ArticlePDF Available

Evaluating the Cancer Therapeutic Potential of Cardiac Glycosides

Wiley
BioMed Research International
Authors:

Abstract and Figures

Cardiac glycosides, also known as cardiotonic steroids, are a group of natural products that share a steroid-like structure with an unsaturated lactone ring and the ability to induce cardiotonic effects mediated by a selective inhibition of the Na(+)/K(+)-ATPase. Cardiac glycosides have been used for many years in the treatment of cardiac congestion and some types of cardiac arrhythmias. Recent data suggest that cardiac glycosides may also be useful in the treatment of cancer. These compounds typically inhibit cancer cell proliferation at nanomolar concentrations, and recent high-throughput screenings of drug libraries have therefore identified cardiac glycosides as potent inhibitors of cancer cell growth. Cardiac glycosides can also block tumor growth in rodent models, which further supports the idea that they have potential for cancer therapy. Evidence also suggests, however, that cardiac glycosides may not inhibit cancer cell proliferation selectively and the potent inhibition of tumor growth induced by cardiac glycosides in mice xenografted with human cancer cells is probably an experimental artifact caused by their ability to selectively kill human cells versus rodent cells. This paper reviews such evidence and discusses experimental approaches that could be used to reveal the cancer therapeutic potential of cardiac glycosides in preclinical studies.
This content is subject to copyright. Terms and conditions apply.
Review Article
Evaluating the Cancer Therapeutic Potential of
Cardiac Glycosides
José Manuel Calderón-Montaño,1Estefanía Burgos-Morón,1Manuel Luis Orta,2
Dolores Maldonado-Navas,1Irene García-Domínguez,1and Miguel López-Lázaro1
1Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain
2DepartmentofCellBiology,FacultyofBiology,UniversityofSeville,Spain
Correspondence should be addressed to Miguel L´
opez-L´
azaro; mlopezlazaro@us.es
Received  February ; Revised  April ; Accepted  April ; Published  May 
Academic Editor: Gautam Sethi
Copyright ©  Jos´
e Manuel Calder´
on-Monta˜
no et al. is is an open access article distributed under the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly cited.
Cardiac glycosides, also known as cardiotonic steroids, are a group of natural products that share a steroid-like structure with an
unsaturated lactone ring and the ability to induce cardiotonic eects mediated by a selective inhibition of the Na+/K+-ATPase.
Cardiac glycosides have been used for many years in the treatment of cardiac congestion and some types of cardiac arrhythmias.
Recent data suggest that cardiac glycosides may also be useful in the treatment of cancer. ese compounds typically inhibit cancer
cell proliferation at nanomolar concentrations, and recent high-throughput screenings of drug libraries have therefore identied
cardiac glycosides as potent inhibitors of cancer cell growth. Cardiac glycosides can also block tumor growth in rodent models,
which further supports the idea that they have potential for cancer therapy. Evidence also suggests, however, that cardiac glycosides
may not inhibit cancer cell proliferation selectively and the potent inhibition of tumor growth induced by cardiac glycosides in
mice xenograed with human cancer cells is probably an experimental artifact caused by their ability to selectively kill human cells
versus rodent cells. is paper reviews such evidence and discusses experimental approaches that could be used to reveal the cancer
therapeutic potential of cardiac glycosides in preclinical studies.
1. Introduction
Cardiac glycosides, also known as cardiotonic steroids, are
natural products with a steroid-like structure and an unsat-
urated lactone ring. ey usually contain sugar moieties
in their structure and have cardiotonic activity. Cardiac
glycosides containing the lactone -furanone are known as
cardenolides and those containing the lactone -pyrone are
known as bufadienolides (Figure ). Most cardiac glycosides
(e.g., digitoxin, digoxin, ouabain, and oleandrin) have been
isolated from plants, including Digitalis purpurea, Digitalis
lanata,Strophanthus gratus,andNerium oleander.Some
cardiac glycosides have also been found in amphibians and
mammals, including digoxin, ouabain, bufalin, marinobufa-
genin, and telecinobufagin. Several cardiac glycosides are
used in cardiology for the treatment of cardiac congestion
andsometypesofcardiacarrhythmias.emechanismby
which these drugs aect cardiac contractility is thought to
be mediated by a highly specic inhibition of the Na+/K+-
ATPase pump [].
Over the years, several reports have suggested that cardiac
glycosides may have an anticancer utilization (reviewed in
[]). In vitro and ex vivo experiments have revealed that
some cardiac glycosides (e.g., digitoxin) induce potent and
selective anticancer eects [,,], which may occur at con-
centrations commonly found in the plasma of patients treated
with these drugs []. Recent high-throughput screenings
of drug libraries have identied several cardiac glycosides
(e.g., digoxin, ouabain, and bufalin) as potent inhibitors of
cancer cell growth []. ese cardiac glycosides were also
able to block tumor growth in mice xenotransplanted with
human cancer cells, further supporting the idea that these
compounds should be evaluated in cancer patients [].
e cardiac drugs digitoxin and digoxin, the semisynthetic
cardiac glycoside UNBS, and two extracts from the plant
Nerium oleander have entered clinical trials for the treatment
Hindawi Publishing Corporation
BioMed Research International
Volume 2014, Article ID 794930, 9 pages
http://dx.doi.org/10.1155/2014/794930
BioMed Research International
RO
OH
Cardenolide Bufadienolide
Digitoxin: R = digitoxose-digitoxose-digitoxose Bufalin: R = H
RO
OH
OO
O
O
F : Chemical structure of cardiac glycosides. e basic skeletons of cardenolides and bufadienolides and the structures of the
cardenolide digitoxin and the bufadienolide bufalin are shown.
of cancer (see http://clinicaltrials.gov/ and ref. [,,,,
]).
Research results also suggest, however, that cardiac gly-
cosides may not inhibit cancer cell proliferation selectively
in particular types of cancer []andthepotentinhi-
bition of tumor growth induced by cardiac glycosides in
mice xenograed with human cancer cells is probably an
experimental artifact caused by their ability to selectively
kill human cells versus rodent cells rather than by their
ability to selectively kill human cancer cells versus human
normal cells []. Aer reviewing such evidence, this
paper discusses experimental approaches that can be used to
reveal the cancer therapeutic potential of cardiac glycosides
in preclinical studies.
2. Possible Misinterpretation of
Data from Preclinical Studies
Inhibition of cancer cell proliferation at low concentrations
and inhibition of tumor growth in animal models are the
most common parameters used by researchers to assess the
therapeutic potential of drug candidates in preclinical studies.
Based on this approach, researchers have proposed cardiac
glycosides as candidates for evaluation in clinical trials. is
section of the paper reviews evidence indicating that this
approach may be inadequate to reveal the cancer therapeutic
potential of cardiac glycosides.
2.1. Inhibition of Cancer Cell Proliferation at Low Concen-
trations Does Not Reliably Predict erapeutic Potential. e
key feature of an ecient anticancer drug candidate is its
ability to kill (or to inhibit the proliferation of) human cancer
cells at concentrations that do not signicantly aect human
nonmalignant cells. If the anticancer drug candidate does not
have this feature, it does not really matter whether or not it
can kill cancer cells at low concentrations. e reason is that
the drug concentrations required to kill the tumor cells of
cancer patients would also cause the death of their normal
cells and, therefore, would be lethal to these patients. It is
important to note that the therapeutic potential of a drug able
to kill cancer cells at a concentration of  millimolar without
signicantly aecting nonmalignant cells at a concentration
of  millimolar is probably higher than that of a drug that
kills both cancer and nonmalignant cells at a concentration
of  nanomolar.
Cancer researchers do not commonly use human non-
malignant cells to assess the therapeutic potential drug
candidates. Possible reasons are that they may consider
that the inhibition of human cancer cell proliferation at
low concentrations is an adequate parameter to predict
therapeutic potential or they prefer using animal models
instead. Researchers typically use mice xenotransplanted
with human cancer cells to reveal whether their drug can-
didates inhibit cancer cell growth selectively. If their drugs
inhibit tumor growth in these models without killing or
signicantly aecting the animals, they assume that their
drugs also inhibit the proliferation of human cancer cells
without signicantly aecting that of human nonmalignant
cells. Following this approach, researchers have proposed
several cardiac glycosides as candidates for clinical testing in
cancer patients [,,].
Several research groups have evaluated the cancer thera-
peutic potential of cardiac glycosides by using human cancer
cells and human nonmalignant cells. For instance, we recently
observed that the cytotoxicity of digitoxin, digoxin, and
ouabain in breast cancer cells (MCF-) and melanoma cells
(UACC-) was similar than that in nonmalignant breast cells
(MCF-) and nonmalignantskin cells (VH-) []. Cliord
and Kaplan [] have recently reported that human breast
cancer cells were even more resistant to ouabain, digitoxin,
and bufalin toxicity than human nonmalignant breast cells.
Evidence has also shown, however, that digitoxin, digoxin,
and ouabain were approximately  times more cytotoxic
against human A lung cancer cells than against human
MRC- nonmalignant lung cells (– nM versus – nM)
[]. Ex vivo experiments, using cells from adult patients
with B-precursor or T-acute lymphoblastic leukemia (ALL),
acute myeloid leukemia (AML), and chronic lymphocytic
leukemia (CLL), as well as peripheral blood mononuclear
cells from healthy donors, have also shown that digitoxin
(but not ouabain) induced selective cytotoxicity (approxi-
mately-fold)incellsfrompatientswithT-andB-precursor
ALL []. In brief, although cardiac glycosides can inhibit
BioMed Research International
the proliferation of cancer cells at very low concentrations
(nM), they usually inhibit the proliferation of human nonma-
lignant cells at similar concentrations; this strongly suggests
that their potential for cancer therapy is low. In contrast,
specic cardiac glycosides (e.g., digitoxin) can inhibit the
proliferation of particular types of cancer cells (e.g., lung can-
cer and acute lymphoblastic leukemia) at concentrations that
do not signicantly aect human nonmalignant cells; these
cardiac glycosides may have cancer therapeutic potential.
2.2. e Anticancer Activity of Cardiac Glycosides in Mice
Xenograed with Human Cancer Cells Is Probably an Exper-
imental Artifact. Several cardiac glycosides that are equally
toxic to human cancer cells and human nonmalignant cells
haveshownpotentanticancereectsinanimalmodels.
For instance, Cliord and Kaplan observed that human
nonmalignant breast cells were more sensitive than human
breast cancer cells (e.g., MDA-MB-) to the cytotoxic
eects of bufalin [], and it has recently been reported
that bufalin reduces tumor growth in mice xenotransplanted
with human MDA-MB- breast cancer cells []. ese
apparent controversies can be explained by the ability of
cardiac glycosides to kill human cells at concentrations much
lower (approximately – fold) than those required to
kill rodent cells [,].
Gupta and colleagues [] evaluated some time ago the
cytotoxicity of numerous cardiac glycosides (i.e., ouabain,
digitoxin, digoxin, convallatoxin, SC, bufalin, gitaloxin,
digoxigenin, actodigin, oleandrin, digitoxigenin, gitoxin,
strophanthidin, gitoxigenin, lanatosides A, B, and C, alpha-
and beta-acetyl digoxin, and alpha- and beta-methyl digoxin)
against a number of independent cell lines established
from human, monkey, mouse, Syrian hamster, and Chinese
hamster. e authors observed that all cardiac glycosides
exhibited greater than -fold higher toxicity towards the
human and monkey cells in comparison to the rodent cells
(mouse, Syrian hamster, and Chinese hamster). ey also
provided strong evidence that the species-related dierences
in sensitivity to cardiac glycosides were mediated by the
Na+/K+-ATPase enzyme. ey obser ve d that th e Na+/K+-
ATPase enzyme of rodent cells was inhibited at much higher
concentrations of cardiac glycosides than the Na+/K+-ATPase
of human cells. ey also observed a good correlation
between these concentrations and those reported for inhibi-
tion of the Na+/K+-ATPasefromisolatedheartmusclesof
thesamespecies[]. More recent evidence suggests that
the expression and cellular location of Na+/K+-ATPase alpha
subunits in dierent types of cells may explain why they are
more or less susceptible to the cytotoxic activity of cardiac
glycosides [].
Several years ago, a PNAS paper reported that digoxin
blocked tumor growth in mice xenotransplanted with several
types of human cancer cells []. e authors observed
that digoxin prolonged tumor latency and inhibited tumor
xenogra growth in mice when treatment was initiated
before the implantation of P-Myc, P-Myc-Luc, PC,
and HepB cells. Digoxin also arrested tumor growth when
treatment was initiated aer the establishment of PC and
P-Myc tumor xenogras []. Based on the observations
of Gupta and colleagues []andontheplasmalevels
of digoxin in cardiac patients, we discussed the fact that
the potent anticancer eects induced by digoxin in mice
harboring human cancer cells []werenotrelevanttothe
treatment of human cancer and these anticancer eects were
probably due to interspecies dierences in sensitivity []. In
other words, the marked reduction in tumor growth induced
by digoxin in mice xenograed with human cancer cells
was probably caused by the ability of cardiac glycosides to
selectively kill human cells versus rodent cells rather than
by their ability to selectively kill cancer cells versus normal
cells. Perne et al. [] later reported experimental data that
further supported this idea. Despite these and other reports
[,], numerous publications containing this probable
experimental artifact continue to appear in the scientic
literature.
is section of the paper now reviews reports that have
used mice xenotransplanted with human cancer cells to
evaluate anticancer eects of cardiac glycosides (Tab le ).
e results of the following reports should probably be
reinterpreted.
Digoxin. Svensson et al. [] carried out in vitro and in vivo
studies to evaluate the anticancer activity of the cardenolide
digoxin. ey studied the eect of digoxin on the growth of
tumor cell lines and primary endothelial cells from dierent
species. e most sensitive cell lines in vitro were the human
SH-SYY and SK-N-AS neuroblastoma cell lines; the IC50
values were  and  ng/mL, respectively. ey also reported
that digoxin signicantly reduced the growth of human SH-
SYY neuroblastoma cells xenotransplanted in immunode-
cient mice. e authors concluded that digoxin might be
a specic neuroblastoma growth inhibitor. e authors also
reported that the in vitro and in vivo anticancer eects of
digoxin were dramatically reduced when the murine Neuro-
a neuroblastoma cell line was used instead of the human
neuroblastoma cell lines []. Zavareh et al. []reported
data suggesting that cardiac glycosides were inhibitors of
N-glycan biosynthesis. Since aberrant N-linked glycans are
known to contribute to cancer progression and metastasis,
the authors studied whether digoxin could inhibit cellular
migration and invasion. ey used two mouse models of
metastatic cancer in which human PPC- prostate cancer
cells were injected into immunodecient mice. ey found
that digoxin reduced distant tumor formation in both models
and concluded that this cardiac glycoside could be a lead
for the development of antimetastasis therapies. As discussed
before,Zhangetal.[] found that digoxin inhibited hypoxia-
inducible factor  (HIF-), a transcription factor highly
involved in cancer development, and suggested that this
eect might be observed in patients taking this drug. ey
also reported that digoxin blocked tumor growth in mice
xenotransplanted with several types of human cancer cells.
ese data suggested that digoxin had anticancer potential
[]. Wong et al. [] reported data suggesting that digoxin
was a potential antimetastasis compound. ey investigated
whether digoxin could reduce metastases in human MDA-
MB- tumor-bearing mice. Digoxin blocked metastatic
BioMed Research International
T : e antitumor activity of cardiac glycosides in mice xenograed with human cancer cells is probably caused by their ability to
selectively kill human cells versus rodent cells rather than by their ability to selectively kill human cancer cells versus human nonmalignant
cells.
Cardiac
glycoside
Antitumor activity in mice
xenograed with human cancer
cells
Selective cytotoxicity
against human cells
versus rodent cells
Selective cytotoxicity against human cancer cells versus
human nonmalignant cells
Arenobufagin Liver HepG/ADM [] N.D. N.D.
Bufalin
Breast MDA-MB- [],
osteosarcoma UOS/MTX
[], and pancreatic Mia Paca-
[]
>-fold []
NO: breast cancer versus breast nonmalignant [];
-fold: ovarian cancer versus endometrial
nonmalignant []
Bufotalin Liver R-HepG []>-fold []N.D.
Digitoxin N.D.
>-fold [];
>-fold []
-fold: lung cancer versus lung nonmalignant [];
-fold: lung cancer versus lung nonmalignant [];
-fold: ALL versus PBMCs nonmalignant []; -fold:
AML versus PBMCs nonmalignant []; -fold: CLL
versus PBMCs nonmalignant []; -fold: breast cancer
versus breast nonmalignant []; NO: breast cancer
versus breast nonmalignant []; NO: skin cancer
versus skin nonmalignant []
Digoxin
Brain SH-SYY [], brain
SK-N-AS [], breast
MDA-MB- [,], breast
MDA-MB- [], liver HepB
[], prostate PC [], prostate
PPC- [], and transformed
human B-lymphocytes
P-Myc []
>-fold [,]
NO: breast cancer versus breast nonmalignant []; -
fold: lung cancer versus lung nonmalignant []; NO:
skin cancer versus skin nonmalignant []; -fold: brain
cancer versus umbilical vein endothelial nonmalignant
[]; NO: breast cancer versus umbilical vein
endothelial nonmalignant []; NO: colorectal cancer
versus umbilical vein endothelial nonmalignant []
Lanatoside C Brain U []>-fold []N.D.
Ouabain
Brain SH-SYY [], ocular
YLUC [], pancreatic BON
[], promyelocytic leukemia
HL- [], and prostate PPC-
[]
>-fold [,,]
NO: breast cancer versus breast nonmalignant [,];
- fold: lung cancer versus lung nonmalignant [];
NO: skin cancer versus skin nonmalignant []; -fold:
ALL versus PBMCs nonmalignant []; -fold: AML
versus PBMCs nonmalignant []; NO: CLL versus
PBMCs nonmalignant []
Periplocin Liver Huh- []andlungA
[]
>-fold [];NO:
[]
>-fold: liver cancer versus PBMCs nonmalignant
[]
UNBS
Brain U-MG [], lung A
[,], lung NCI-H [,],
prostate PC- [], and skin
VM- []
>-fold []
-fold: brain cancer versus lung and skin
nonmalignant []; -fold: prostate cancer versus
lung and skin nonmalignant []
N.D.: not determined; NO: no selective cytotoxicity; ALL: acute lymphoblastic leukemia; AML: acute myeloid leukemia; CLL: chronic lymphocytic leukemia;
PBMCs: peripheral blood mononuclear cells; not specied if the PBMCs were human cells or rodent cells (we contacted the authors without success).
niche formation and breast cancer metastasis in the lungs,
andtheauthorsdiscussedthefactthatthiseectwasprobably
due to inhibition of HIF-. e most relevant conclusion of
this work was that digoxin might be useful to treat patients
with HIF--overexpressing breast cancers []. Zhang et al.
[] observed that digoxin reduced tumor growth and inhib-
ited the metastasis of human MDA-MB- breast cancer
cellstothelungsinmicexenograedwiththesecells,without
causing any sign of toxicity in the animals. ey concluded
that clinical trials were warranted to investigate whether the
concentrations of digoxin achievable in patients are sucient
to inhibit tumor growth and metastases []. Schito et al. []
reported that HIF- promoted lymphatic metastases of breast
cancer and the use of the HIF- inhibitor digoxin strongly
decreased tumor growth and blocked lymphangiogenesis and
lymphaticmetastasisinmicebearinghumanbreastcancer
cells. e authors suggested that digoxin might be useful to
treat patients with high risk of lymphatic metastases [].
Gayed et al. [] observed that specic concentrations of
digoxin inhibited blood vessel formation but not tumor
growth in mice injected with the human C- prostate cancer
cell line.
Ouabain. Several cardiac glycosides were identied by
Antczak et al. [] as potent antiretinoblastoma agents in
vitro. One of them, the cardenolide ouabain, induced a drastic
tumor regression in immunodecient mice injected with
human YLUC retinoblastoma cells, without inducing any
BioMed Research International
signicant toxicity on the host. In light of the results of their
study, the authors proposed that digoxin, which is widely
used in patients with cardiac disease, could be repositioned
for the treatment of retinoblastoma []. Simpson et al.
[] identied the cardiac glycosides ouabain, peruvoside,
digoxin, digitoxin, and strophanthidin as anoikis sensitizers.
Because resistance to anoikis permits cancer cells to survive
in the circulation and improves their metastatic potential,
the authors evaluated in mouse models of metastasis whether
ouabain could block distant tumor formation. ey observed
that ouabain reduced the number of tumors in human PPC-
prostate cancer cells bearing mice. ey also reported that
systemic administration of ouabain decreased the survival
and growth of human PPC- prostate cancer cells and human
BON pancreatic cancer cells xenograed into nude mice
[]. Hiyoshi et al. [] reported that ouabain induced quies-
cence in neuroblastoma cells in vitro and a marked reduction
in tumor growth when human neuroblastoma cells were
xenograed into immune-decient mice. Based on these
ndings, the authors concluded that ouabain could be used in
chemotherapies to suppress tumor growth and/or arrest cells
to increase the therapeutic index in combination therapies.
Tail ler e t al. [ ] identied the cardiac glycoside ouabain
as a potential antileukemic compound. ey observed that
ouabain was highly ecient in inhibiting the growth of
human acute myeloid leukemia cells xenotransplanted in
immunodecient mice, without exerting signicant toxic-
ity on the host. e authors concluded that ouabain was
a promising antileukemic agent whose activity should be
evaluated in prospective clinical studies [].
UNBS1450. Mijatovic et al. []investigatedthein vitro and
in vivo anticancer activity of UNBS, a semisynthetic
derivative of the natural cardenolide UNBS (isolated
from the African plant Calotropis procera). ey observed
that UNBS was able to inhibit cell growth of four
dierentnon-smallcelllungcancercells(A,NCI-H,
A, and CAL-T) at nanomolar concentrations. is
cardenolide also signicantly decreased tumor growth in
nude mice xenograed with human NCI-H cancer cells
andincreasedthesurvivalratesinmicexenograedwith
human A cancer cells. e authors observed in another
study [] that the cytotoxic potency of UNBS in A
lungcancercellswassimilarthanthatoftheanticancerdrugs
paclitaxel and SN (the active metabolite of irinotecan)
and much higher than that of cisplatin, carboplatin, and
oxaliplatin. UNBS also decreased tumor growth in mice
xenotransplanted with A lung cancer cells and human
NCI-H lung cancer cells []. Another study revealed
that UNBS inhibited the proliferation of human prostate
cancer cells (LNCaP, PC-, and DU) and increased the
survival of mice transplanted with human PC- prostate can-
cer cells []. Lefranc et al. [] reported that UNBS was
more cytotoxic on human glioblastoma cells (U-MG and
TG)thanonhumannormalbroblasts(WI-andWSI)
at nanomolar concentrations. is compound also inhibited
the proliferation of rat C glioblastoma cells at micromolar
concentrations. UNBS increased the survival of mice
graed with human U-MG glioblastomas cells, without
observabletoxiceectsontheanimals.Mathieuetal.[]
reported that UNBS blocked cell proliferation in several
human melanoma cell lines in vitro (IC50 values between 
and  nM) and improved the survival of immunodecient
mice graed with human VM- melanoma brain metastasis
cells.
Periplocin. Lu et al. [] reported that the natural cardenolide
periplocin induced similar cytotoxicity against a panel of
human lung cancer cell lines than against a rodent lung cancer
cell line (LL/). ey also observed antitumor activity in mice
transplanted with both the human A lung cancer cell
line and the murine LL/ Lewis lung cancer cell line. Cheng
et al. [] have recently reported that periplocin displayed a
potent cancer cell growth inhibitory activity in vitro and in
vivo. Periplocin inhibited cell growth of human HAT/VGH
hepatocellular carcinoma with an IC50 of  nM and was less
toxic to normal peripheral blood mononucleated cells. e
authors also observed that periplocin showed an inhibition
of tumor growth when human Huh- hepatoma cells were
injected into immunodecient mice, without observing clear
side eects on the host.
Lanatoside C. Badr et al. [] identied the cardenolide
lanatoside C as a sensitizer of glioblastoma cells to tumor
necrosis factor-related apoptosis-inducing ligand (TRAIL)-
induced cell death. ey observed that lanatoside C, alone or
in combination with TRAIL, reduced tumor growth in nude
mice harboring human U glioblastoma cells.
Bufalin. Chen et al. [] identied the bufadienolide bufalin
as a potential agent for the treatment of pancreatic cancer in
combination with the standard anticancer drug gemcitabine.
ey found that bufalin inhibited the growth on three pancre-
atic cancer cell lines (Bxpc-, Mia PaCa-, and Panc-) and
it synergistically increased gemcitabine-induced cancer cell
growth inhibition and apoptosis. e combination of bufalin
with gemcitabine was also found to signicantly reduce
tumor growth in mice bearing human Mia Paca- pancreatic
cancer cells. Xie et al. []investigatedthein vitro and in
vivo antiosteosarcoma activity of bufalin. ey observed that
bufalin strongly inhibited the cell growth of dierent human
osteosarcoma cell lines, including the methotrexate-resistant
UOS/MTX cell line. ey also found that the treatment
with bufalin induced signicant tumor growth inhibition
in mice xenotransplanted with the human UOS/MTX
osteosarcoma cell line, without decreasing the body weight
oftheanimals.eauthorsconcludedthatbufalinmightbe
an alternative chemotherapeutic agent to treat osteosarcoma,
particularly in methotrexate-resistant cancers []. Wang
et al. [] have recently reported that bufalin was a potent
inhibitor of the steroid receptor coactivators SRC- and SRC-
. Because these coactivators have been implicated in cancer
progression, the authors investigated whether bufalin could
also block cancer cell growth in cell culture and animal
models. ey observed that bufalin inhibited the growth of
human MCF- breast cancer cells and human A lung
cancer cells at nanomolar concentrations (–nM); these
concentrations also resulted in inhibition of the steroid
BioMed Research International
receptor coactivator SRC- and were below those reported
to be tolerated by humans (. nM). ey also found that
bufalin inhibited tumor growth in mice xenotransplanted
with human MDA-MB- breast cancer cells.
Arenobufagin. Zhang et al. [] recently observed that the
bufadienolide arenobufagin induced a potent cell growth
inhibitory activity on cancer cells both in vitro and in
vivo. ey tested its anticancer activity on several human
cancer cell lines (hepatoma, breast adenocarcinoma, cervix
adenocarcinoma, lung cancer, colon cancer, leukemia, and
gastric adenocarcinoma). Arenobufagin inhibited the growth
of all cancer cell lines at nanomolar concentrations, includ-
ing multidrug-resistant cancer cell lines. Arenobufagin also
inhibited the growth of human HepG/ADM hepatocellular
carcinoma cells xenograed into immunodecient mice,
without causing side eects on the hosts. e authors con-
cluded that their results may provide a rationale for future
clinical application using arenobufagin as a chemotherapeu-
tic agent for the treatment of patients with hepatocarcinoma
[].
Bufotalin. Zhang et al. [] observed that four bufadienolides
from Venenum Bufonis, a traditional Chinese medicine,
displayed inhibitory eects on the growth of human HepG
hepatocarcinoma cells and human R-HepG multidrug hep-
atocarcinoma cells. One of them, bufotalin, was also able
to signicantly inhibit the growth of human R-HepG cells
xenograed into immunodecient mice, without observing
any life-threatening toxicity in the animals. e authors
discussed the fact that their study supports the possible
development of bufotalin as a potential agent in the treatment
of multidrug resistant hepatocellular carcinoma [].
Data from preclinical studies reporting antitumor eects
in rodent xenogras of plant extracts containing cardiac
glycosides may also need reinterpretation. For instance,
Han et al. [] reported that an extract from the plant
Streptocaulon juventas induced a strong inhibitory eect
on the proliferation of human lung A adenocarcinoma
cells. A bioassay-guided fractionation revealed that the most
cytotoxic fraction in vitro also induced antitumor eects in
athymicnudemicetransplantedwithhumanAcancer
cells without exerting side eects on the mice. Following
HPLC and NMR spectrometry, the main components of
this active fraction were identied as the cardiac glycosides
digitoxigenin, periplogenin, and periplogenin glucoside [].
3. Possible Approaches to Reveal the Cancer
Therapeutic Potential of Cardiac Glycosides
in Preclinical Studies
As discussed before, the key feature of an ecient anticancer
drug candidate is its ability to kill (or to inhibit the pro-
liferation of ) human cancer cells at concentrations that do
not signicantly aect human nonmalignant cells. Ideally, the
drugcandidateshouldkillallthecancercellsofthepatients
without signicantly aecting their normal cells. Because this
is dicult to achieve, one can settle for less. A drug that
improves the ability of our current anticancer drugs to kill
cancer cells at concentrations that do not signicantly aect
nonmalignant cells could be therapeutically useful.
In vitro, one can evaluate whether the drug candidate
improves the selective cytotoxicity of the standard anticancer
drugs towards cancer cells by using the following approach.
e rst step in this approach is the selection of a panel of
human cancer cell lines and human nonmalignant cell lines
(or primary cells). Because the cytotoxicity of some drugs
dependsonthenatureofthetissuefromwhichtheyoriginate,
one should select nonmalignant cell lines of the same tissue
origin than that of the selected cancer cell lines. A small
number of cancer cell lines may be sucient to reveal the
therapeutic potential of a drug for a particular type of cancer.
However, the selection of a low number of nonmalignant cell
lines reduces the chances of nding toxicity on a specic
tissue that would limit the possible therapeutic use of the
drug. e next step is to treat the selected cell lines with
several concentrations of the drug candidate and of the
anticancer drugs most commonly used in the treatment of the
selected cancers. en, cell viability or cell death is estimated
with a cytotoxicity test (e.g., SRB assay and MTT assay), and
cytotoxic parameters (e.g., IC50 values) are calculated. e
following step is to calculate one or several selectivity indexes
for the drug candidate and for the anticancer drug. ese
selectivity indexes can be calculated by dividing the IC50
values in the nonmalignant cell lines by the IC50values in the
cancer cell lines. For instance, if the mean IC50 value of a drug
in a variety of nonmalignant cells originated from several
tissues is  𝜇MandthemeanIC
50 value of the drug in
several cell lines derived from a specic cancer is  𝜇M, the
selectivity index for this particular cancer would be . Finally,
the following question must be answered: is the selectivity
index of the drug candidate higher (or at least similar) than
that of the standard anticancer drug? If the answer is no,
the drug candidate does not have therapeutic potential and
shouldnotbetestedinanimalmodels.Iftheanswerisyes,
the drug candidate has chemotherapeutic potential, which
should be conrmed by using in vivo experiments.
Rodent xenogra models are the most common animal
models used by researchers to evaluate the therapeutic
potential of anticancer drug candidates in vivo.However,as
discussed before, these models may be inadequate to evaluate
the therapeutic potential of cardiac glycosides. To the authors’
knowledge, all cardiac glycosides tested in human cells and
rodent nonmalignant cells have shown greater than -
fold higher toxicity towards the human cells in comparison
to the rodent cells. is does not mean, however, that all
compounds having the basic chemical structure of cardiac
glycosides (a steroid skeleton with an unsaturated lactone
ring)willbemoretoxicagainsthumancellsthanagainst
rodent cells. One can test the suitability of using tumor
xenogras to evaluate the in vivo therapeutic potential of a
particular cardiac glycoside by testing if the cytotoxicity of
the cardiac glycoside against a panel of human nonmalignant
cells is similar than that against a panel of rodent nonmalig-
nant cells. If the compound behaves similarly in both types
of cell lines, its in vivo anticancer activity can be evaluated
in mice xenograed with human cancer cells. If the rodent
BioMed Research International
cell lines are more resistant than the human cell lines to
the cytotoxicity of the cardiac glycoside, these models are
probably inadequate to evaluate its anticancer eects in vivo.
Animal models using mice transplanted with mouse cancer
cells may also be inadequate when human cells are more
sensitive than rodent cells to the cytotoxicity of the cardiac
glycoside. e reason is that the therapeutic target responsible
for the death of the human cells may be dierent than that
responsible for the death of the rodent cells and, therefore,
resultsobtainedinmicemaynotbeextrapolatedtohumans.
e anticancer activity of cardiac glycosides displaying
a similar cytotoxic prole in nonmalignant cells originated
from both human and mouse tissues can be assessed by using
tumor xenogras or other rodent models. It is important
to remember that most cancer patients requiring therapy
with anticancer drugs have metastatic disease and patient
survival is the parameter used by oncologists as an endpoint
of clinical interventions designed to assess drug ecacy
in patients with cancer (other parameters used by many
preclinical researchers as an endpoint for their experiments,
such as measurements of tumor volumes, do not necessarily
predictsurvival).Itisessential,therefore,toselectanimal
models of metastasis and to assess animal survival as an
endpoint for the experiments. In our opinion, animals with
metastasis should be treated with equitoxic concentrations
of the cardiac glycoside and of the standard anticancer drug
usedinthetypeofcancerunderstudy.en,oneshould
evaluate whether the cardiac glycoside improves the survival
rates induced by the standard anticancer drug. If the cardiac
glycoside improves (or at least matches) the selectivity index
(in vitro)andthesurvivalrates(in vivo)ofthestandard
anticancer drugs, it should be considered for clinical trials
testing.
Rodent models are inappropriate for testing the anti-
cancer activity of cardiac glycosides that kill human non-
malignant cells at lower concentrations than those required
to kill rodent nonmalignant cells. ese models, however,
could provide information on the pharmacokinetics of the
cardiac glycoside. ese models may also help detect possible
toxicity not detected by using a panel of human nonmalignant
cell lines; they could help detect toxicity not mediated by
inhibition of the Na+/K+-ATPase (which seems to be the
main determinant for the species dierences in sensitivity
to cardiac glycosides). In our opinion, a cardiac glycoside
that kills human nonmalignant cells at lower concentrations
than rodent nonmalignant cells should pass the following
tests before being considered for evaluation in clinical trials.
First, it should match or improve the selectivity indexes of the
standard anticancer drugs when they are evaluated in a panel
of human cancer cell lines derived from a particular type
of cancer versus a variety of human nonmalignant cell lines
and primary cells derived from a variety of human tissues.
Second, in vivo experiments (e.g., rodent models) should
exclude pharmacokinetic and toxicological limitations that
may compromise the in vivo anticancer activity of the cardiac
glycoside. Finally, if the cardiac glycoside is in clinical use
for the management of other diseases or if clinical data
already exist on its plasma and tissue concentrations, one
should also consider whether the anticancer eects observed
in preclinical studies may occur at concentrations within or
below the concentration range tolerated by humans.
4. Conclusion
Preclinical research has shown that cardiac glycosides can
both inhibit cancer cell proliferation at very low concen-
trations and induce potent anticancer eects in mice trans-
planted with human cancer cells. Based on these observa-
tions, cardiac glycosides have been considered as potential
anticancer drug candidates that should be evaluated in
clinical studies. is paper has reviewed evidence indicating
that cardiac glycosides may not selectively inhibit the prolif-
eration of human cancer cells and these compounds have the
ability of killing human cells at concentrations much lower
than those required to kill rodent cells (approximately –
 fold). is strongly suggests that the potent anticancer
eects induced by cardiac glycosides in mice transplanted
with human cancer cells may be an experimental artifact
caused by their ability to selectively kill human cells versus
rodent cells rather than by their ability to kill human cancer
cells versus human nonmalignant cells. It has also been
discussed that inhibition of cancer cell proliferation at low
concentrations is not an adequate parameter to predict the
therapeutic potential of a drug candidate. e key feature of
an ecient anticancer drug is its ability to kill (or inhibit the
proliferation of ) human cancer cells at concentrations that
do not signicantly aect human nonmalignant cells. Based
on this principle, an approach to evaluate the therapeutic
potential of cardiac glycosides in preclinical in vitro studies
has been proposed. is approach is also based on the
idea that only drug candidates that match or improve the
ability of the approved anticancer drugs to kill human cancer
cells at concentrations that do not signicantly aect human
nonmalignant cells have a chance to be ultimately used in
cancer therapy. A test for revealing the suitability of using
rodent models for the evaluation of the anticancer activities
of cardiac glycosides in vivo has also been proposed. If the
cardiac glycoside passes this test, several recommendations
have been made for the evaluation of its cancer therapeutic
potential in these models. If the cardiac glycoside fails to pass
this test, an alternative approach for revealing its possible
therapeutic potential has been discussed. It is the hope of
the authors that this paper may help researchers evaluate
the therapeutic potential of cardiac glycosides in preclinical
studies.
Conflict of Interests
e authors declare that there is no conict of interests
regarding the publication of this paper.
References
[] S. H. Rahimtoola and T. Tak, “e use of digitalis in heart
failure,Current Problems in Cardiology,vol.,no.,pp.
, .
BioMed Research International
[] Z. Xie and A. Askari, “Na+/K+-ATPase as a signal transducer,
European Journal of Biochemistry,vol.,no.,pp.
, .
[] W. Schoner and G. Scheiner-Bobis, “Endogenous and exoge-
nous cardiac glycosides: their roles in hypertension, salt
metabolism, and cell growth,AmericanJournalofPhysiology-
Cell Physiology,vol.,no.,pp.CC,.
[] J. Haux, “Digitoxin is a potential anticancer agent for several
types of cancer,Medical Hypotheses,vol.,no.,pp.,
.
[] M. L ´
opez-L´
azaro, “Digitoxin as an anticancer agent with selec-
tivity for cancer cells: possible mechanisms involved,Expert
OpiniononerapeuticTargets,vol.,no.,pp.,
.
[] R.A.Newman,P.Yang,A.D.Pawlus,andK.I.Block,“Cardiac
glycosides as novel cancer therapeutic agents,Molecular Inter-
ventions,vol.,no.,pp.,.
[] I.PrassasandE.P.Diamandis,“Noveltherapeuticapplications
of cardiac glycosides,Nature Reviews Drug Discovery,vol.,no.
, pp. –, .
[] T.Mijatovic,E.VanQuaquebeke,B.Delest,O.Debeir,F.Darro,
and R. Kiss, “Cardiotonic steroids on the road to anti-cancer
therapy,” Biochimica et Biophysica Acta-Reviews on Cancer,vol.
,no.,pp.,.
[] T. Mijatovic, F. Dufrasne, and R. Kiss, “Cardiotonic steroids-
mediated targeting of the Na+/K+-ATPase to combat chemore-
sistant cancers,” Current Medicinal Chemistry,vol.,no.,pp.
–, .
[] T. Mijatovic and R. Kiss, “Cardiotonic steroids-mediated
Na+/K+-ATPase targeting could circumvent various chemore-
sistance pathways,Planta Medica, vol. , no. -, pp. –,
.
[] M. Slingerland, C. Cerella, H. J. Guchelaar, M. Diederich, and
H. Gelderblom, “Cardiac glycosides in cancer therapy: from
preclinical investigations towards clinical trials,Invest New
Drugs,vol.,no.,pp.,.
[] P.Babula,M.Masarik,V.Adam,I.Provaznik,andR.Kizek,
“From Na+/K+-ATPase and cardiac glycosides to cytotoxicity
and cancer treatment,Anti-Cancer Agents in Medicinal Chem-
istry,vol.,no.,pp.,.
[] C.Cerella,M.Dicato,andM.Diederich,“Assemblingthepuzzle
of anti-cancer mechanisms triggered by cardiac glycosides,
Mitochondrion,vol.,no.,pp.,.
[] Y. Sreenivasan, P. B. Raghavendra, and S. K. Manna,
“Oleandrin-mediated expression of Fas potentiates apoptosis
in tumor cells,Journal of Clinical Immunology,vol.,no.,
pp. –, .
[] H. Hallb¨
o¨
ok,J.Felth,A.Erikssonetal.,“Exvivoactivityof
cardiac glycosides in acute leukaemia,PLoS ONE,vol.,no.
, Article ID e, .
[] M. L ´
opez-L´
azaro, N. Pastor, S. S. Azrak, M. J. Ayuso, C.
A. Austin, and F. Cort´
es, “Digitoxin inhibits the growth of
cancer cell lines at concentrations commonly found in cardiac
patients,Journal of Natural Products,vol.,no.,pp.
, .
[] H. Zhang, D. Z. Qian, Y. S. Tan et al., “Digoxin and other cardiac
glycosides inhibit HIF-𝛼synthesis and block tumor growth,
Proceedings of the National Academy of Sciences of the United
States of America,vol.,no.,pp.,.
[] M. Tailler, L. Senovilla, E. Lainey et al., “Antineoplastic activity
of ouabain and pyrithione zinc in acute myeloid leukemia,
Oncogene,vol.,no.,pp.,.
[] Y.Wang,D.M.Lonard,Y.Yuetal.,“Bufalinisapotentsmall
molecule inhibitor of the steroid receptor coactivators SRC-
and SRC-,Cancer Research,vol.,no.,pp.,.
[] L. J. Rashan, K. Franke, M. M. Khine et al., “Characterization of
the anticancer properties of monoglycosidic cardenolides iso-
lated from Nerium oleander and Streptocaulon tomentosum,
Journal of Ethnopharmacology,vol.,no.,pp.,.
[] J. M. Calderon-Montano, E. Burgos-Moron, M. L. Orta, S.
Mateos, and M. Lopez-Lazaro, “A hydroalcoholic extract from
theleavesofNeriumoleanderinhibitsglycolysisandinduces
selective killing of lung cancer cells,Planta Medica,vol.,no.
, pp. –, .
[] A. Perne, M. K. Muellner, M. Steinrueck et al., “Cardiac
glycosides induce cell death in human cells by inhibitinggeneral
protein synthesis,PloS ONE,vol.,no.,ArticleIDe,
.
[] R. J. Cliord and J. H. Kaplan, “Human breast tumor cells
are more resistant to cardiac glycoside toxicity than non-
tumorigenic breast cells,PLoS ONE,vol.,no.,ArticleID
e, .
[] J. M. Calderon-Montano, E. Burgos-Moron, and M. Lopez-
Lazaro, “e in vivo antitumor activity of cardiac glycosides
in mice xenograed with human cancer cells is probably an
experimental artifact,Oncogene,.
[] M. Lopez-Lazaro, “Digoxin, HIF-, and cancer,” Proceedings of
the National Academy of Sciences of the United States of America,
vol.,no.,p.E,.
[] J. M. Calderon-Montano, E. Burgos-Moron, and M. Lopez-
Lazaro, “Comment on “Quiescence and gammaHAX in neu-
roblastoma are regulated by Ouabain/Na, K-ATPase”: ouabain
and cancer,British Journal of Cancer,vol.,no.,pp.
, .
[] H. Hiyoshi, S. Abdelhady, L. Segerstrom et al., “Quiescence and
gammaHAX in neuroblastoma are regulated by ouabain/Na,
K-ATPase,British Journal of Cancer,vol.,no.,pp.
, .
[] D. M. Zhang, J. S. Liu, L. J. Deng et al., “Arenobufagin, a
natural bufadienolide from toad venom, induces apoptosis and
autophagy in human hepatocellular carcinoma cells through
inhibition of PIK/Akt/mTOR pathway,Carcinogenesis,vol.,
no. , pp. –, .
[] R. S. Gupta, A. Chopra, and D. K. Stetsko, “Cellular basis
for the species dierences in sensitivity to cardiac glycosides
(digitalis),Journal of Cellular Physiology,vol.,no.,pp.
, .
[] T. Mijatovic, I. Roland, E. Van Quaquebeke et al., “e 𝛼
subunit of the sodium pump could represent a novel target to
combat non-small cell lung cancers,Journal of Pathology,vol.
, no. , pp. –, .
[] P. Yang, D. G. Menter, C. Cartwright et al., “Oleandrin-mediated
inhibition of human tumor cell proliferation: importance of
Na,K-ATPase 𝛼subunits as drug targets,Molecular Cancer
erapeutics,vol.,no.,pp.,.
[] P. Yang, C. Cartwright, E. Efuet et al., “Cellular location and
expression of Na+,K
+-ATPase alpha subunits aect the anti-
proliferative activity of oleandrin,Mol Carcinog,.
[] A. Svensson, F. Azarbayjani, U. B¨
ackman,T.Matsumoto,andR.
Christoerson, “Digoxin inhibits neuroblastoma tumor growth
in mice,Anticancer Research,vol.,no.,pp.,.
[] R. B. Zavareh, K. S. Lau, R. Hurren et al., “Inhibition of the
sodium/potassium ATPase impairs N-glycan expression and
function,Cancer Research, vol. , no. , pp. –, .
BioMed Research International
[]C.C.Wong,H.Zhang,D.M.Gilkesetal.,“Inhibitorsof
hypoxia-inducible factor  block breast cancer metastatic niche
formation and lung metastasis,Journal of Molecular Medicine,
vol.,no.,pp.,.
[] H. Zhang, C. C. L. Wong, H. Wei et al., “HIF--dependent
expression of angiopoietin-like  and LCAM mediates vascu lar
metastasis of hypoxic breast cancer cells to the lungs,Oncogene,
vol.,no.,pp.,.
[] L. Schito, S. Rey, M. Tafani et al., “Hypoxia-inducible factor
-dependent expression of platelet-derived growth factor B
promotes lymphatic metastasis of hypoxic breast cancer cells,
Proceedings of the National Academy of Sciences of the United
States of America,vol.,no.,pp.EE,.
[] B. A. Gayed, K. J. O. ’Malley, J. Pilch, and Z. Wang,
“Digoxin inhibits blood vessel density and HIF-a expression
in castration-resistant C- xenogra prostate tumors,Clinical
and Translational Science,vol.,no.,pp.,.
[] C. Antczak, C. Kloepping, C. Radu et al., “Revisiting old
drugs as novel agents for retinoblastoma: in vitro and in vivo
antitumor activity of cardenolides,Investigative Ophthalmolog y
and Visual Science,vol.,no.,pp.,.
[] C. D. Simpson, I. A. Mawji, K. Anyiwe et al., “Inhibition of the
sodium potassium adenosine triphosphatase pump sensitizes
cancer cells to anoikis and prevents distant tumor formation,
Cancer Research,vol.,no.,pp.,.
[] T. Mijatovic, A. O. De Beeck, E. Van Quaquebeke et al., “e
cardenolide UNBS is able to deactivate nuclear factor 𝜅B-
mediated cytoprotective eects in human non-small cell lung
cancer cells,Molecular Cancer erapeutics,vol.,no.,pp.
–, .
[] T. Mijatovic, V. Mathieu, J. Gaussin et al., “Cardenolide-
induced lysosomal membrane permeabilization demonstrates
therapeutic benets in experimental human non-small cell lung
cancers,Neoplasia,vol.,no.,pp.,.
[] T. Mijatovic, N. De N`
eve, P. Gailly et al., “Nucleolus and c-Myc:
potential targets of cardenolide-mediated antitumor activity,
Molecular Cancer erapeutics,vol.,no.,pp.,.
[] F. Lefranc, T. Mijatovic, Y. Kondo et al., “Targeting the 𝛼
subunit of the sodium pump to combat glioblastoma cells,
Neurosurgery,vol.,no.,pp.,.
[]V.Mathieu,C.Pirker,E.MartindeLassalleetal.,“e
sodium pump 𝛼 sub-unit: a disease progression-related target
for metastatic melanoma treatment,Journal of Cellular and
Molecular Medicine,vol.,no.,pp.,.
[] Z. J. Lu, Y. Zhou, Q. Song et al., “Periplocin inhibits growth of
lung cancer in vitro and in vivo by blocking AKT/ERK signaling
pathways,Cellular Physiology and Biochemistry,vol.,no.-,
pp.,.
[] C.F.Cheng,I.H.Lu,H.W.Tsengetal.,“Antitumoreectof
periplocin in TRAIL-resistant human hepatocellular carcinoma
cells through downregulation of IAPs,Evidence-Based Comple-
mentary and Alternative Medicine, vol. , Article ID ,
 pages, .
[] C. E. Badr, T. Wurdinger, J. Nilsson et al., “Lanatoside C
sensitizes glioblastoma cells to tumor necrosis factor-related
apoptosisinducing ligand and induces an alternative cell death
pathway,Neuro-Oncology, vol. , no. , pp. –, .
[] Y.Chen,Q.Guo,B.Zhang,M.Kang,Q.Xie,andY.Wu,“Bufalin
enhances the antitumor eect of gemcitabine in pancreatic
cancer,Oncology Letters, vol. , no. , pp. –, .
[] X. B. Xie, J. Q. Yin, L. L. Wen et al., “Critical role of heat shock
protein  in bufalin-induced apoptosis in human osteosarco-
mas: a proteomic-based research,” PLoS ONE,vol.,no.,
Article ID e, .
[] D. M. Zhang, J. S. Liu, M. K. Tang et al., “Bufotalin from
Venenum Bufonis inhibits growth of multidrug resistant HepG
cells through G/M cell cycle arrest and apoptosis,European
Journal of Pharmacology,vol.,no.,pp.,.
[] N. Han, J. Yang, L. Li et al., “Inhibitory activity of a phytochemi-
cally characterized fraction from streptocaulon juventas on lung
cancer in nude mice,Planta Medica,vol.,no.,pp.,
.
[]N.Takai,T.Ueda,M.Nishida,K.Nasu,andH.Narahara,
“Bufalin induces growth inhibition, cell cycle arrest and apop-
tosis in human endometrial and ovarian cancer cells,Interna-
tional Journal of Molecular Medicine,vol.,no.,pp.,
.
[] Y.B.Moreno,E.Urban,M.Gelbckeetal.,“Structure-activity
relationship analysis of bufadienolide-induced in vitro growth
inhibitory eects on mouse and human cancer cells,Journal of
Natural Products,vol.,no.,pp.,.
[] H. A. Elbaz, T. A. Stueckle, H. L. Wang et al., “Digitoxin and
a synthetic monosaccharide analog inhibit cell viability in lung
cancer cells,Toxicology and Applied Pharmacology,vol.,no.
,pp.,.
... Cardiac glycosides have been reported to inhibit the expression of EBV early antigen, with EC50 values less than 60 nM [176]. Cardiac glycosides have also shown potent anticancer activities, and there is a growing interest in repurposing them for cancer treatments [177][178][179][180][181]. Lanatoside C has specifically exhibited anti-proliferative and cytotoxic effects on cervical cancer cells [118]. ...
Article
Full-text available
Cervical cancer is the fourth most diagnosed cancer and the fourth leading cause of cancer death in women globally. Its onset and progression have been attributed to high-risk human papillomavirus (HPV) types, especially 16 and 18, while the Epstein–Barr virus (EBV) is believed to also significantly contribute to cervical cancer growth. The E6 protein associated with high-risk HPV strains, such as HPV16 and HPV18, is known for its role in promoting cervical cancer and other anogenital cancers. E6 proteins contribute to the malignant transformation of infected cells by targeting and degrading tumor suppressor proteins, especially p53. On the other hand, EBV nuclear antigen 1 (EBNA1) plays a crucial role in the maintenance and replication of the EBV genome in infected cells. EBNA1 is believed to increase HPV E6 and E7 levels, as well as c-MYC, and BIRC5 cellular genes in the HeLa cell line, implying that HPV/EBV co-infection accelerates cervical cancer onset and growth. Thus, the E6 and EBNA1 antigens of HPV and EBV, respectively, are attractive targets for cervical cancer immunotherapy. This study, therefore, virtually screened for potential drug candidates with good binding affinity to all three oncoviral proteins, HPV16 E6, HPV18 E6, and EBNA1. The compounds were further subjected to ADMET profiling, biological activity predictions, molecular dynamics (MD) simulations, and molecular mechanics Poisson–Boltzmann surface area (MM/PBSA) calculations. A total of six compounds comprising ZINC000013380012, ZINC000070454124, ZINC000014588133, ZINC000085568136, ZINC000095909247, and ZINC000085597263 demonstrated very strong affinity (≤−60 kJ/mol) to the three oncoviral proteins (EBNA1, HPV16 E6, and HPV18 E6) after being subjected to docking, MD, and MM/PBSA. These compounds demonstrated relatively stronger binding than the controls used, inhibitors of EBNA1 (VK-1727) and HPV E6 (baicalein and gossypetin). Biological activity predictions also corroborated their antineoplastic, p53-enhancing, Pin1 inhibitory, and JAK2 inhibitory activities. Further experimental testing is required to validate the ability of the shortlisted compounds to silence the insidious effects of HPV E6 and EBNA1 proteins in cervical cancers.
... Recent discoveries have proposed new signaling mechanisms of action for Na+/K + -ATPase, involving cardiac glycosides in the control of numerous essential cellular processes and revealing possible new therapeutic functions for these compounds in a variety of disorders (Prassas and Diamandis, 2008;Škubník et al., 2021). The enhanced vulnerability of cancer cells to these compounds, perhaps most notably, indicates their prospective application as cancer therapeutics, and the first generation of glycoside-based anticancer medicines is now in clinical trials (Calderón-Montaño et al., 2014). To date, 14 compounds had been isolated from the Bersama genus and their chemical structures were depicted in Figure 3. Compounds from 20-34 were isolated from the stem bark, root bark and bark of B. abyssinica (Kupchan et al., 1968;Kubo and Matsumoto, 1984;Bowen et al., 1985;Nyamboki et al., 2021). ...
Article
Full-text available
Bersama (Melianthaceae) has been used in traditional medicine for a wide range of ailments, including blood purifier, immune booster, psychotropic medication, and treatment for malaria, hepatitis, infertility, diabetes, impotency, meningitis, and stroke. This review gathers fragmented information from the literature on ethnomedicinal applications, phytochemistry, pharmacology, and toxicology of the Bersama genus. It also explores the therapeutic potential of the Bersama genus in ethnophytopharmacology, allowing for further investigation. All the available information published in the English language on Bersama genus was compiled from electronic databases such as Academic Journals, Ethnobotany, Google Scholar, PubMed, Science Direct, Web of Science, and library search using the following keywords: “Bersama genus,” “traditional use,” “phytochemistry,” “pharmacological effects,” and “toxicology”. The ethnomedical applications of the Bersama genus have been recorded, and it has been used traditionally for more than 30 different types of ailments. Thus far, more than 50 compounds have been isolated from the genus. Cardiac glycosides and terpenoids are the main compounds isolated from the Bersama genus. Different plant parts of Bersama genus extracts demonstrated a wide range of pharmacological properties, including antioxidant, antimalarial, antidiabetic, antiviral, anti-inflammatory, and cytotoxic activity. Exemplary drug leads from the genus include mangiferin and quercetin-3-O-arabinopyranoside, both of which have antioxidant activities. Bersama genus has long been used to cure a wide range of ailments. Bersama genus extracts and phytochemicals have been found to have promising pharmacological activities. Further study on promising crude extracts and compounds is required to develop innovative therapeutic candidates.
... Cardenolides are a group of biocompounds produced by plant species mainly belonging to the Apocynaceae, Plantaginaceae, and Brassicaceae families [7,8]. They exhibit a broad range of biological activities including cardiotonic and anti-tumor activities [9,10]. They are characterized by a steroid core structure and a lactone ring [11]. ...
Article
Full-text available
Specialized metabolites possess diverse interesting biological activities and some cardenolides- and monoterpene indole alkaloids- (MIAs) derived pharmaceuticals are currently used to treat human diseases such as cancers or hypertension. While these two families of biocompounds are produced by specific subfamilies of Apocynaceae, one member of this medicinal plant family, the succulent tree Pachypodium lamerei Drake (also known as Madagascar palm), does not produce such specialized metabolites. To explore the evolutionary paths that have led to the emergence and loss of cardenolide and MIA biosynthesis in Apocynaceae, we sequenced and assembled the P. lamerei genome by combining Oxford Nanopore Technologies long-reads and Illumina short-reads. Phylogenomics revealed that, among the Apocynaceae whose genomes have been sequenced, the Madagascar palm is so far the species closest to the common ancestor between MIA producers/non-MIA producers. Transposable elements, constituting 72.48% of the genome, emerge as potential key players in shaping genomic architecture and influencing specialized metabolic pathways. The absence of crucial MIA biosynthetic genes such as strictosidine synthase in P. lamerei and non-Rauvolfioideae species hints at a transposon-mediated mechanism behind gene loss. Phylogenetic analysis not only showcases the evolutionary divergence of specialized metabolite biosynthesis within Apocynaceae but also underscores the role of transposable elements in this intricate process. Moreover, we shed light on the low conservation of enzymes involved in the final stages of MIA biosynthesis in the distinct MIA-producing plant families, inferring independent gains of these specialized enzymes along the evolution of these medicinal plant clades. Overall, this study marks a leap forward in understanding the genomic dynamics underpinning the evolution of specialized metabolites biosynthesis in the Apocynaceae family, with transposons emerging as potential architects of genomics restructuring and gene loss.
... Cardiac glycosides have been shown to reduce tumor size, trigger apoptosis, and make cancer cells more susceptible to treatment. Oleandrin, a cardiac glycoside derived from Nerium oleander, has also demonstrated potential in slowing the expansion of several cancer cell lines [14][15][16]. Other class flavonoids glycosides, quercetin glycosides have been demonstrated to trigger apoptosis and cell cycle arrest in cancer cells in addition to having antioxidant capabilities. ...
... In herbal medicine, cosmetics, phytosterol are important component (Sharma et al. 2021). Cardiac glycosides are considered as cancer (Calderón-Montaño et al. 2014) and cardiac therapeutics agent (Hou et al. 2021, Sharma et al. 2021. Coumarins is well known for its anti-hypertensive (Jebir & Mustafa 2022), anti-Inflammatory (Padureanu et al. 2019), antidiabetic (Li et al. 2017), anticoagulant (Sharifi-Rad et al. 2020), anticancer (Buga et al. 2019), antibacterial (Jebir & Mustafa 2022), antifungal (Chou et al. 2007). ...
Article
Full-text available
Background: India is a land of diverse ethnicity with plethora of ethnic foods. One of the ethnic foods is Posa kumura(a form prepared from matured fruit of Benincasa hispida) which is consumed in Assam. However, being not so popular, till date the there is no written document to provide an evidence of its origin. Methods: The present ethnobotanical study consists of both online (Google form) and offline (field visits and interviews) survey with 918 informers of the state having different gender, age groups and occupation through semi- structured questionnaires along withIn this article an attempt has been made to document the contemporary process of making Posa kumura by means of survey through a semi-structured questionnaire along with the preliminary phytochemical analysis. Results: The findings revealed that of 918 respondents (559 male, 358 female and 01 transgender) from 35 districts of Assam, 372 consumes Posa kumura in various forms. Of the 372 people, 75.81% opined that the matured Benincasa hispida (Thunb.) Cogn. is placed in shade for varying periods of time for conversion to Posa kumura. Though the production process reported is similar, it is different in terms of how people intend to consume. Frying of Posa kumura (49%) was the most favoured mode of consumption followed by curry (27%), dry (6%) and pitha (6%). The age-old health benefits claimed by the consumer of Posa kumura includes improved digestion, anti-diabetic, promotes weight loss, etc. The preliminary phytochemical analysis revealed the presence of carbohydrates, reducing sugars, alkaloids, flavonoids, amino acids, phytosterols, saponins, coumarins. However, phenolics, tannins, phlobatannins, triterpenoids, lignins, quinones, anthraquinones, resins, fixed oils and fats were absent. Besides, Posa kumura also enhances milk production and helps prevent foot and mouth disease of cows. Conclusion: Thus, it can be inferred that the functional ingredients of this food lead to improvement in health in a holistic way.
... Both South African and Nigerian extracts revealed richness of cardiac glycosides, whereas Nigerian DCM and MeOH extracts showed greater richness compared to the SADCM and MeOH extracts. This observation partly validates the use of the extracts against cancer, as it is known that cardiac glycosides have potent activity against every cancer cell line [14,15]. Saponins are strongly present in the methanol extract compared to the DCM extracts. ...
Article
Full-text available
Loranthus micranthus Linn. (Loranthaceae) is a botanically significant hemiparasite that grows on tree branches or trunks and is used in traditional medicine. This study compares the antioxidant activity and qualitative phytochemical screening of Nigerian and South Africa Loranthus micranthus Linn. Standard techniques for phytochemical screening were deployed while thin layer chromatography (TLC) bio-autography was utilized to analyze antioxidants qualitatively. Quantitative antioxidant analysis was performed using 2,2-diphenyl-1-picrylhydrazyl (DPPH); hydrogen peroxide (H2O2) free-radical scavenging; and ferric chloride reducing power. The results of qualitative phytochemical screening revealed the presence of flavonoids; glycosides; saponins; phenolic compounds; phlobatannins; tannins; and terpenoids. As for the antioxidant potentials of the four extracts—i.e., Nigerian dichloromethane (NGDCM); South African dichloromethane (SADCM); Nigerian methanol (NGMeOH); and South African methanol (SAMeOH)—the SADCM extract had more creamy bands compared to the Nigerian, thus indicating potentially more antioxidant compounds. A better complementary antioxidant potential was observed for the Nigerian methanol extracts over their South African counterparts. The DPPH quantitative analyses underpin that the SADCM exhibited greater scavenging activity compared to the NGDCM, but this was less than the gallic acid control, with the highest activity at a concentration of 0.2 mg/mL and 0.4 mg/mL, respectively. However, as the concentration increased from 0.6 to 1.0 mg/mL, the SADCM again dominated in its antioxidant potential over all the analytes. The half maximal inhibitory concentration (IC50) values obtained were [SADCM = 0.31 mg/mL, NGDCM = 0.51 mg/mL, SAMeOH = 0.51 mg/mL, NGMeO = 0.17, gallic acid = 1.17 mg/mL, and BHT = 1.47 Mg/mL)]. Both the H2O2 scavenging and the Fe3+ to Fe2+ reduction assays mirrored a similar trend in the antioxidant potentials of all the analytes except for the BHT, which performed better in the ferric chloride reduction assay at a concentration of 0.2–06 mg/mL. Based on the facts gathered, it can be inferred that the South African and Nigerian Loranthus micranthus Linn. are chemically equivalent. This is in support of their similar morphology and taxonomical classification, notwithstanding the environmental, biological, and edaphic impacts experienced by each plant.
... Bufalin is an effective anti-cancer agent that has various biological activities. Its structure is similar to digitalis toxin (10)(11)(12). Some studies have demonstrated that bufalin exerts significant anti-tumor activity by inhibiting cell proliferation and angiogenesis and inducing programmed cell death (13)(14)(15). ...
Article
Full-text available
Background: Epithelial-mesenchymal transition (EMT) is a biological process involved in tumor migration, invasion, and radiotherapy resistance. Bufalin can affect the proliferation, apoptosis and invasion of tumor cells by regulating multiple signaling pathways. Whether bufalin can increase radiosensitivity through EMT deserves further investigation. Methods: In this study, we investigated the effect of bufalin on the EMT and radiosensitivity of non-small cell lung cancer (NSCLC) and the underlying molecular mechanism. NSCLC cells were treated with bufalin (at a dose of 0-100 nM) or irradiated with 6 MV X-rays (4 Gy/min). The effects of bufalin on cell survival, cell cycle, radiosensitivity, cell migration, and invasion were detected. Western blot was used to analyze the gene expression changes of Src signaling in NSCLC cell induced by Bufalin. Results: Bufalin significantly inhibited cell survival, migration, and invasion and induced G2/M arrest and apoptosis. Cells co-treated with bufalin and radiation manifested a higher inhibitory effect compared to those treated with radiation or bufalin alone. Furthermore, the levels of p-Src and p-STAT3 were considerably reduced following bufalin treatment. Interestingly, elevated p-Src and p-STAT3 were observed in cells treated with radiation. Bufalin inhibited radiation-induced p-Src and p-STAT3, whereas the knockdown of Src abrogated the effects of bufalin on cell migration, invasion, EMT, and radiosensitivity. Conclusions: Bufalin inhibits EMT and enhances radiosensitivity through targeting Src signaling in NSCLC.
Chapter
Phytoconstituents are compounds present in plants which performs various functions to protect plant’s vitality. Besides awarding tremendous benefits to plants, phytoconstituents also provides several benefits to human beings. There are two types of metabolites synthesized in plants, primary and secondary. Primary metabolites are essential for the growth and development of the plant while secondary metabolites play important role in plant defence. These secondary metabolites or phytochemicals are used as drugs, detoxifying agents, flavourings, cardioprotective agents, antioxidants, anti-cancer agents, antimicrobial agent etc. phytoconstituents play a significant role in the secondary metabolism of plants. They only exist in trace amounts and typically have pharmacological effects. They have health promoting properties including antioxidant, anti-inflammatory and disease preventing properties too. Moreover, they also serve as attractants for pollinators and seed-dispersing agents. Major classes of secondary metabolites are terpenes, phenolic compounds, glycosides and alkaloids. Phenolic compounds are the largest group of phytoconstituents.
Chapter
Breast cancer is an aggressive and primary cause of death among women globally. Triple negative breast cancer (TNBC) is one of the sub types of breast cancer. TNBC lacks the expression of progesterone receptor (PR), estrogen receptor (ER), and human epidermal factor 2 (HER2), which leads to poor diagnosis resulting in lack of targeted therapies. On the other hand, natural products are also cost efficient, non-toxic, and abundantly available in nature. Natural products have also been reported to exert various pharmacological activities including cardioprotective, anti-diabetic, antimicrobial, anti-inflammatory, etc. In this chapter, summarization of 12 well known natural products such as chebulinic acid, maslinic acid, apigenin, piperlongumine, Liquiritigenin, berberine, icariin, bufalin, which are targeted against TNBC through regulation of different pathways, and their mechanism are briefly explained. These natural products are already used to treat various diseases at the preclinical level and also have shown to have effective anti-tumor effect and can act as potent anti-TNBC agents.
Article
Full-text available
Cardiotonic steroids (CTS), specific inhibitors of Na,K-ATPase activity, have been widely used for treating cardiac insufficiency. Recent studies suggest that low levels of endogenous CTS do not inhibit Na,K-ATPase activity but play a role in regulating blood pressure, inducing cellular kinase activity, and promoting cell viability. Higher CTS concentrations inhibit Na,K-ATPase activity and can induce reactive oxygen species, growth arrest, and cell death. CTS are being considered as potential novel therapies in cancer treatment, as they have been shown to limit tumor cell growth. However, there is a lack of information on the relative toxicity of tumor cells and comparable non-tumor cells. We have investigated the effects of CTS compounds, ouabain, digitoxin, and bufalin, on cell growth and survival in cell lines exhibiting the full spectrum of non-cancerous to malignant phenotypes. We show that CTS inhibit membrane Na,K-ATPase activity equally well in all cell lines tested regardless of metastatic potential. In contrast, the cellular responses to the drugs are different in non-tumor and tumor cells. Ouabain causes greater inhibition of proliferation and more extensive apoptosis in non-tumor breast cells compared to malignant or oncogene-transfected cells. In tumor cells, the effects of ouabain are accompanied by activation of anti-apoptotic ERK1/2. However, ERK1/2 or Src inhibition does not sensitize tumor cells to CTS cytotoxicity, suggesting that other mechanisms provide protection to the tumor cells. Reduced CTS-sensitivity in breast tumor cells compared to non-tumor cells indicates that CTS are not good candidates as cancer therapies.
Article
Full-text available
Recent evidence suggests that cardiac glycosides might be used for the treatment of cancer. The ornamental shrub Nerium oleander has been used in traditional medicine for treating several disorders including cancer, and extracts from the leaves of this plant have already entered phase I clinical trials. In this communication, we have prepared a hydroalcoholic extract from the leaves of Nerium oleander (containing 4.75 ± 0.32 % of cardenolides) and have assessed its cytotoxic activity in A549 lung cancer cells vs. MRC5 nonmalignant lung fibroblasts. The results showed that the cytotoxicity of the Nerium oleander extract against the cancer cell line was significantly higher than that against the nonmalignant cell line, with a potency and selectivity similar to those of the anticancer drug cisplatin. Pretreatment of A549 cells with the antioxidants N-acetylcysteine and catalase slightly prevented the cytotoxicity of the extract, therefore suggesting that the formation of reactive oxygen species participates in its cytotoxic activity but does not play a major role. Nerium oleander extract-induced cytotoxicity and DNA damage (gamma-H2AX focus formation) were slightly higher in cells lacking BRCA2 (deficient in homologous recombination repair) than in parental cells; this indicates that the induction of DNA damage may also play a role in the cytotoxicity of the extract. Nerium oleander extract induced a marked inhibition of glycolysis (glucose consumption and lactate production) in A549 cells, comparable to that of the glycolysis inhibitor dichloroacetate (currently in clinical development for cancer therapy). Because platinum compounds are widely used in the treatment of lung cancer, we tested the cytotoxicity of several combinations of cisplatin with the extract and found a moderate synergism when Nerium oleander extract was administered after cisplatin but a moderate antagonism when it was added before cisplatin. Our results suggest that extracts from Nerium oleander might induce anticancer effects in patients with lung cancer and support their possible advancement into phase II clinical trials for the treatment of this type of cancer.
Article
Full-text available
Oncogene is one of the world’s leading cancer journals. It is published weekly and covers all aspects of the structure and function of Oncogenes.
Article
Full-text available
The BJC is owned by Cancer Research UK, a charity dedicated to understanding the causes, prevention and treatment of cancer and to making sure that the best new treatments reach patients in the clinic as quickly as possible. The journal reflects these aims. It was founded more than fifty years ago and, from the start, its far-sighted mission was to encourage communication of the very best cancer research from laboratories and clinics in all countries. The breadth of its coverage, its editorial independence and it consistent high standards, have made BJC one of the world's premier general cancer journals. Its increasing popularity is reflected by a steadily rising impact factor.
Article
Full-text available
Cardiac glycosides represent group of compounds isolated from plants and some animals. They have been using in the therapy of heart failure for many years. In spite of the fact that cytotoxic effect of many cardiac glycosides has been demonstrated. The mechanism of the cytotoxic action is very complicated and complex, where Na+/K+-ATPase plays crucial role. On the other hand, Na+/K+-ATPase is regulated by many endogenous factors including hormones or FXYD proteins, which role in the regulation of cell cycle is intensively studied. This review focuses the role of Na+/K+-ATPase in the regulation of cell growth, cell cycle and cell proliferation and involvement of cardiac glycosides in the regulation of Na+/K+-ATPase. Cytotoxic effect of cardiac glycosides is discussed in the connection with possible apoptotic mechanisms induced by these compounds. Novel strategies in cancer therapy based on the cardiac glycosides as well as possibilities in the overcoming multidrug resistance by cardiac glycosides are discussed too. The goal of this review is to present cardiac glycosides as not only pharmaceuticals used in heart failure, but also as potent cytotoxic agents with possible involvement in cancer treatment.
Article
Full-text available
Many cancer patients fail to respond to chemotherapy because of the intrinsic resistance of their cancer to pro-apoptotic stimuli or the acquisition of the multidrug resistant phenotype during chronic treatment. Previous data from our groups and from others point to the sodium/potassium pump (the Na+/K+-ATPase, i.e., NaK) with its highly specific ligands (i.e., cardiotonic steroids) as a new target for combating cancers associated with dismal prognoses, including gliomas, melanomas, non-small cell lung cancers, renal cell carcinomas, and colon cancers. Cardiotonic steroid-mediated Na+/K+-ATPase targeting could circumvent various resistance pathways. The most probable pathways include the involvement of Na+/K+-ATPase β subunits in invasion features and Na+/K+-ATPase α subunits in chemosensitisation by specific cardiotonic steroid-mediated apoptosis and anoïkis-sensitisation; the regulation of the expression of multidrug resistant-related genes; post-translational regulation, including glycosylation and ubiquitinylation of multidrug resistant-related proteins; c-Myc downregulation; hypoxia-inducible factor downregulation; NF-κB downregulation and deactivation; the inhibition of the glycolytic pathway with a reduction of intra-cellular ATP levels and an induction of non-apoptotic cell death. The aims of this review are to examine the various molecular pathways by which the NaK targeting can be more deleterious to biologically aggressive cancer cells than to normal cells.
Article
Na+/K+-ATPase as an energy transducing ion pump has been studied extensively since its discovery in 1957. Although early findings suggested a role for Na+/K+-ATPase in regulation of cell growth and expression of various genes, only in recent years the mechanisms through which this plasma membrane enzyme communicates with the nucleus have been studied. This research, carried out mostly on cardiac myocytes, shows that in addition to pumping ions, Na+/K+-ATPase interacts with neighboring membrane proteins and organized cytosolic cascades of signaling proteins to send messages to the intracellular organelles. The signaling pathways that are rapidly elicited by the interaction of ouabain with Na+/K+-ATPase, and are independent of changes in intracellular Na+ and K+ concentrations, include activation of Src kinase, transactivation of the epidermal growth factor receptor by Src, activation of Ras and p42/44 mitogen-activated protein kinases, and increased generation of reactive oxygen species by mitochondria. In cardiac myocytes, the resulting downstream events include the induction of some early response proto-oncogenes, activation of the transcription factors, activator protein-1 and nuclear factor kappa-B, regulation of a number of cardiac growth-related genes, and stimulation of protein synthesis and myocyte hypertrophy. For these downstream events, the induced reactive oxygen species and rise in intracellular Ca2+ are essential second messengers. In cells other than cardiac myocytes, the proximal pathways linked to Na+/K+-ATPase through protein–protein interactions are similar to those reported in myocytes, but the downstream events and consequences may be significantly different. The likely extracellular physiological stimuli for the signal transducing function of Na+/K+-ATPase are the endogenous ouabain-like hormones, and changes in extracellular K+ concentration.
Article
Virtually all transcription factors partner with coactivators that recruit chromatin remodeling factors and interact with the basal transcription machinery. Coactivators have been implicated in cancer cell proliferation, invasion and metastasis, including the p160 steroid receptor coactivator (SRC) family comprised of SRC-1 (NCOA1), SRC-2 (TIF2/GRIP1/NCOA2), and SRC-3 (AIB1/ACTR/NCOA3). Given their broad involvement in many cancers, they represent candidate molecular targets for new chemotherapeutics. Here we report on the results of a high throughput screening effort which identified the cardiac glycoside bufalin as a potent small molecule inhibitor for SRC-3 and SRC-1. Bufalin strongly promoted SRC-3 protein degradation and was able to block cancer cell growth at nanomolar concentrations. When incorporated into a nanoparticle delivery system, bufalin was able to reduce tumor growth in a mouse xenograft model of breast cancer. Our work identifies bufalin as apotentially broad-spectrum small molecule inhibitor for cancer.
Article
Cardiac glycosides have a long history in the treatment of cardiac disease. However, several preclinical studies as well as two phase I studies have shown that cardenolides may also possess anticancer effects. The mechanisms of these anticancer effects may include intracellular decrease of K(+) and increase of Na(+) and Ca(2+); intracellular acidification; inhibition of IL-8 production and of the TNF-α/NF-κB pathway; inhibition of DNA topoisomerase II and activation of the Src kinase pathway. To date three cardiac glycosides have been developed for treatment of cancer and were tested in a phase 1 clinical trial to determine dose limiting toxicities and maximum tolerated dose. Future studies of this novel class of anticancer drugs are warranted to determine their possible role in cancer treatment.
Article
The in vitro growth inhibitory effects of 27 bufadienolides and eight degradation products, with two cardenolides (ouabain and digoxin) chosen as reference compounds, were analyzed by means of an MTT colorimetric assay in six human and two mouse cancer cell lines. A structure-activity analysis was then performed to highlight the most important substituents relating to the in vitro growth inhibitory activity of bufadienolides in cancer cells. Thus, the current study revealed that various bufadienolides, including gamabufotalin rhamnoside (1a), bufotalin (2a), and hellebrin (3a), displayed higher growth inhibitory activities for various human cancer cell lines when compared to ouabain and digoxin. Gamabufotalin rhamnoside (1a) was the only compound that displayed growth inhibitory effects of <1 μM in mouse cancer cells that expressed mutated forms of the Na(+),K(+)-ATPase α-1 subunit. In addition, all genins and degradation products displayed weaker (if any) in vitro growth inhibitory effects on cancer cells when compared to their respective glycosylated homologue, with the exception of hellebrigenin (3b), which was as active as hellebrin (3a).