Sie sind auf Seite 1von 11

Bisphosphonates: Preclinical Review

JONATHAN R. GREEN
Novartis Pharma AG, Basel, Switzerland
Key Words. Bisphosphonates Zoledronic acid Bone resorption Apoptosis Antitumor effects Mevalonate pathway
ABSTRACT
Bisphosphonates effectively inhibit osteoclast-medi-
ated bone resorption and are integral in the treatment of
benign and malignant bone diseases. The evolution of bis-
phosphonates over the past 30 years has led to the devel-
opment of nitrogen-containing bisphosphonates (N-BPs),
which have a mechanism of action different from that of
the nonnitrogen-containing bisphosphonates. Studies
conducted over the past decade have elucidated the
mechanism of action and pharmacologic properties of the
N-BPs. N-BPs exert their effects on osteoclasts and tumor
cells by inhibiting a key enzyme in the mevalonate path-
way, farnesyl diphosphate synthase, thus preventing pro-
tein prenylation and activation of intracellular signaling
proteins such as Ras. Recent evidence suggests that
N-BPs also induce production of a unique adenosine
triphosphate analogue (Apppi) that can directly induce
apoptosis. Our increased understanding of the pharma-
cologic effects of bisphosphonates is shedding light on the
mechanisms by which they exert antitumor effects. As a
result of their biochemical effects on protein prenylation,
N-BPs induce caspase-dependent apoptosis, inhibit
matrix metalloproteinase activity, and downregulate

3
and
v

5
integrins. In addition, zoledronic acid
(Zometa

; Novartis Pharmaceuticals Corp.; East


Hanover, NJ and Basel, Switzerland) exerts synergistic
antitumor activity when combined with other anticancer
agents. Zoledronic acid also inhibits tumor cell adhesion
to the extracellular matrix and invasion through
Matrigel and has antiangiogenic activity. A growing
body of evidence from animal models demonstrates that
zoledronic acid and other bisphosphonates can reduce
skeletal tumor burden and prevent metastasis to bone.
Further studies are needed to fully elucidate these bio-
chemical mechanisms and to determine if the antitumor
potential of bisphosphonates translates to the clinical set-
ting. The Oncologist 2004;9(suppl 4):3-13
The Oncologist 2004;9(suppl 4):3-13 www.TheOncologist.com
Correspondence: Jonathan R. Green, Ph.D., Novartis Pharma AG, Klybeckstrasse 141, WKL-125.901, CH-4002 Basel,
Switzerland. Telephone: 41-61-696-4415; Fax: 41-61-696-3849; e-mail: jonathan.green@pharma.novartis.com Received
July 19, 2004; accepted for publication August 3, 2004. AlphaMed Press 1083-7159/2004/$12.00/0
INTRODUCTION
Bisphosphonates are ideally suited for the treatment of
metabolic bone disease because they bind avidly to the bone
mineral at sites of active bone metabolism, where they
achieve therapeutic concentrations. The pioneering work of
Fleisch and colleagues showed that bisphosphonates not only
inhibit dissolution of hydroxyapatite crystals, but also affect
osteoclast metabolism and function [1-3]. Bisphosphonates
The
Oncologist

LEARNING OBJECTIVES
After completing this course, the reader will be able to:
1. Describe the mechanism of action of first-generation and nitrogen-containing bisphosphonates.
2. Explain how the mechanism of action of the bisphosphonates might directly affect tumor growth.
3. Discuss how the bisphosphonates might be incorporated into both the prevention and treatment of cancer.
Access and take the CME test online and receive 1 hour of AMA PRA category 1 credit at CME.TheOncologist.com CME CME
This material is protected by U.S. Copyright law.
Unauthorized reproduction is prohibited.
For reprints contact: Reprints@AlphaMedPress.com
are released during bone resorption and are internalized by
osteoclasts, leading to inhibition of bone resorption and
induction of osteoclast apoptosis [4-6].
There is now extensive preclinical evidence that bis-
phosphonates also have antitumor activity, as evidenced by
reduced proliferation and viability of tumor cell lines in
vitro and reduced skeletal tumor burden and slower pro-
gression of bone lesions in animal models. Several mecha-
nisms have been proposed to explain these observations.
Bisphosphonates may render the bone a less favorable
microenvironment for tumor cell growth by reducing
tumor-induced osteolysis and local release of growth fac-
tors, and bisphosphonates may also have direct antitumor
effects. Bisphosphonates inhibit proliferation and induce
apoptosis of a variety of human tumor cell lines in vitro [7-
17]. Bisphosphonates also inhibit tumor cell adhesion to the
extracellular bone matrix, inhibit invasion of tumor cells
through Matrigel

, and have antiangiogenic and immuno-


modulatory activities. Consistent with these findings,
bisphosphonates have been shown to inhibit the formation
or progression of bone metastases and/or reduce skeletal
tumor burden in a variety of animal models. The mecha-
nisms responsible for the observed antitumor effects of
bisphosphonates are beginning to be elucidated.
MECHANISM OF ACTION
Bisphosphonates accumulate in the mineralized bone
matrix and are released during bone resorption. First-gener-
ation, nonnitrogen-containing bisphosphonates are metabo-
lized by osteoclasts to nonhydrolyzable cytotoxic ATP
analogues [18-21]. For example, clodronate is metabolized
to AppCC12p, which, at high concentrations, inhibits mito-
chondrial ATP/adenosine diphosphate (ADP) translocase,
thereby causing loss of the mitochondrial membrane poten-
tial and direct induction of apoptosis [22-24]. The high
affinity of bisphosphonates for bone mineral and subse-
quent uptake by activated osteoclasts during bone resorp-
tion ensures that cytotoxic concentrations of these
metabolites only accumulate within osteoclasts. However,
they may also accumulate within tumor cells growing in the
bone because tumor cells stimulate osteolysis.
In contrast, nitrogen-containing bisphosphonates (N-BPs),
which include pamidronate (Aredia

; Novartis Pharma-
ceuticals Corp.; East Hanover, NJ), alendronate (Fosamax

;
Merck and Company, Inc.; West Point, PA), ibandronate
(Bondronat

; Hoffmann-La Roche Inc.; Nutley, NJ), rise-


dronate (Actonel

; Proctor and Gamble Pharmaceuticals, Inc.;


Cincinnati, OH), and zoledronic acid (Zometa

; Novartis
Pharmaceuticals Corp.), affect osteoclast activity and survival
through a different mechanism. After internalization, these
compounds inhibit a key enzyme, farnesyl diphosphonate
(FPP) synthase, in the biosynthetic mevalonate pathway (Fig.
1) [5, 25-28]. As a result, N-BPs interfere with a variety of
cellular functions essential for the bone-resorbing activity
and survival of osteoclasts [28, 29]. Several intermediates in
this pathway, including farnesyl pyrophosphate and geranyl-
geranyl pyrophosphate, are required for the posttranslational
modification (i.e., prenylation) of guanosine triphosphate-
binding proteins such as Ras, Rho, and Rac [30]. These sig-
naling molecules are involved in the regulation of cell
proliferation, cell survival, and cytoskeletal organization [26,
28, 31, 32]. In particular, inhibition of protein prenylation
and Ras signaling within osteoclasts leads to defects in intra-
cellular vesicle transport [33]. As a result, osteoclasts cannot
form a tight-sealing zone or ruffled borders, which are
required for bone resorption.
Recent studies have shown that N-BPs may have yet
another mechanism of action. As shown in Figure 1 [25, 34],
N-BPs also induce production of an intracellular ATP ana-
logue known as Apppi (triphosphoric acid 1-adenosin-5-yl
ester 3-[3-methylbut-3-enyl] ester), which may directly
induce apoptosis similar to AppCC12p (i.e., a metabolite of
clodronate) [34]. Inhibition of FPP synthase results in accu-
mulation of isopentenyl diphosphonate, which can be
metabolized to Apppi. The ability of various bisphospho-
nates (at 0.1-M concentrations) to inhibit recombinant
human FPP synthase activity (Fig. 2) [35] correlates well
with their ability to induce production of Apppi in J774
4 Bisphosphonates: Preclinical Review
Statins
HMG CoA
reductase
HMG CoA
Mevalonate
Mevalonate
pyrophosphate
Dimethylallyl
pyrophosphate
Isopentenyl
pyrophosphate
+ AMP Apppi
Geranyl
pyrophosphate
FPP-synthase N-BPs
Farnesyl
pyrophosphate
Farnesol
Dolichol
Ubiquinone
Protein
farnesylation
Squalene
Cholesterol
Geranyl-Geranyl
pyrophosphate
Protein geranyl-
geranylation
Figure 1. Schematic representation of the mevalonate pathway and
the effects of nitrogen-containing bisphosphonates. Abbreviation:
HMG CoA = 3-hydroxy-3-methylglutaryl coenzymeA. Adapted with
permission from Gober et al. [25].
cells. In this assay, clodronate serves as a negative control
with little or no effect on FPP synthase activity, and it
induces no Apppi production. Of the N-BPs tested, zole-
dronic acid demonstrated the highest potency in terms of
both inhibition of FPP synthase activity (Fig. 2) [35] and
production of Apppi [34]. The implication of this work is
that N-BPs, by inhibiting FPP synthase, can potentially
induce apoptosis of osteoclasts and tumor cells by at least
two distinct pathways.
It has been demonstrated that the potency of various
N-BPs with respect to inhibition of FPP synthase activity
also correlates well with their observed potency in terms of
inhibiting bone resorption in vitro (Table 1) [35, 36]. Among
the N-BPs tested, zoledronic acid was the most potent
inhibitor of FPP synthase activity, followed by risedronate
and ibandronate. The relative potencies of these three N-BPs
in the FPP synthase assay correlate closely with their relative
potencies in terms of inhibiting vitamin D
3
-induced calcium
release from mouse calvaria cultures [36]. These data suggest
that inhibition of FPP synthase is a central mechanism by
which N-BPs inhibit osteoclast-mediated bone resorption.
Moreover, osteoclasts isolated from animals treated with N-
BPs show a profound suppression of FPP synthase activity
[37]. Therefore, FPP synthase appears to be one of the key
molecular targets of N-BPs. Inhibition of protein prenylation
in cancer cells is also thought to be responsible for many of
the observed antitumor effects of N-BPs.
EVIDENCE OF ANTITUMOR EFFECTS
In Vitro Studies
Bisphosphonates have demonstrated direct antitumor
activity against a variety of tumor cell lines at concentrations
ranging from 5-2,000 M [38]. Bisphosphonates cause dose-
and time-dependent inhibition of proliferation and induce
apoptosis of myeloma, breast cancer, prostate cancer, pancre-
atic cancer, lung cancer, and osteosarcoma cell lines in vitro
[7-17, 39]. For example, in studies with the human MDA-
MB-231 breast cancer cell line, clodronate, pamidronate, and
zoledronic acid demonstrated dose-dependent effects on cell
viability, with 50% inhibitory concentration (IC
50
) values of
700, 40, and 15 M, respectively [12]. This effect was not
caused by calcium chelation and appeared to be specific to
tumor cells. Similar results have been observed with other
breast cancer cell lines. For instance, dose-dependent
increases in the proportion of apoptotic cells have been
observed when Hs 578T cells were incubated with zoledronic
acid [12]. Studies with MCF-7 cells have further shown that
the effects of zoledronic acid on tumor cell viability and apop-
tosis could be reversed by incubation with geranylgeraniol,
indicating that inhibition of protein prenylation can induce
tumor cell apoptosis [39]. In vitro studies with several differ-
ent prostate cancer cell lines, including PC-3 and LNCaP
23.1, have shown that zoledronic acid inhibits proliferation,
induces apoptosis, and causes cell-cycle arrest in a dose-
dependent manner [40, 41]. These are just a few examples of
the antitumor activity of bisphosphonates against a wide vari-
ety of human and murine tumor cell lines, and in every
instance where several bisphosphonates have been tested,
zoledronic acid has demonstrated the most potent activity.
In vitro studies have also shown that combining N-BPs
with a variety of standard anticancer agents results in addi-
tive or synergistic antitumor effects against a range of
tumor cell lines [39, 42-49]. Jagdev et al. [39] were the first
to show that the combination of zoledronic acid (10 M)
plus paclitaxel (Taxol

; Bristol-Myers Squibb; Princeton,


NJ; 2 nM) enhanced apoptosis of MCF-7 breast cancer cells
Green 5
F
P
P

s
y
n
t
h
a
s
e

(
%

c
o
n
t
r
o
l
)
100
75
50
25
0
Clodronate Ibandronate Risedronate Zoledronic acid
*
*
*
Figure 2. The effects of equimolar concentrations (0.1 m) of
bisphosphonates on recombinant human FPP synthase activity.
*p < 0.001. Adapted with permission from Dunford et al. [35].
Table 1. Relative potencies of bisphosphonates with respect to
inhibition of FPP synthase and bone resorption activity
FPP synthase Bone resorption
Bisphosphonate IC
50
(M)
a
IC
50
(M)
b
Zoledronic acid 0.02 0.002
Risedronate 0.10 0.01
Ibandronate 0.31 0.02
Alendronate 0.50 0.05
Pamidronate 0.85 0.2
a
Mean values calculated from dose-response plots of inhibition of
FPP synthase in J774 cell homogenates based on three experiments.
Data from Dunford et al. [35].
b
Inhibition of 1,25-dihydroxyvitamin D
3
-induced calcium release
from mouse calvaria in vitro. Data represent means of several
experiments. Data from Green et al. [36].
fourfold compared with either agent alone. Similar results
were reported when ibandronate or zoledronic acid were
combined with epirubicin (Ellence

; Pharmacia and
Upjohn; Portage, MI) plus docetaxel (Taxotere

; Aventis
Pharmaceuticals Inc.; Bridgewater, NJ) [50]. In cultures of
primary breast cancer cells, the concentrations of epirubicin
plus docetaxel were gradually reduced to suboptimal levels
by serial dilution until there was little or no inhibition of
tumor cell growth with these two drugs alone. However, the
addition of 15 M ibandronate or zoledronic acid resulted in
35% and 70% inhibitions of tumor cell growth, respectively.
Combinations of zoledronic acid with taxanes also have
demonstrated synergistic antitumor activity against prostate
cancer cell lines. The combination of zoledronic acid
(12.5 or 25 M) with suboptimal concentrations of doc-
etaxel (0.1 ng/ml) demonstrated additive and dose-depen-
dent cytotoxic effects on PC-3 prostate cancer cells at 72
hours [45]. In addition, recent studies with DU-145 prostate
cancer and MCF-18 breast cancer cell lines have demon-
strated that the combination of zoledronic acid with the
cyclooxygenase-2 inhibitor SC236 had additive inhibitory
effects on tumor cell growth [48, 49]. These and many other
studies have shown that N-BPs can enhance the cytotoxic
and cytostatic effects of standard anticancer agents used to
treat a variety of solid tumors. Recently, zoledronic acid
also has been shown to possess antileukemic activity
against a Philadelphia chromosome-positive cell line, and
the combination of zoledronic acid with imatinib mesylate
(Gleevec

; Novartis Pharmaceuticals Corp.) demonstrated


synergistic antileukemic activity [51]. These findings are
intriguing given that Bcr-Abl stimulates the Ras signaling
pathway.
Two recent studies with breast cancer cell lines have
begun to shed light on the possible mechanisms underlying
the observed synergy between N-BPs and anticancer agents.
In the first of these studies, MCF-7 breast cancer cells were
incubated with zoledronic acid (25 M) and doxorubicin
(Adriamycin

; Pharmacia and Upjohn; Peapack, NJ; 0.05


M), and the effects of different sequences and incubation
periods were tested [43]. The only combination that resulted
in synergistic enhancement of apoptosis was doxorubicin for
24 hours followed by zoledronic acid for 1 hour, whereas
zoledronic acid administered either before doxorubicin or
together with doxorubicin did not increase apoptosis (Fig. 3)
[43]. This suggests that exposure to doxorubicin at a concen-
tration that was not cytotoxic sensitized tumor cells to the
cytotoxic effects of zoledronic acid. A similar but opposite
effect of sequencing was also reported by the same group
when zoledronic acid (25 M) was combined with tumor
necrosis factor (TNF)-related apoptosis inducing ligand
(TRAIL) at a concentration of 10 ng/ml. Among five combi-
nations tested, the only combination that produced synergis-
tic apoptotic effects was zoledronic acid for 48 hours fol-
lowed by TRAIL for 24 hours. This combination yielded
14.7% apoptotic cells compared with 0.7% for zoledronic
acid alone and 2.7% for TRAIL alone (p < 0.001). The
intriguing results of these in vitro studies suggest a novel
approach to enhance the synergy between N-BPs and
chemotherapeutic agents in the clinical setting.
Animal Models
These in vitro findings are supported by data from many
animal models showing that the newer N-BPs can significantly
reduce the number and size of osteolytic lesions in tumor-bear-
ing mice, reduce skeletal tumor burden, induce tumor cell
apoptosis in bone lesions, reduce serum levels of tumor mark-
ers, and prevent formation of bone metastases (Table 2) [9, 40,
52-60]. Some models have even shown effects on visceral
tumors and an improvement in survival of tumor-bearing
mice, but these findings have been less consistently observed.
Animal studies have focused on models of multiple myeloma,
breast cancer, and prostate cancer. Using radiographic, histo-
logic, and histomorphometric techniques, these studies have
shown that N-BPs can inhibit the formation or progression of
bone metastases and/or reduce skeletal tumor burden.
Bisphosphonates have been administered either at the time of
tumor cell inoculation (i.e., prevention setting) or after bone
metastases are established (i.e., treatment setting).
Several models of multiple myeloma have been estab-
lished and have consistently shown reductions in tumor-
induced osteolysis and skeletal tumor burden by N-BPs [52,
53, 59]. For example, in the 5T2 model of murine myeloma,
treatment with zoledronic acid (120 g/kg s.c. twice weekly)
after osteolytic lesions were established significantly
reduced development of new osteolytic lesions, reduced the
bone surface occupied by osteoclasts and inhibited tumor-
induced osteolysis, and reduced skeletal tumor burden as
evidenced by serum paraprotein levels [53]. Moreover,
treatment with zoledronic acid at the first sign of circulating
6 Bisphosphonates: Preclinical Review
15
10
5
0
A
p
o
p
t
o
s
i
s

(
%
)
Control DOX alone
50 nM, 24 hours
ZOL alone
25 M, 1 hour
DOX +
ZOL
DOX then
ZOL
1.5% 1.7%
13.6%
ZOL then
DOX
Figure 3. Synergistic apoptotic effect of zoledronic acid (ZOL) in
combination with doxorubicin (DOX) on MCF-7 breast cancer cells
in vitro. Adapted with permission from Neville-Webbe et al. [43].
paraprotein significantly prolonged disease-free survival in
those mice.
Numerous studies in breast cancer models have also
been reported. Zoledronic acid was shown to inhibit pro-
gression of established bone metastases and development of
new bone metastases in two models of breast cancer [54,
55]. In nude mice injected with human MDA-MB-231
breast cancer cells and allowed to develop osteolytic lesions,
mice treated with zoledronic acid (0.2, 1.0, or 5.0 g/day
s.c.) had significantly less radiographic bone lesion area, by
>80%, than controls [54]. Ibandronate (1.0 g/day) and
alendronate (10 g/day) resulted in nonsignificant reduc-
tions in bone lesion area (65% and 55% reductions, respec-
tively). These findings were confirmed in another
independent study using highly sensitive in vivo imaging of
MDA-MB-231 cells genetically engineered to express green
fluorescent protein [55]. Similar studies have been con-
ducted with ibandronate in nude mice bearing MDA-MB-
231 breast cancer cells, which typically form both adrenal
and bone metastases. Treatment with ibandronate (4 g/day
s.c.) inhibited the radiographic progression of established
osteolytic lesions and decreased skeletal tumor burden com-
pared with controls [9]. However, administration of iban-
dronate at the time of tumor cell inoculation resulted in a
twofold increase in adrenal tumor load [61]. In a murine
breast cancer model, treatment with zoledronic acid
(5 g/day) for 7 days after injection of 4T1 murine mam-
mary tumor cells (i.e., prevention setting) markedly
decreased the formation of new bone metastases at day 28
[54]. The observed decrease in radiographic bone lesion
area was accompanied by an increase in the number of apop-
totic osteoclasts and apoptotic tumor cells in bone lesions.
Notably, the 4T1 mammary tumor model has provided
the first in vivo evidence of synergy between zoledronic acid
and chemotherapy. In animals with established orthotopic
tumors, the combination of zoledronic acid (250 g/kg) with
20 mg/kg/day of oral UFT (a combination of uracil and tega-
fur [4:1 molar ratio]) reduced skeletal tumor burden more
effectively than either agent alone [56]. Similarly, the com-
bination of ibandronate with doxorubicin (150 g/day) has
been investigated in the MDA-MB-231 model and was
shown to have additive antitumor effects in bone [61].
Studies in a prostate cancer model have also recently been
reported. In those studies PC-3 and LuCaP 23.1 cells were
injected directly into the tibia of mice [40]; PC-3 cells form
osteolytic lesions, and LuCaP 23.1 cells form osteoblastic
lesions. The treatment group received zoledronic acid (5 g
s.c. twice weekly) either at the time of tumor cell injection or
after tibial tumors were established (7 days for PC-3 tumors
and 33 days for LuCaP 23.1 tumors). Treatment with zole-
dronic acid significantly inhibited growth of both osteolytic
and osteoblastic metastases by radiographic analysis (Fig. 4)
[40] and also reduced skeletal tumor burden as evidenced by
a significant decrease in serum levels of prostate-specific
antigen in animals bearing LuCaP 23.1 tumors [40]. The
observed reduction in serum prostate-specific antigen levels
provides compelling direct evidence of the antitumor activity
of zoledronic acid in this animal model.
The potential of zoledronic acid to prevent bone metas-
tasis was also demonstrated in an animal model of prostate
cancer [62]. In that model, mice were injected with PC-3
cells, and the incidence of bone metastases was studied in
normal mice and in mice that were rendered androgen defi-
cient by surgical castration. Androgen-deficient mice devel-
Green 7
Table 2. Animal models demonstrating antitumor effects of zoledronic acid
Study Tumor cells Bisphosphonate Dose and schedule
Yaccoby et al. [52] Primary human myeloma cells Zoledronic acid 100 g/kg/week s.c.
Pamidronate 1.3 mg/kg every 2 weeks s.c.
Croucher et al. [53] 5T2 murine myeloma Zoledronic acid 120 g/kg twice weekly s.c.
Green et al. [54] MDA-MB-231 human breast cancer Zoledronic acid 0.2-5.0 g/day s.c.
Ibandronate 1.0 g/day s.c.
Alendronate 10 g/day s.c.
Peyruchaud et al. [55] MDA-MB-231 human breast cancer Zoledronic acid 3 g/day 12 days s.c.
Hiraga et al. [56] 4T1 murine mammary carcinoma Zoledronic acid + 250 g/kg
UFT 20 mg/kg/day
Hiraga et al. [9] MDA-MB-231 human breast cancer Ibandronate 4 g/day s.c.
Nobuyuki et al. [57] 4T1 murine mammary carcinoma Zoledronic acid 0.5 or 5 g every 4 days i.v.
Corey et al. [40] PC-3 and LuCaP 23.1 prostate cancer Zoledronic acid 5 g/mouse twice weekly s.c.
Adapted with permission from Green [58].
oped significantly more bone metastases than did intact con-
trol mice. This strongly suggests that excess bone resorption
caused by androgen ablation can stimulate metastasis of PC-
3 cells to the bone. This is consistent with current hypotheses
of tumor cell metastasis to bone. Moreover, daily treatment
with zoledronic acid significantly reduced the incidence of
bone metastases in both normal and castrated mice.
Although the majority of animal models suggest that the
primary antitumor effect of bisphosphonates is manifested in
the bone, where they reach the highest concentrations, pre-
liminary data from the 4T1 mammary tumor model have
demonstrated that zoledronic acid can also inhibit visceral
metastases [57]. 4T1 cells expressing luciferase were used to
quantitate tumor burden in mice. After tumor cell inocula-
tion, mice were treated with zoledronic acid (5.0 g every
4 days), which resulted in lower tumor burdens than in con-
trols not only in bone but also in the liver and lungs [57, 63].
Treatment with zoledronic acid also prolonged survival of
tumor-bearing mice. In the same model, ibandronate
(4 g/day s.c.) had no effect on lung or liver metastases or on
survival [61, 63]. As mentioned above, treatment of mice
bearing 5T2 myeloma cells with zoledronic acid (120 g/kg
s.c. twice weekly) also resulted in a significantly longer dis-
ease-free survival time (median, 47 days versus 35 days for
untreated controls; p < 0.01) [53].
These animal models provide convincing evidence of the
potential of bisphosphonates, particularly the more potent
N-BPs, to reduce tumor burden in bone and inhibit formation
and progression of bone metastases in a variety of tumor mod-
els. It is less clear whether bisphosphonates have antitumor
activity beyond the bone, but it appears that zoledronic acid
may be sufficiently potent to inhibit extraskeletal tumor cell
growth and to prolong survival in some animal models.
MECHANISMS OF ANTITUMOR EFFECTS
The precise mechanisms responsible for the antitumor
effects of bisphosphonates are beginning to be elucidated.
Bisphosphonates appear to make the bone a less favorable
site for tumor cell growth via the inhibition of osteoclast-
mediated bone resorption and osteoclastogenesis, thereby
reducing the release of growth factors that stimulate tumor
growth in bone. In addition, bisphosphonates directly
inhibit tumor cell growth and survival and the ability of
tumor cells to colonize the bone. Either or both of these
mechanisms may be at work.
Apoptosis
One of the primary mechanisms responsible for the
direct antitumor activity of bisphosphonates is induction
of tumor cell apoptosis. Both N-BPs and non-N-BPs
appear to induce apoptosis of osteoclasts and tumor cells
by activation of caspases [9, 12-14, 16, 17, 64, 65]. One
mechanism by which bisphosphonates induce apoptosis
is through production of ATP analogues (either as direct
metabolites or as a result of inhibition of the mevalonate
pathway), which can disrupt mitochondrial ATP/ADP
translocase. A recent study investigating the mechanism
by which zoledronic acid induced apoptosis in human
breast cancer cell lines (MDA-MB-231 and MCF-7)
indicated that this response was associated with
cytochrome c release from the mitochondria and subse-
quent caspase-3 activation [66]. It appears that N-BPs
may induce cytochrome c release by modulating expres-
sion of Bcl-2, a key antiapoptotic regulatory protein
[66]. These events may be precipitated by inhibition of
Ras activation, which requires protein prenylation
(specifically farnesylation) [66].
8 Bisphosphonates: Preclinical Review
Figure 4. The effects of
zoledronic acid (5 g s.c.
twice weekly) on tumor
volume (assessed radi-
ographically) in mice with
tibial tumors from PC-3
and LuCaP 23.1 prostate
cancer cell lines. Zole-
dronic acid was adminis-
tered either at the time of
tumor cell injection (i.e.,
prevention) or after tibial
tumors were established
(i.e., treatment), which
was 7 days postinjection
for PC-3 and 33 days
postinjection for LuCaP.
*p < 0.03; **p < 0.001.
Adapted with permission
from Corey et al. [40].
80
60
40
20
0
T
u
m
o
r

v
o
l
u
m
e

(
%

t
i
s
s
u
e

v
o
l
u
m
e
)
Vehicle Prevent Treat Vehicle Prevent Treat
Zoledronic acid: 5 g/mouse (twice a week)
*
*
**
**
PC-3 (lytic) LuCaP 23.1 (blastic)
Inhibition of Tumor Cell Adhesion and Invasion of the
Extracellular Bone Matrix
Bisphosphonates have also been shown to inhibit adhe-
sion of tumor cells to extracellular matrix (ECM) proteins and
to inhibit the process of tumor cell invasion and metastasis
[67-70]. Using an in vitro Matrigel

-based invasion assay,


Boissier et al. have shown that bisphosphonates inhibit the
ability of human breast and prostate cancer cells to invade the
ECM [67]. In this assay, zoledronic acid and ibandronate
caused dose-dependent inhibition of cell invasion through the
Matrigel

at extremely low concentrations (10


10
M) that did
not inhibit tumor cell motility or induce significant apoptosis.
Clodronate was approximately six orders of magnitude less
potent in the same assay. Furthermore, the combination of
ibandronate with taxanes enhanced the inhibitory effect on
MDA-MB-231 cell invasion [69].
One contributory mechanism may be the inhibition of
matrix metalloproteinase (MMP) activity, which is necessary
for tumor cell invasion of the ECM [67, 71, 72]. Bisphos-
phonates have been shown to inhibit the activity of MMPs
produced by tumor cell lines, and this seems to correlate with
reduced invasiveness in the Matrigel

assay [71, 72]. For


example, zoledronic acid was shown to inhibit the production
of MMP-2 and MMP-9 by PC-3 cells [40]. These data sug-
gest a potential mechanism by which N-BPs may inhibit tumor
cell invasion of the bone but cannot explain the apparent
dependence of this effect on protein prenylation.
Recent studies suggest that inhibition of tumor cell
adhesion to ECM proteins and invasion through Matrigel

is dependent on inhibition of protein prenylation. In one


study, zoledronic acid was shown to dose-dependently
inhibit adhesion of MCF-7 and MDA-MB-231 cells to a
variety of matrix proteins (Fig. 5) [73], and this inhibitory
effect was overcome by addition of either farnesol or ger-
anylgeraniol or by the addition of a broad spectrum caspase
inhibitor [73]. Similar findings have been reported for alen-
dronate. The inhibitory effect of alendronate on tumor cell
invasion through Matrigel

was reversed by the addition


of geranylgeraniol and
trans-trans-farnesol
[74]. Therefore, inhibition of the mevalonate pathway and
induction of caspase activity are important for the
inhibitory effects of N-BPs on tumor cell adhesion to the
ECM and on invasiveness. Further, it has been shown that
an activating Ras mutation enhances adhesion of a normal
breast epithelial cell line to ECM proteins, suggesting that
increased Ras activation in response to growth factor recep-
tor signaling may increase the metastatic potential of breast
cancer cells [73]. Thus, by inhibiting protein prenylation
and Ras signaling, zoledronic acid should reduce the
metastatic potential of tumor cells.
Antiangiogenic Effects
In vitro and in vivo studies have further demonstrated that
zoledronic acid has antiangiogenic effects. In vitro assays
with human umbilical vein endothelial cells (HUVECs) have
shown that zoledronic acid dose-dependently inhibited the
proliferation of HUVECs induced by fetal calf serum and
basic fibroblast growth factor (bFGF), and these findings have
been confirmed in vivo [75]. Systemic administration of zole-
dronic acid to mice resulted in potent inhibition of angiogen-
esis induced by s.c. implants impregnated with bFGF, with
a dose of 3 g/kg producing a 50% efficacy (ED
50
) [75]. It
has also been reported that zoledronic acid can reduce bone-
tumor-associated angiogenesis in the murine 5T2 myeloma
model [53]. In another series of experiments, zoledronic
acid, as well as ibandronate, risedronate, and clodronate,
inhibited formation of capillary-like tubules by HUVECs in
vitro [76]. In vivo, zoledronic acid and ibandronate, but not
clodronate, decreased revascularization (as measured by
vessel area) of the ventral prostate gland in castrated rats
treated with testosterone [76].
The inhibitory effect of zoledronic acid on endothelial
cell adhesion and migration appears to be mediated, at least
in part, by modulation of integrins (e.g.,
v

3
and
v

5
) that
are involved in angiogenesis [77, 78]. Interestingly,
v

3
integrin is also required for osteoclasts to adhere tightly to
the bone and form resorption lacunae during active bone
Green 9
Figure 5. Percent adherent
cells versus controls when
MDA-MB-231 human
breast cancer cells were
incubated with zoledronic
acid for 24 hours (0.1 or
100 M) before culture
on plates coated with vari-
ous extracellular matrix
proteins. Adapted with
permission from Pickering
et al. [73].
Control
0.1 M Zoledronic acid
100 M Zoledronic acid
24-hour treatment
Collagen I Vitronectin Fibronectin Laminin
A
d
h
e
r
e
n
t

c
e
l
l
s

(
%

c
o
n
t
r
o
l
)
100
50
0
resorption, and
v

3
expression confers on tumor cells a
greater propensity to metastasize to bone [79]. In fact, a
small molecule inhibitor of
v

3
was recently shown to
effectively prevent metastasis of MDA-MB-435 breast can-
cer cells to bone [80]. Therefore, effects on
v

3
could have
pleiotropic effects on bone resorption and tumor metastasis.
In addition, it has recently been reported that zoledronic
acid decreases the survival of HUVECs by sensitizing them
to TNF-induced programmed cell death [78]. Zoledronic
acid also appears to modulate serum levels of proangio-
genic growth factors such as vascular endothelial growth
factor and bFGF in cancer patients [81]. These studies sug-
gest a variety of potential mechanisms to account for the
observed antiangiogenic effects of bisphosphonates.
CONCLUSIONS AND FUTURE DIRECTIONS
There is now extensive in vitro and in vivo preclinical
evidence that bisphosphonates, particularly the more
potent N-BPs, have antitumor activity and can reduce
skeletal tumor burden. The evidence that they have antitu-
mor activity outside the bone is more tenuous. A variety
of potential mechanisms to explain these observed antitu-
mor effects have been proposed, including indirect effects
on tumor cell growth in bone via inhibition of bone
resorption and osteoclastogenesis. In addition, bisphos-
phonates clearly have the potential to directly induce
apoptosis of tumor cells, inhibit tumor cell adhesion to the
ECM, reduce the metastatic potential of tumor cells, and
inhibit angiogenesis. Further research is ongoing to fully
elucidate the molecular mechanisms involved and to
determine the most effective dose and schedule of bispho-
sphonates to maximize their antitumor potential in the clin-
ical setting, either alone or in combination with standard
antineoplastic agents.
ACKNOWLEDGMENT
Jonathan Green is a full time employee of Novartis
Pharma AG and holds stock in the company.
10 Bisphosphonates: Preclinical Review
REFERENCES
1 Fleisch H. Development of bisphosphonates. Breast Cancer
Res 2002;4:30-34.
2 Russell RG, Croucher PI, Rogers MJ. Bisphosphonates: phar-
macology, mechanisms of action and clinical uses. Osteoporos
Int 1999;9(suppl 2):S66-S80.
3 Stronski SA, Bettschen-Camin L, Wetterwald A et al. Bisphos-
phonates inhibit 1,25-dihydroxyvitamin D3-induced increase of
osteocalcin in plasma of rats in vivo and in culture medium of rat
calvaria in vitro. Calcif Tissue Int 1988;42:248-254.
4 Flanagan AM, Chambers TJ. Inhibition of bone resorption by
bisphosphonates: interactions between bisphosphonates,
osteoclasts, and bone. Calcif Tissue Int 1991;49:407-415.
5 Rogers MJ, Gordon S, Benford HL et al. Cellular and molec-
ular mechanisms of action of bisphosphonates. Cancer
2000;88(suppl 12):2961-2978.
6 Sato M, Grasser W, Endo N et al. Bisphosphonate action.
Alendronate localization in rat bone and effects on osteoclast
ultrastructure. J Clin Invest 1991;88:2095-2105.
7 Aparicio A, Gardner A, Tu Y et al. In vitro cytoreductive
effects on multiple myeloma cells induced by bisphosphonates.
Leukemia 1998;12:220-229.
8 Derenne S, Amiot M, Barille S et al. Zoledronate is a potent
inhibitor of myeloma cell growth and secretion of IL-6 and
MMP-1 by the tumoral environment. J Bone Miner Res
1999;14:2048-2056.
9 Hiraga T, Williams PJ, Mundy GR et al. The bisphosphonate
ibandronate promotes apoptosis in MDA-MB-231 human breast
cancer cells in bone metastases. Cancer Res 2001;61:4418-4424.
10 Lee MV, Fong EM, Singer FR et al. Bisphosphonate treat-
ment inhibits the growth of prostate cancer cells. Cancer Res
2001;61:2602-2608.
11 Mackie PS, Fisher JL, Zhou H et al. Bisphosphonates regulate
cell growth and gene expression in the UMR 106-01 clonal rat
osteosarcoma cell line. Br J Cancer 2001;84:951-958.
12 Senaratne SG, Pirianov G, Mansi JL et al. Bisphosphonates
induce apoptosis in human breast cancer cell lines. Br J Cancer
2000;82:1459-1468.
13 Shipman CM, Rogers MJ, Apperley JF et al. Bisphosphonates
induce apoptosis in human myeloma cell lines: a novel anti-
tumour activity. Br J Haematol 1997;98:665-672.
14 Shipman CM, Croucher PI, Russell RG et al. The bisphos-
phonate incadronate (YM175) causes apoptosis of human
myeloma cells in vitro by inhibiting the mevalonate pathway.
Cancer Res 1998;58:5294-5297.
15 Sonnemann J, Eckervogt V, Truckenbrod B et al. The bisphos-
phonate pamidronate is a potent inhibitor of human osteosar-
coma cell growth in vitro. Anticancer Drugs 2001;12:459-465.
16 Takahashi R, Shimazaki C, Inaba T et al. A newly developed
bisphosphonate, YM529, is a potent apoptosis inducer of
human myeloma cells. Leuk Res 2001;25:77-83.
17 Tassone P, Tagliaferri P, Viscomi C et al. Zoledronic acid
induces antiproliferative and apoptotic effects in human pan-
creatic cancer cells in vitro. Br J Cancer 2003;88:1971-1978.
18 Rogers MJ, Russell RG, Blackburn GM et al. Metabolism of
halogenated bisphosphonates by the cellular slime mould
Dictyostelium discoideum. Biochem Biophys Res Commun
1992;189:414-423.
19 Rogers MJ, Ji X, Russell RG et al. Incorporation of bisphospho-
nates into adenine nucleotides by amoebae of the cellular slime
mould Dictyostelium discoideum. Biochem J 1994;303:303-311.
20 Pelorgeas S, Martin JB, Satre M. Cytotoxicity of dichloro-
methane diphosphonate and of 1-hydroxyethane-1,1-diphos-
phonate in the amoebae of the slime mould Dictyostelium
discoideum. A 31P NMR study. Biochem Pharmacol
1992;44:2157-2163.
21 Frith JC, Mnkknen J, Blackburn GM et al. Clodronate and
liposome-encapsulated clodronate are metabolized to a toxic
ATP analog, adenosine 5-(beta, gamma-dichloromethylene)
triphosphate, by mammalian cells in vitro. J Bone Miner Res
1997;12:1358-1367.
22 Lehenkari PP, Kellinsalmi M, Napankangas JP et al. Further
insight into mechanism of action of clodronate: inhibition of
mitochondrial ADP/ATP translocase by a nonhydrolyzable, ade-
nine-containing metabolite. Mol Pharmacol 2002;61:1255-1262.
23 Rogers MJ, Watts DJ, Russell RGG. Overview of bisphos-
phonates. Cancer 1997;80(suppl 8):1652-1660.
24 Selander KS, Monkkonen J, Karhukorpi EK et al. Character-
istics of clodronate-induced apoptosis in osteoclasts and
macrophages. Mol Pharmacol 1996;50:1127-1138.
25 Gober HJ, Kistowska M, Angman L et al. Human T cell
receptor gammadelta cells recognize endogenous mevalonate
metabolites in tumor cells. J Exp Med 2003;197:163-168.
26 Luckman SP, Hughes DE, Coxon FP et al. Nitrogen-contain-
ing bisphosphonates inhibit the mevalonate pathway and pre-
vent post-translational prenylation of GTP-binding proteins,
including Ras. J Bone Miner Res 1998;13:581-589.
27 Rogers MJ, Frith JC, Luckman SP et al. Molecular mecha-
nisms of action of bisphosphonates. Bone 1999;24(suppl
5):73S-79S.
28 Russell RG, Rogers MJ, Frith JC et al. The pharmacology of
bisphosphonates and new insights into their mechanisms of
action. J Bone Miner Res 1999;14(suppl 2):53-65.
29 Hughes DE, Wright KR, Uy HL et al. Bisphosphonates promote
apoptosis in murine osteoclasts in vitro and in vivo. J Bone
Miner Res 1995;10:1478-1487.
30 Luckman SP, Coxon FP, Ebetino FH et al. Heterocycle-con-
taining bisphosphonates cause apoptosis and inhibit bone
resorption by preventing protein prenylation: evidence from
structure-activity relationships in J774 macrophages. J Bone
Miner Res 1998;13:1668-1678.
31 Oliff A. Farnesyltransferase inhibitors: targeting the molecular
basis of cancer. Biochim Biophys Acta 1999;1423:C19-C30.
32 Zhang FL, Casey PJ. Protein prenylation: molecular mecha-
nisms and functional consequences. Annu Rev Biochem
1996;65:241-269.
33 Alakangas A, Selander K, Mulari M et al. Alendronate dis-
turbs vesicular trafficking in osteoclasts. Calcif Tissue Int
2002;70:40-47.
34 Mnkknen H, Lehenkari PP, Kellinsalmi M et al. A new
mechanism of action for bisphosphonates: apppi dedicated
cytotoxicity of N-BPs. Bone 2004;34:S66-S67.
35 Dunford JE, Thompson K, Coxon FP et al. Structure-activity rela-
tionships for inhibition of farnesyl diphosphate synthase in vitro
and inhibition of bone resorption in vivo by nitrogen-containing
bisphosphonates. J Pharmacol Exp Ther 2001;296:235-242.
36 Green JR, Mller K, Jaeggi KA. Preclinical pharmacology of
CGP 42'446, a new, potent, heterocyclic bisphosphonate
compound. J Bone Miner Res 1994;9:745-751.
37 Fisher JE, Rodan GA, Reszka AA. In vivo effects of bisphos-
phonates on the osteoclast mevalonate pathway. Endocrinology
2000;141:4793-4796.
38 Clezardin P. The antitumor potential of bisphosphonates.
Semin Oncol 2002;29(suppl 21):33-42.
39 Jagdev SP, Coleman RE, Shipman CM et al. The bisphos-
phonate, zoledronic acid, induces apoptosis of breast cancer
cells: evidence for synergy with paclitaxel. Br J Cancer
2001;84:1126-1134.
40 Corey E, Brown LG, Quinn JE et al. Zoledronic acid exhibits
inhibitory effects on osteoblastic and osteolytic metastases of
prostate cancer. Clin Cancer Res 2003;9:295-306. Erratum in:
Clin Cancer Res 2003;9:1574-1575.
41 Oades GM, Senaratne SG, Clarke IA et al. Nitrogen contain-
ing bisphosphonates induce apoptosis and inhibit the meval-
onate pathway, impairing Ras membrane localization in
prostate cancer cells. J Urol 2003;170:246-252.
42 Matsumoto S, Kimura S, Segawa H et al. Efficacy of com-
bining the third generation bisphosphonate, zoledronate with
imatinib mesylate in suppressing small cell lung cancer cell
line proliferation. Proc Am Soc Clin Oncol 2003;22:684.
43 Neville-Webbe HL, Evans CA, Coleman RE et al. Zoledronic
acid (ZOL) in combination with doxorubicin (DOX) has syn-
ergistic effects on apoptosis of breast cancer cells (BCCs) in
vitro [abstract]. Presented at the IVth International Conference
on Cancer-Induced Bone Diseases, December 7-9, 2003, San
Antonio, TX.
44 Tassone P, Forciniti S, Galea E et al. Growth inhibition and syn-
ergistic induction of apoptosis by zoledronate and dexametha-
sone in human myeloma cell lines. Leukemia 2000;14:841-844.
45 Ulln A, Lennartsson L, Hjelm-Eriksson M et al. Additive/syn-
ergistic anti-tumoral effects on prostate cancer cells in vitro fol-
lowing treatment with a combination of docetaxel and
zoledronate [poster]. Presented at the European Winter
Oncology Conference (EWOC-8), January 19-24, 2003, Flims,
Switzerland.
46 Ulln A, Lennartsson L, Hjelm-Eriksson M et al. Additive/syn-
ergistic anti-tumoral effect on prostate cancer cells in vitro fol-
lowing treatment with a combination of gemcitabine and
zoledronic acid. Proc Am Soc Clin Oncol 2003;22:432.
47 Vogt U, Wassmann K, Bosse U et al. Enhancement of breast
tumor growth inhibition (BTGI) in vitro through combination of
CMF, epirubicin/cyclophosphamide (EC), epirubicin/paclitaxel
(ET) and epirubicin/docetaxel (EDoc) with ibandronate (IB) or
zoledronic acid (ZOL). Proc Am Soc Clin Oncol 2003;22:55.
48 Witters L, Crispino J, Javeed M et al. Inhibition of growth of
a human prostate cancer cell line with the combination of
zoledronic acid and a COX-2 inhibitor. Proc Am Soc Clin
Oncol 2002;21:5b.
Green 11
49 Witters LM, Crispino J, Fraterrigo T et al. Effect of the com-
bination of docetaxel, zoledronic acid, and a COX-2 inhibitor
on the growth of human breast cancer cell lines. Am J Clin
Oncol 2003;26:S92-S97.
50 Schlotter CM, Vogt U, Bosse U et al. Enhancement of breast
tumor growth inhibition (BTGI) in vitro through combination
of CMF, epirubicin/cyclophosphamide (EC), and epiru-
bicin/paclitaxel (ET) with ibandronate (IB) or zoledronic acid
(ZOL). J Bone Miner Res 2001;16(suppl 1):S191.
51 Kuroda J, Kimura S, Segawa H et al. The third-generation
bisphosphonate zoledronate synergistically augments the
anti-Ph
+
leukemia activity of imatinib mesylate. Blood
2003;102:2229-2235.
52 Yaccoby S, Pearse RN, Johnson CL et al. Myeloma interacts
with the bone marrow microenvironment to induce osteoclasto-
genesis and is dependent on osteoclast activity. Br J Haematol
2002;116:278-290.
53 Croucher PI, De Raeve H, Perry MJ et al. Zoledronic acid treat-
ment of 5T2MM-bearing mice inhibits the development of
myeloma bone disease: evidence for decreased osteolysis,
tumor burden and angiogenesis, and increased survival. J Bone
Miner Res 2003;18:482-492.
54 Green J, Gschaidmeier H, Yoneda T et al. Zoledronic acid
potently inhibits tumour-induced osteolysis in two models of
breast cancer metastasis to bone. Ann Oncol 2000;11(suppl 4):14.
55 Peyruchaud O, Winding B, Pecheur I et al. Early detection of
bone metastases in a murine model using fluorescent human
breast cancer cells: application to the use of the bisphosphonate
zoledronic acid in the treatment of osteolytic lesions. J Bone
Miner Res 2001;16:2027-2034.
56 Hiraga T, Ueda A, Tamura D et al. Effects of oral UFT com-
bined with or without zoledronic acid on bone metastasis in the
4T1/luc mouse breast cancer. Int J Cancer 2003;106:973-979.
57 Nobuyuki H, Hiraga T, Williams PJ et al. The bisphospho-
nate zoledronic acid inhibits metastases to bone and liver with
suppression of osteopontin production in mouse mammary
tumor. J Bone Miner Res 2001;16(suppl 1):S191.
58 Green JR. Bisphosphonates in cancer therapy. Curr Opin Oncol
2002;14:609-615.
59 Cruz JC, Alsina M, Craig F et al. Ibandronate decreases bone
disease development and osteoclast stimulatory activity in an in
vivo model of human myeloma. Exp Hematol 2001;29:441-447.
60 Alvarez E, Westmore M, Galvin RJ et al. Properties of bisphos-
phonates in the 13762 rat mammary carcinoma model of
tumor-induced bone resorption. Clin Cancer Res 2003;9:5705-
5713.
61 Michigami T, Hiraga T, Williams PJ et al. The effect of the
bisphosphonate ibandronate on breast cancer metastasis to
visceral organs. Breast Cancer Res Treat 2002;75:249-258.
62 Padalecki SS, Carreon M, Grubbs B et al. Androgen deprivation
enhances bone loss and prostate cancer metastases to bone:
prevention by zoledronic acid. Oncology 2003;17(suppl):32.
63 Yoneda T, Hashimoto N, Hiraga T. Bisphosphonate actions
on cancer. Calcif Tissue Int 2003;73:315-318.
64 Benford HL, McGowan NWA, Helfrich MH et al.
Visualization of bisphosphonate-induced caspase-3 activity
in apoptotic osteoclasts in vitro. Bone 2001;28:465-473.
65 Fromigue O, Lagneaux L, Body JJ. Bisphosphonates induce
breast cancer cell death in vitro. J Bone Miner Res
2000;15:2211-2221.
66 Senaratne SG, Mansi JL, Colston KW. The bisphosphonate
zoledronic acid impairs Ras membrane [correction of impairs
membrane] localisation and induces cytochrome c release in
breast cancer cells. Br J Cancer 2002;86:1479-1486. Erratum
in: Br J Cancer 2002;87:1340.
67 Boissier S, Ferreras M, Peyruchaud O et al. Bisphosphonates
inhibit breast and prostate carcinoma cell invasion, an early
event in the formation of bone metastases. Cancer Res
2000;60:2949-2954.
68 Boissier S, Magnetto S, Frappart L et al. Bisphosphonates
inhibit prostate and breast carcinoma cell adhesion to unmin-
eralized and mineralized bone extracellular matrices. Cancer
Res 1997;57:3890-3894.
69 Magnetto S, Boissier S, Delmas PD et al. Additive antitumor
activities of taxoids in combination with the bisphosphonate
ibandronate against invasion and adhesion of human breast
carcinoma cells to bone. Int J Cancer 1999;83:263-269.
70 van der Pluijm G, Vloedgraven H, van Beek E et al.
Bisphosphonates inhibit the adhesion of breast cancer cells to
bone matrices in vitro. J Clin Invest 1996;98:698-705.
71 Teronen O, Heikkila P, Konttinen YT et al. MMP inhibition
and downregulation by bisphosphonates. Ann NY Acad Sci
1999;878:453-465.
72 Heikkila P, Teronen O, Moilanen M et al. Bisphosphonates
inhibit stromelysin-1 (MMP-3), matrix metalloelastase
(MMP-12), collagenase-3 (MMP-13) and enamelysin (MMP-
20), but not urokinase-type plasminogen activator, and dimin-
ish invasion and migration of human malignant and
endothelial cell lines. Anticancer Drugs 2002;13:245-254.
73 Pickering LM, Mansi JL, Colston KW. Adhesion of breast
cancer cells to extracellular matrices is inhibited by zole-
dronic acid and enhanced by aberrant Ras signalling. Proc
Am Soc Clin Oncol 2003;22:863.
74 Virtanen SS, Vnnen HK, Hrknen PL et al. Alendronate
inhibits invasion of PC-3 prostate cancer cells by affecting
the mevalonate pathway. Cancer Res 2002;62:2708-2714.
75 Wood J, Bonjean K, Ruetz S et al. Novel antiangiogenic
effects of the bisphosphonate compound zoledronic acid.
J Pharmacol Exp Ther 2002;302:1055-1061.
76 Fournier P, Boissier S, Filleur S et al. Bisphosphonates
inhibit angiogenesis in vitro and testosterone-stimulated vas-
cular regrowth in the ventral prostate in castrated rats. Cancer
Res 2002;62:6538-6544.
77 Bonjean K, Bellahcene A, Locigno R et al. Zoledronate modu-
lates endothelial cell surface receptors involved in angiogenesis.
Proc Am Assoc Cancer Res 2001;42:106.
78 Bezzi M, Hasmim M, Bieler G et al. Zoledronate sensitizes
endothelial cells to tumor necrosis factor-induced programmed
12 Bisphosphonates: Preclinical Review
cell death. Evidence for the suppression of sustained activa-
tion of focal adhesion kinase and protein kinase B/Akt. J Biol
Chem 2003;278:43603-43614.
79 Pcheur I, Peyruchaud O, Serre CM et al. Integrin
alpha(v)beta3 expression confers on tumor cells a greater
propensity to metastasize to bone. FASEB J 2002;16:1266-
1268.
80 Welch DR, Harms JF, Samant RS et al. The small molecule

3
antagonist (S247) inhibits MDA-MB-435 breast cancer
metastasis to bone. Oncology 2003;17(suppl):18.
81 Santini D, Vincenzi B, Dicuonzo G et al. Zoledronic acid
induces significant and long-lasting modifications of circulat-
ing angiogenic factors in cancer patients. Clin Cancer Res
2003;9:2893-2897.
Green 13

Das könnte Ihnen auch gefallen