Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

DOI:10.1158/0008-5472.

CAN-07-5725

Aplidin, a Marine Organism−Derived Compound with Potent


Antimyeloma Activity In vitro and In vivo
Constantine S. Mitsiades, Enrique M. Ocio, Atanasio Pandiella, et al.

Cancer Res 2008;68:5216-5225. Published online July 1, 2008.

Updated Version Access the most recent version of this article at:
doi:10.1158/0008-5472.CAN-07-5725
Supplementary Access the most recent supplemental material at:
Material http://cancerres.aacrjournals.org/content/suppl/2008/06/30/68.13.5216.DC1.html

Cited Articles This article cites 40 articles, 25 of which you can access for free at:
http://cancerres.aacrjournals.org/content/68/13/5216.full.html#ref-list-1
Citing Articles This article has been cited by 1 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/68/13/5216.full.html#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR
Subscriptions Publications Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, contact the AACR Publications
Department at permissions@aacr.org.

Downloaded from cancerres.aacrjournals.org on April 12, 2011


Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725

Research Article

Aplidin, a Marine Organism–Derived Compound with Potent


Antimyeloma Activity In vitro and In vivo
1,2 3,4 3 3
Constantine S. Mitsiades, Enrique M. Ocio, Atanasio Pandiella, Patricia Maiso,
3,5 3 3 3
Consuelo Gajate, Mercedes Garayoa, David Vilanova, Juan Carlos Montero,
1,2 1,2 1,2 1,2
Nicholas Mitsiades, Ciaran J. McMullan, Nikhil C. Munshi, Teru Hideshima,
1,2 6,7 6,7 6,7
Dharminder Chauhan, Pablo Aviles, Gabriel Otero, Glynn Faircloth,
4 1,2 3
M. Victoria Mateos, Paul G. Richardson, Faustino Mollinedo,
3,4 1,2
Jesus F. San-Miguel, and Kenneth C. Anderson
1
Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute; 2Department of Medicine,
Harvard Medical School, Boston, Massachusetts; 3Centro de Investigación del Cáncer, Instituto de Biologı́a Molecular y Celular del
Cáncer, Universidad de Salamanca-CSIC, Salamanca; 4Departamento de Hematologı́a and 5Unidad de investigación,
Hospital Universitario de Salamanca, Spain; 6PharmaMar, Madrid, Spain; and 7PharmaMar, Inc., Cambridge, Massachusetts

Abstract malignancy in the Western world (2). The identification of new


Despite recent progress in its treatment, multiple myeloma classes of agents with anti-MM activity, especially in patients who
(MM) remains incurable, thus necessitating identification of relapse from or do not respond to available therapies, remains
novel anti-MM agents. We report that the marine-derived an urgent priority. Within this context, we evaluated a series of
cyclodepsipeptide Aplidin exhibits, at clinically achievable candidate antitumor agents derived from marine organisms. This
concentrations, potent in vitro activity against primary MM effort was based on the rationale that several established or
tumor cells and a broad spectrum of human MM cell lines, investigational anticancer drugs are derived from natural products
including cells resistant to conventional (e.g., dexamethasone, (3). These compounds are produced by various plants and other
alkylating agents, and anthracyclines) or novel (e.g., thalido- organisms (mostly above sea level), and their antimitotic pro-
mide and bortezomib) anti-MM agents. Aplidin is active perties, which prompted the evaluation of their antitumor activity,
against MM cells in the presence of proliferative/antiapoptotic may serve the teleologic purpose of protecting from predators
cytokines or bone marrow stromal cells and has additive or or infections. Recently, increasing interest has been placed on
synergistic effects with some of the established anti-MM oceanic organisms, primarily invertebrates (e.g., sponges, tunicates,
agents. Mechanistically, a short in vitro exposure to Aplidin bryozoans, and mollusks), as unique natural sources of a diverse
induces MM cell death, which involves activation of p38 and array of natural products with potent antimitotic and/or antitumor
c-jun NH2-terminal kinase signaling, Fas/CD95 translocation properties, including compounds that have advanced to late-
to lipid rafts, and caspase activation. The anti-MM effect of stage clinical trials (4, 5). An intriguing hypothesis about marine
Aplidin is associated with suppression of a constellation of organism–derived compounds is that they may have evolved to be
proliferative/antiapoptotic genes (e.g., MYC, MYBL2, BUB1, more potent than similar compounds from above-water organisms
MCM2, MCM4, MCM5, and survivin) and up-regulation of to compensate for the increased diffusion and thus rapidly
several potential regulators of apoptosis (including c-JUN, decreasing protective concentration gradient of these compounds
TRAIL, CASP9, and Smac). Aplidin exhibited in vivo anti-MM under water.
activity in a mouse xenograft model. The profile of the anti- In this study, we characterize the anti-MM activity of one such
MM activity of Aplidin in our preclinical models provided marine-derived compound, Aplidin, which is a cyclic depsipeptide
the framework for its clinical testing in MM, which has already isolated from the Mediterranean tunicate Aplidium albicans (6). We
provided favorable preliminary results. [Cancer Res 2008; show that Aplidin, at clinically relevant concentrations, has a
68(13):5216–25] potent activity against a broad panel of human MM cell lines and
primary MM tumor cells, including cells resistant to conventional
Introduction or novel anti-MM agents. Although cytokine- or cell adhesion–
mediated interactions with the bone marrow microenvironment
Although recent advances in the therapeutic management of
protect MM cells from conventional therapies (e.g., dexamethasone
multiple myeloma (MM) have improved its prognosis (1), no
or cytotoxic chemotherapy; refs. 7, 8), Aplidin is able to overcome
curative therapy currently exists for this plasma cell dyscrasia,
these protective effects in coculture models of MM cells with bone
which is the second most commonly diagnosed hematologic
marrow stromal cells. Moreover, Aplidin has additive or synergistic
effects with established anti-MM agents including bortezomib and
immunomodulatory agents. Mechanistically, in vitro exposure of
Note: Supplementary data for this article are available at Cancer Research Online MM cells to clinically relevant doses of Aplidin for even a few hours
(http://cancerres.aacrjournals.org/). is sufficient to induce their cell death. This involves activation of
C.S. Mitsiades, E.M. Ocio, and A. Pandiella contributed equally to this work. p38 signaling and is associated with suppression of a constellation
Requests for reprints: Constantine S. Mitsiades, Jerome Lipper Multiple Myeloma
Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard of proliferative/antiapoptotic genes and up-regulation of several
Medical School, 44 Binney Street, Boston, MA 02115. Phone: 617-632-1962; Fax: 617- positive regulators of apoptosis. Aplidin also exhibited in vivo anti-
812-7701; E-mail: Constantine_Mitsiades@dfci.harvard.edu.
I2008 American Association for Cancer Research.
MM activity in a mouse xenograft model. The profile of the anti-
doi:10.1158/0008-5472.CAN-07-5725 MM activity of Aplidin in preclinical in vitro and in vivo models,

Cancer Res 2008; 68: (13). July 1, 2008 5216 www.aacrjournals.org


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Antimyeloma Activity of Aplidin

coupled with its favorable safety profile in clinical trials for solid Results
tumors, provided the framework for clinical studies of this agent in In vitro activity of Aplidin against MM cell lines. We first
MM, where Aplidin has already shown encouraging preliminary evaluated, using MTT assays, the in vitro activity of Aplidin against
results. a large panel of MM cell lines including cells resistant to various
anti-MM agents. We observed that Aplidin exhibited potent in vitro
anti-MM activity (Fig. 1): The majority (18 of 19) of MM cell lines
Materials and Methods
treated with Aplidin at 10 nmol/L, a clinically achievable con-
MM cell lines, primary MM tumor cells, stromal cells, normal centration (20), exhibited V30% viability compared with their
hematopoietic cells, reagents, and antibodies. Detailed information on respective controls (Fig. 1A; Supplementary Fig. S1; Supplementary
the MM cell lines, primary MM tumor cells, stromal cells, normal
Table S1). Aplidin-responsive cell lines included cells resistant to
hematopoietic cells, reagents, and antibodies is included in Supplementary
data.
conventional anti-MM agents [e.g., melphalan (RPMI-8226/LR5),
Evaluations of cell viability, cell proliferation, cell cycle profiles, mitoxantrone (RPMI-8226/MR20), and doxorubicin (RPMI-8226/
induction of apoptosis, and mitochondrial membrane potential. Dox40)] as well as cells with low sensitivity to novel anti-MM
Previously described protocols were used to evaluate MM cell viability agents [e.g., immunomodulatory thalidomide derivatives (S6B45,
[with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide RPMI-8226/S); Fig. 1A]. Similarly, on comparison of the in vitro
(MTT) colorimetric survival assay; refs. 9–13]; cell proliferation and cell effect of Aplidin on dexamethasone-sensitive MM1S and melpha-
cycle profiles (with the CD38-CD138/propidium iodide double-staining lan-sensitive U266 cells versus the effect of the drug on the MM1R
technique; ref. 14); induction of apoptosis (based on Annexin V-FITC/ and U266-LR7 isogenic sublines, which are resistant to dexameth-
propidium iodide staining of myelomatous plasma cells; ref. 13); and
asone and melphalan, respectively, we observed that Aplidin
changes in mitochondrial transmembrane potential (Dw m; using
3,3¶-dihexyloxacarbocyanine iodide [DiOC6(3)] staining of cells; ref. 13). potently suppressed MM cell viability in a dose-dependent manner
Detailed information on these protocols is included in Supplementary both in the drug-sensitive lines and in their drug-resistant sublines
data. (Fig. 1B). Consistent with the studies on other MM cell lines, the
Oligonucleotide microarray analyses, Western blots, c-jun NH2- IC50 values for these four cell lines were within the 1 to 10 nmol/L
terminal kinase in vitro kinase assays, and lipid raft isolation. Gene range (4.1, 4.7, 5, and 8.2 nmol/L for MM1S, MM1R, U266, and
expression profiling, Western blot analyses, c-jun NH2-terminal kinase (JNK) U266-LR7, respectively). Interestingly, this range of potency is
in vitro kinase assays, and lipid raft isolation studies were done as in similar to that observed for bortezomib (data not shown), a potent
previous studies (12, 15–18) and are described in detail in Supplementary anti-MM agent used in the clinical practice (21, 22).
data.
The effect of Aplidin on MM1S cells was time dependent:
Combinations of Aplidin with other anti-MM agents. The interac-
tion between Aplidin and other anti-MM agents (dexamethasone,
Inhibition of cell viability progressively increased from day 1 to day
melphalan, doxorubicin, thalidomide, lenalidomide, and bortezomib) was 3 of treatment with any given Aplidin dose (Fig. 1C). Interestingly,
analyzed with the Calcusyn software (Biosoft). Data from cell viability even a 24-hour exposure to Aplidin at concentrations as low as
assays (MTT) were expressed as fraction of cells affected by the dose in 5 nmol/L was sufficient to induce a >50% decrease in MM cell
drug-treated cells compared with untreated cells (control). This program viability. To address the question of whether constant exposure to
is based on the Chou-Talalay method (19) according to the following Aplidin was required for its anti-MM effect, we performed cell
equation: CI = (D)1/(Dx)1 + (D)1(D)2/(Dx)1(Dx)2, where (D)1 and (D)2 are death commitment experiments: MM cells were exposed to a
the doses of drug 1 and drug 2 that have the same x effect when used
clinically relevant dose of Aplidin (40 nmol/L) for variable periods
alone, and CI is the combination index. CI < 1.0 indicates synergism; CI ffi
of time (ranging from 0 to 10 hours) and then were washed to
1.0 indicates an additive effect; and CI > 1.0 corresponds to an
antagonistic effect. MM1S and U266-LR7 cells were treated for 72 h with remove the drug, followed by culture in drug-free media for 3 days.
combinations of Aplidin and other anti-MM drugs at either a nonconstant At that time, cell viability was assessed by MTT and expressed as
ratio ( for suboptimal doses of Aplidin) or a constant ratio combination percent cell viability compared with control cultures. These cell
design, and MTT assays were done as previously described. CI values for death commitment assays evaluated whether a short exposure of
each drug combination were obtained from three different experiments MM cells to Aplidin irreversibly commits them to undergo cell
fulfilling experimental prerequisites of the Calcusyn program. death. In all five MM cell lines tested (JJN-3, S6B45, NCI-H929,
MM xenograft murine model. CB17-SCID mice (The Jackson SKMM1, and KMS-18), even a 1-hour exposure to Aplidin was
Laboratory) were s.c. inoculated into their right flank with 3  106
sufficient to trigger, despite the subsequent washout of the drug, a
MM1S cells in 100 AL of RPMI 1640 and 100 AL of Matrigel basement
membrane matrix (Becton Dickinson). When tumors became palpable,
>50% decrease in cell viability (Fig. 1D). In fact, in four of five lines
mice were randomly assigned into treatment and control groups. Aplidin tested (JJN-3, S6B45, SKMM1, and KMS-18), a 1-hour exposure to
was administered via i.p. injection and mice in the control group Aplidin was sufficient to trigger a >70% decrease in MM cell
received an equal volume of vehicle (cremophor EL/ethanol/water viability. These results supported the notion that, whereas up to 24
for injection, 15%:15%:70%). Caliper measurements of the tumor dia- hours may elapse before the full effect of Aplidin-induced cell death
meters were done every other day and tumor volumes were calculated can be observed, a short exposure to the drug (1 hour) may be
according to the formula of the volume of an ellipse: V = 4/3p  (a/2)  sufficient to irreversibly commit MM cells to their death.
(b /2)2, where a and b correspond to the longest and shortest diameters Effect of Aplidin on MM cells cultured with exogenous
of the tumor, respectively. Animals were sacrificed, according to
interleukin 6, insulin-like growth factor I, and bone marrow
institutional guidelines, when the diameter of their tumors reached
stromal cells. We next evaluated the effect of Aplidin on MM cell
2 cm or when significant toxicity was observed. Differences in tumor
volumes between treated and control groups were evaluated using the proliferation induced by the stimulatory cytokines interleukin 6
unpaired Student t test. Statistical significance was defined as P < 0.05. (IL-6) and insulin-like growth factor I (IGF-I), as well as bone
Survival was estimated from the day of initiation of treatment, and marrow stromal cells (Fig. 2). MM1S cells were treated with
statistical differences were assessed by Kaplan-Meier curves with the increasing doses of Aplidin in the presence or absence of IL-6
log-rank test. (1 nmol/L) or IGF-I (10 nmol/L). Neither of these growth factors

www.aacrjournals.org 5217 Cancer Res 2008; 68: (13). July 1, 2008


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Cancer Research

Figure 1. Activity of Aplidin on MM cell lines. A, dose-response matrix of human MM cell lines to Aplidin in vitro (0–100 nmol/L for 72 h). The viability of Aplidin-treated
MM cells (expressed for each cell line as the percent of its respective control) is visualized in color format according to their values on a linear scale (0–100%).
B, dose effect of Aplidin after 48 h of treatment in MM1S, MM-1R, U266, and U266-LR7 cell lines using the MTT uptake assay (as described in Materials and Methods).
The mean cell viability values are presented for each condition as percent of the respective control of each cell line. A similar pattern of decrease in cell viability
in a dose-dependent manner was observed for the four cell lines tested. C, time course analysis of Aplidin-induced death of MM1S cells. D, results of cell death
commitment experiments of five MM cell lines. MM cells were cultured in the presence of Aplidin (40 nmol/L) for 0 to 10 h and, after washout of the drug, were cultured
for an additional 72 h in the presence of drug-free media to assess the shortest duration of exposure to Aplidin that could commit MM cells to eventual death.
A 1-h exposure to Aplidin was sufficient, despite the subsequent absence of drug in the culture medium, to trigger a significant decrease in viability in the five MM cell
lines tested.

protected against Aplidin-induced growth inhibition (Fig. 2B and was 83% (SD, 14.4; 95% confidence interval, 74.1–91.6). Even at a
C). Binding of MM cells to patient bone marrow stromal cells 10-fold lower dose of Aplidin (10 nmol/L), the average cytotoxic
triggered increased MM cell proliferation, which was attenuated by activity on MM plasma cells remained close to 60%. Supplementary
Aplidin (Fig. 2A). Even low doses of Aplidin (e.g., 10 nmol/L) display Fig. S2A illustrates the effect of Aplidin on MM plasma cells, as well
a potency similar to that of higher concentrations of bortezomib as on normal lymphoid and myeloid cells, from four representative
(e.g., 50 nmol/L). Importantly, Aplidin did not affect the viability of patient cases. Interestingly, whereas Aplidin had a pronounced
bone marrow stromal cells, as determined by MTT assays (data not effect on the viability of MM cells, minimal toxicity was observed
shown). on normal lymphocytes and myeloid cells from the same patient
Efficacy of Aplidin against freshly isolated cells from MM samples (Supplementary Table S2; Supplementary Fig. S2A). Sup-
patients. The activity of Aplidin against MM plasma cells freshly plementary Fig. S2B depicts the results from the analysis of a
isolated cells from 16 patients (8 newly diagnosed and 8 with representative primary MM patient sample, which exhibited a
relapsed/refractory disease) was next analyzed using a multi- dose-dependent induction of cell death in MM plasma cells. Again,
parametric cytometry protocol. This test allows for separate only minimal toxicity was observed on normal mononuclear and
analysis of the induction of cell death in the myelomatous plasma myeloid cells, even at high doses of Aplidin.
cell population versus other bone marrow cells. Thirteen of 16 Evaluation of combinations of Aplidin with other anti-MM
primary MM cell samples tested in vitro were sensitive to the drug. agents. Clinical experience in the therapeutic management of
Two of the three nonresponsive MM primary cell samples were MM patients supports the concept that drug combinations can
isolated from newly diagnosed patients and the third one was induce higher response rates compared with single-agent treat-
from a patient with refractory disease. Interestingly, of the 13 ment (23–26). We therefore evaluated the effect of combinations
cases sensitive to Aplidin in vitro, 7 patients had shown clinical of Aplidin with other anti-MM drugs on the viability of MM1S
resistance to several treatments including alkylating agents and and U266-LR7 cells. Specifically, Aplidin was combined with con-
high-dose dexamethasone (all of them), autologous stem cell ventional agents used in MM treatment (such as dexamethasone,
transplantation (5 cases), bortezomib (3 cases), and thalidomide- doxorubicin, or melphalan), as well as with recently developed
based regimens (2 cases) (Supplementary Table S2). Aplidin drugs (bortezomib, lenalidomide, or thalidomide). In these ex-
(100 nmol/L) induced cell death in >80% of MM plasma cells from periments, both nonconstant ratio ( for suboptimal doses of
8 of 16 patients, and between 60% to 80% in 5 more patients. The drugs) and constant ratio experimental designs were used. The
average cytotoxicity of Aplidin (100 nmol/L) in sensitive patients effects of single and combined treatments after 72 hours were

Cancer Res 2008; 68: (13). July 1, 2008 5218 www.aacrjournals.org


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Antimyeloma Activity of Aplidin

evaluated by MTT assays, and data were analyzed with the combined with lenalidomide or thalidomide (Fig. 3C), a synergistic
Calcusyn program. effect on U266-LR7 cells was again observed, although less
As shown in Fig. 3A, Aplidin increased the anti-MM effect pronounced than that against MM1S cells. The effect of Aplidin
of dexamethasone, melphalan, lenalidomide, thalidomide, and and bortezomib on U266-LR7 cells remained moderately synergis-
bortezomib. Analysis with the Calcusyn program indicated that tic at lower doses and additive at higher doses. Conversely, in
Aplidin exhibited a synergistic effect when combined with lenali- U266-LR7 cells, the Aplidin plus doxorubicin combination
domide or thalidomide (Fig. 3B); Aplidin combinations with dexa- remained antagonistic.
methasone or melphalan were additive; combinations of Aplidin Effects of Aplidin on the cell cycle and viability of MM cells.
and bortezomib presented an additive effect that was even Treatment with 10 nmol/L Aplidin induced a modest increase in
moderately synergistic when low doses of both drugs were used. the percentage of MM1S cells in the G2-M phase (Supplementary
No synergism was observed in combinations with doxorubicin Fig. S3A). A similar increase in the percent of cells in G2-M was
(data not shown). In experiments with U266-LR7 cells (resistant observed after treatment of MM1R cells with 1 nmol/L Aplidin:
to melphalan), combinations with dexamethasone or melphalan Percent of cells in G2-M was 19% in control cells versus 29% in 1
were not amenable to evaluation with the Calcusyn program nmol/L Aplidin–treated cells (data not shown). Higher doses
because the prerequisite of a dose-effect relationship for each of of Aplidin (100 nmol/L) triggered a clear sub-G0 accumulation
these drugs as single agents was not fulfilled. When Aplidin was (data not shown). The latter fact, together with the increased
Annexin V staining obtained in patient plasma cells, indicates
that Aplidin does not merely block the proliferation of MM cells
but also actively triggers cell death. This hypothesis is also
supported by a time-dependent increase in Annexin V staining of
MM1S cells treated with Aplidin (Supplementary Fig. S3B). In
addition, Aplidin induced DNA laddering in MM1S cells (data not
shown).
To gain insights into the mechanism(s) of action of Aplidin on
MM cells, we performed gene expression profiling, as well as
biochemical and cell biological analyses, with emphasis on the
pathways involved in the regulation of apoptosis. Gene expression
profiling identified 805 transcripts that were significantly up-
regulated or down-regulated by z2-fold with Aplidin treatment.
The classification of these genes according to functional categories
indicated that 9.8% were involved in apoptosis/responses to stress,
22.4% in the control of cell cycle/proliferation, 5.8% were kinases
that participate in cell cycle and survival signaling, and 1%
participated in adhesion (Supplementary Fig. S3C). These results
suggest that genes involved in cell cycle and cell death regulation
may participate in the mechanism of action of Aplidin. Indeed,
microarray data showed that Aplidin down-regulated several genes
required for cell proliferation and/or cell survival (MYBL2, BUB1,
BUB1B, cyclin B1, cyclin E1, CENPF, MCM2, MCM4, MCM5, and
survivin; Supplementary Fig. S3). In addition, Aplidin up-regulated
the transcript levels of potential regulators of apoptosis, such as
GADD45A, GADD45B, TRAIL, CASP9, CASP6, CIDEC, and Smac
(Supplementary Fig. S3D). Of interest, we also observed an increase
in the expression of c-JUN and a decrease in the expression of MYC
(Fig. 4; Supplementary Figs. S4–S8). Previous data from our group
and others have shown that activation of the pathway and/or up-
regulation of c-JUN expression is associated with the apoptosis
induced by novel drugs such as bortezomib (11). Furthermore,
extensive work in MM and other neoplasias has indicated that myc
is an important oncogenic protein and a putative therapeutic
target (as reviewed in ref. 27). A complete list of transcripts modu-
lated by Aplidin treatment is included as a Supplementary table,
and schematic representations of functional pathways involving
Aplidin-modulated transcripts are depicted in Supplementary
figures.
Drug-induced apoptotic cell death is frequently linked to the
Figure 2. Effect of Aplidin on MM cells stimulated with proliferative cytokines or
cocultured with bone marrow stromal cells: MM1S cells were treated for 48 h
release from mitochondria to the cytosol of proteins, which then
with the indicated concentrations of Aplidin in the presence or absence of bone partner with other cytosol-resident proteins to activate caspases.
marrow stromal cells (BMSC ) derived from a MM patient (A), IL-6 (B), Loss of mitochondrial membrane potential (Dw m) often reflects an
or IGF-I (C ). DNA synthesis was determined by measuring bromodeoxyuridine
(BrdUrd ) incorporation during the last 8 h of 48-h cultures. Columns, mean of increase in mitochondrial outer membrane permeability. Treat-
quadruplicates; bars, SD. ment of MM1S cells with Aplidin caused a decrease in Dw m,

www.aacrjournals.org 5219 Cancer Res 2008; 68: (13). July 1, 2008


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Cancer Research

indicative of an increase in mitochondrial outer membrane Bcl-2 family member Mcl-1 (Fig. 4B). No changes in Bcl-2
permeability (Fig. 4A). Bcl-2 family members act as important expression were induced by Aplidin, whereas a small increase in
regulators of mitochondrial outer membrane permeability, and Bcl-X was noticed (Fig. 4B). Apoptosis triggered by Aplidin was
Western blot analyses indicated that Aplidin down-regulated the further confirmed by cleavage of poly(ADP-ribose) polymerase,

Figure 3. Evaluation of combinations of Aplidin with other


anti-MM agents. A, Aplidin (1.25 nmol/L) was combined with
dexamethasone, bortezomib, melphalan, lenalidomide, or
thalidomide; after 48 h, MTT assays were done on MM1S cells.
B, calculated CIs of different double combinations for the
MM1S cell line. A fraction affected-CI plot simulation is
provided when a constant ratio design is used. CR, constant
ratio; NCR, nonconstant ratio. C, CIs observed for the
U266-LR7 cell line. CIs of <0.3, 0.3 to 0.7, 0.7 to 0.85, 0.85 to
0.90, 9.0 to 1.10, and >1.10 indicate strong synergism,
synergism, moderate synergism, slight synergism, additive
effect, and antagonism, respectively.

Cancer Res 2008; 68: (13). July 1, 2008 5220 www.aacrjournals.org


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Antimyeloma Activity of Aplidin

Figure 4. Analysis of the pathways involved in


Aplidin-induced apoptosis. A, MM1S cells were
treated with Aplidin (100 nmol/L), and Dw m analyses
done with [DiOC6(3)low] by flow cytometry.
B, MM1S cells were treated with Aplidin (100 nmol/L)
for the indicated times, and expressions of
poly(ADP-ribose) polymerase (PARP ), caspase-3,
caspase-7, caspase-8, caspase-9, Bcl-2, Bcl-X,
Mcl-1, and myc proteins were analyzed by Western
blotting of cell extracts with specific antibodies.
C, effect of the caspase inhibitors z-VAD-fmk,
z-IETD-fmk, and z-LEHD-fmk on Aplidin-induced cell
death. MM1S cells were plated and pretreated,
where indicated, with caspase inhibitors (50 Amol/L)
for 60 min. Aplidin (100 nmol/L) was added to the
corresponding samples and the experiment was
continued for 6 and 12 h. Annexin V staining was
carried out as described above.

caspase-3, caspase-7, caspase-8, and caspase-9, with generation Fas/CD95 death receptor in lipid rafts by isolating these membrane
of active low M r cleaved fragments (Fig. 4B). To investigate the microdomains from untreated and drug-treated MM cells by taking
importance of caspases in cell death triggered by Aplidin, we advantage of their insolubility in Triton X-100 lysis buffer at 4jC,
evaluated the ability of the pan-caspase inhibitor z-VAD-fmk, followed by discontinuous sucrose gradient centrifugation. GM1-
the caspase-8 inhibitor z-IETD-fmk, and the caspase-9 inhibitor containing lipid rafts, at the upper part ( fractions 3–5) of the
z-LEHD-fmk to protect against Aplidin-induced apoptosis. As sucrose gradient (Fig. 5A), were identified using cholera toxin B
shown in Fig. 4C, preincubation with z-VAD-fmk attenuated subunit conjugated to horseradish peroxidase. We observed that
Aplidin-induced cell death, whereas no effect was observed with Fas/CD95 was located in the soluble fractions ( fractions 9–12) of
caspase-8 and caspase-9 inhibitors, suggesting that only the the sucrose gradient and not in the detergent-insoluble lipid
blockade of both the intrinsic and extrinsic pathways can partially raft region in untreated MM144 cells, indicating that Fas/CD95
block Aplidin-induced apoptosis. is excluded from the lipid rafts in untreated MM144 cells, in
Aplidin-induced translocation of Fas/CD95 death receptor agreement with previous studies (18). Importantly, Aplidin treat-
into lipid rafts. A novel mechanism regulating Fas/CD95 death ment induced a partial recruitment of Fas/CD95 into the lipid raft
receptor activation in apoptosis involves its translocation into lipid region ( fractions 3–5) of the sucrose gradient (Fig. 5A). Conversely,
rafts (16). In this regard, Aplidin-induced apoptosis in leukemic disruption of lipid rafts by methyl-h-cyclodextrin, which interferes
cells involves translocation of Fas/CD95 in lipid rafts (28), and with protein association with lipid rafts by cholesterol depletion
recruitment of death receptors in lipid rafts has recently been (16), partially inhibited Aplidin-induced cell death in MM144 cells
shown to be a crucial event in the anti-MM activity of the synthetic (Fig. 5B). These results suggest that Aplidin-induced cell death
alkyl-lysophospholipid analogues edelfosine and perifosine (18). in MM cells involves, at least in part, Fas/CD95 translocation
Therefore, we analyzed whether Aplidin induced translocation of into lipid rafts.

www.aacrjournals.org 5221 Cancer Res 2008; 68: (13). July 1, 2008


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Cancer Research

The role of the JNK and p38 apoptotic pathways in the primary MM patient cells, we next studied the in vivo anti-MM
mechanism of action of Aplidin. To further explore the effect of Aplidin in a human plasmocytoma model in immunode-
mechanism of action of Aplidin on MM cells, we analyzed the ficient mice. To address the possibility that the side effect profile of
expression and activation status of JNK, p38, extracellular signal– Aplidin is strain dependent, we first conducted a pilot study to
regulated kinase (Erk)-1/2, and Erk5 in Aplidin-treated MM cells. explore the potential toxicity of this drug in CB17-SCID mice. For
Aplidin (100 nmol/L) induced activation of JNK, which was already this purpose, 20 mice were injected i.p. with different doses of
detectable by 15 minutes of Aplidin treatment and reached a Aplidin and were serially monitored for the development of any
maximum at 1 to 3 hours (Fig. 5C). Activation of JNK was also significant toxicity. Maximum tolerated dose for this strain was
observed in MM1R cells (data not shown). Aplidin also rapidly established at a total weekly dose of 700 Ag/kg.
increased phosphorylation of p38. On the other hand, no significant A separate cohort of 16 mice were treated with a total weekly
effect on the Erk1/2 or Erk5 pathways was observed at times up to dose of 700 Ag/kg Aplidin at two different schedules of treatment:
6 hours of treatment. At later times, Aplidin decreased Erk1/2 and nine mice received Aplidin 100 Ag/kg i.p. daily and seven were given
Erk5 expression. To further characterize the biological consequen- Aplidin 140 Ag/kg i.p. 5 days a week. Nine mice served as control
ces of the activation of the JNK and p38 signaling pathways, cells cohort and received i.p. injections of the vehicle alone. The toxicity
were treated with or without inhibitors for p38 kinase (SB203580; profile of Aplidin was quite favorable, evidenced by a body weight
25 Amol/L) and JNK (SP600125; 25 Amol/L) before Aplidin treat- loss of only 6% to 8% in the treated groups as compared with the
ment. Both the p38 inhibitor SB203580 and JNK inhibitor SP600125 control group. Only one mouse in the 140 Ag/kg cohort had to be
partially rescued MM1S cells from Aplidin-induced cell death euthanized (on day 26 after initiation of treatment) due to a
(Fig. 5D), implicating p38 and JNK activation in the mechanism of significant loss in body weight (>20%). Aplidin treatment decreased
action of this drug. Comparison with bortezomib indicated tumor growth in both cohorts of treated mice, and this effect was
differences in the sensitivity to mitogen-activated protein kinase greater in the group receiving 140 Ag/kg of Aplidin 5 days a week
(MAPK) inhibitors because the p38 inhibitor (SB203580) was (Fig. 6A). The differences in tumor volumes between the vehicle and
more efficient in preventing the action of Aplidin (Fig. 5D). In the 140 Ag/kg cohort were statistically significant from day 9 of
fact, inhibition of the p38 route had a small effect on bortezomib- treatment (P = 0.01) until the end of the experiment. With 100 Ag/kg
induced cell death (Fig. 5D), in agreement with the poor treatment, the tumor volume also decreased compared with the
p38-stimulating activity of bortezomib (Fig. 5C). controls, but differences did not reach statistical significance
Aplidin exhibits in vivo anti-MM activity in a xenograft- (P = 0.08). Kaplan-Meier survival analysis revealed a significant
plasmocytoma murine model. In view of the potent in vitro prolongation of survival for the Aplidin-treated groups compared
activity of Aplidin against MM cell lines and freshly isolated with the vehicle-treated controls (log-rank P = 0.02; Fig. 6B).

Figure 5. Effects of Aplidin treatment on Fas/CD95 translocation into lipid rafts and on JNK and p38 signaling. A, translocation of Fas/CD95 into lipid rafts in
Aplidin-treated MM cells. Untreated MM144 cells (Control ) and MM144 cells treated with 10 nmol/L Aplidin for 15 h (Aplidin ) were lysed in 1% Triton X-100 and
fractionated by centrifugation on a discontinuous sucrose density gradient. An equal volume of each collected fraction was subjected to SDS-PAGE before analysis of
Fas/CD95 with a specific antibody. Location of GM1-containing lipid rafts (fractions 3–5) was determined using cholera toxin B subunit conjugated to horseradish
peroxidase. Representative of three experiments. B, MM144 cells were untreated (Control ) or pretreated with methyl-h-cyclodextrin (MCD ) and then incubated with
10 nmol/L Aplidin (APL ) for 24 h and examined for the percentage of apoptotic cells by flow cytometry. Columns, mean of three independent determinations;
bars, SE. C, MM1S cells were treated for the indicated times with 100 nmol/L Aplidin (left ) or 25 nmol/L bortezomib (right ) and lysed. One part of the cell extracts
were used for the analyses of JNK activity using glutathione S-transferase-c-Jun. Another part of the extracts were used to detect p-Erk1/2, Erk1/2, p-p38, and
poly(ADP-ribose) polymerase by Western blots. Erk5 was analyzed by Western blotting of anti-Erk5 immunoprecipitates. Shown are the results of an experiment that
was repeated twice. D, MM1S cells were pretreated for 60 min with inhibitors for p38 kinase (SB203580) and JNK (SP600125) and subsequently treated with
Aplidin for 6 and 12 h. Induction of cell death in each condition was assessed by Annexin V staining.

Cancer Res 2008; 68: (13). July 1, 2008 5222 www.aacrjournals.org


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Antimyeloma Activity of Aplidin

still active against MM cells; the ability to overcome the protective


effect of growth factors, such as IL-6 or IGF-I, on MM cells; and the
additive (bortezomib and melphalan) or synergistic (dexametha-
sone, thalidomide, and lenalidomide) effects when Aplidin is
combined with other anti-MM agents. Another positive feature of
the anti-MM activity of Aplidin is that even a brief exposure of
MM cells to this agent is capable of committing them to undergo
cell death. Indeed, a 1-hour incubation with clinically relevant
concentrations of Aplidin, based on pharmacokinetic studies from
clinical trials in solid tumors (20), was capable of irreversibly
initiating the process of MM cell death in vitro. Finally, this anti-
MM activity of Aplidin in MM cells was confirmed in an in vivo
setting, as evidenced by a delay in tumor growth and prolongation
of survival in Aplidin-treated MM-bearing mice. Importantly, the
toxicity profile of Aplidin in this model was favorable, as evidenced
by the lack of major side effects.
Cell cycle studies indicated that low doses of Aplidin caused G2-
M accumulation. In addition, molecular profiling studies indicated
that Aplidin down-regulated several genes required for cell cycle
progression. In addition to the cell cycle effects, Aplidin triggered
increased Annexin V staining, decreased Dw m, and DNA laddering.
It is important to emphasize the prompt increase in Annexin V
staining, which is detectable early, after 3 hours of treatment. Due
to this rapid proapoptotic action of Aplidin, we selected early times
of exposure to Aplidin (4 hours) to identify genes involved in the
triggering of cell death. These gene expression analyses identified
several genes, including c-JUN and c-MYC, that control cell cycle
and apoptotic phenomena. This apoptotic cell death was also
supported by the induction of poly(ADP-ribose) polymerase pro-
Figure 6. In vivo anti-MM effect of Aplidin. CB17-SCID mice were inoculated cessing as well as caspase-3, caspase-7, caspase-8, and caspase-9
with 3  106 MM1S cells s.c. in the right flank. Treatment was started when cleavage induced by Aplidin. This treatment also induced a sub-
tumors became palpable. Mice were randomly assigned into three groups
(with equal average tumor volumes before initiation of treatments): vehicle stantial decrease in Mcl-1, an important regulator of MM survival.
(n = 9), Aplidin 140 Ag/kg/d i.p.  5 d/wk (n = 7), and Aplidin 100 Ag/kg/d Finally, studies with inhibitors of the different caspases confirmed
i.p.  7 d/wk (n = 9). Caliper measurements of tumor diameters were done that Aplidin induced cell death via the caspase machinery.
every other day, and tumor volumes were estimated with the formula
V = 4/3p  (a/2)  (b/2)2, where a and b correspond to the longest and shortest However, because the inhibitory effect of these caspase inhibitors
diameters, respectively. Aplidin significantly inhibited the increase in tumor was only partial, it remains possible that caspase-independent
growth of the MM xenograft (A; P = 0.01) and increased survival (B ; P = 0.02) mechanisms may also contribute to apoptotic cell death triggered
compared with control mice. A, bars, SE.
by Aplidin.
Our results offer new insights into the mechanism of action of
Aplidin on tumor cells. Here we found that Aplidin induces
Discussion apoptosis in MM cells, at least in part, through the translocation of
The results of this preclinical study suggest a favorable profile of the death receptor Fas/CD95 into lipid rafts. In addition, our data
the in vitro and in vivo anti-MM activities of Aplidin. First, this suggest that this is not the only mechanism of action of Aplidin.
compound exhibits potent in vitro activity against both MM cell The difficulty in delineating a singular molecular target for Aplidin
lines and primary MM tumor cells, including cell lines resistant to results, in part, from its complex molecular structure, which
established anti-MM treatments. Furthermore, Aplidin was able to includes many potential biologically active moieties, and the
induce cytotoxicity in myelomatous plasma cells obtained from pleiotropicity of its molecular sequelae. Both of these factors,
patients with clinical resistance to most of the current treatments which often apply as well for other similarly identified natural
for the clinical management of MM, including combination products, can confound our ability to pinpoint specific candidate
chemotherapy regimens, autologous stem cell transplant, as well molecule(s) that might physically interact with Aplidin and
as novel agents such as bortezomib or thalidomide and their function as exclusive mediator(s) of its antitumor effects. Our
combinations. Interestingly, among the primary tumor samples studies, however, do provide insight into the pathways involved in
tested with Aplidin, one of the most sensitive ones (patient sample the anti-MM activity of this compound. The rapid commitment of
no. 3) had shown clinical refractoriness to all of these treatments. MM cells to undergo cell death on exposure to Aplidin and the
These data suggest a different mechanism of action of Aplidin than early activation of JNK signaling are similar to the effects of the
other antimyeloma agents and suggest that it may overcome proteasome inhibitor bortezomib. However, whereas Aplidin also
resistance generated against currently available therapies for MM. activates p38, bortezomib treatment is enhanced by p38 inhibition
Moreover, our studies also show that the potency of Aplidin is high (29). Therefore, although equimolar concentrations of Aplidin and
and comparable to that of bortezomib, a most potent anti-MM bortezomib have similar potency in rapidly killing MM cells in vitro,
agent in vitro. Additional favorable aspects of Aplidin include the they have distinct differences in their molecular effects (11). This
sparing of normal hematopoietic cells at concentrations that are apparent dichotomy may be related to the ability of the p38 MAPK

www.aacrjournals.org 5223 Cancer Res 2008; 68: (13). July 1, 2008


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Cancer Research

pathway to respond to different stimuli with diverse biological additive effect with other anti-MM agents, coupled with promising
effects, which can also depend on the specific cellular context (30). clinical data on the safety profile and objective clinical responses in
For example, p38 can trigger apoptosis in some cells but inhibit its heavily pretreated refractory patients, indicate that this novel
induction in other pathophysiologic settings (30). It has been compound (alone or in combination with other agents) may
proposed (as reviewed in ref. 31) that such stress-activated MAPK contribute to improve the outcome of MM patients.
pathways may function as a double-edged sword: They may protect
normal cells against environmental stress and malignant cells
against forms of stress inherent to the neoplastic transformation Disclosure of Potential Conflicts of Interest
(e.g., oncogenic ‘‘addiction,’’ drug treatments); conversely, they may C.S. Mitsiades: Honoraria, Pharmion and Millenium. A. Pandiella: Commercial
also trigger apoptosis if the stress levels exceed a threshold (31), research grant, PharmaMar. P. Aviles: Employment, PharmaMar. G. Faircloth:
Employment, PharmaMar. P.G. Richardson: Speakers bureau/honoraria, Millennium
which in turn is influenced by the particular context in which the and Celgene. J.F. San-Miguel: Commercial research support, PharmaMar. K.C.
tumor cells are placed. We hypothesize that p38 activation, in the Anderson: Commercial research support, Millennium and Celgene; speakers bureau/
honoraria, Novartis, Millennium, and Celgene. C.S. Mitsiades and N. Mitsiades are
context of Aplidin treatment, is complemented by other molecular inventors of a patent, co-owned by Dana-Farber and PharmaMar, that relates to the
events (also triggered by the compound) that reinforce the pro- present work. The other authors disclosed no potential conflicts of interest.
apoptotic role of p38. Such complementary molecular events
include the suppression of myc (an important regulator of pro-
Acknowledgments
liferative/antiapoptotic responses for MM cells; ref. 27) or anti-
apoptotic molecules such as survivin (32, 33) and/or the increased Received 10/2/2007; revised 3/21/2008; accepted 4/11/2008.
Grant support: The Multiple Myeloma Research Foundation (C.S. Mitsiades,
expression of proapoptotic modulators such as TRAIL (9, 32, 34, 35) N. Mitsiades, and J.F. San-Miguel), The Lauri Strauss Leukemia Foundation (C.S.
and Smac (36, 37). Mitsiades and N. Mitsiades), The International Waldenstrom’s Macroglobulinemia
This favorable preclinical profile of the anti-MM activity of Foundation (C.S. Mitsiades), The Fund to Cure Myeloma (K.C. Anderson), The
Fulbright Commission (C.J. McMullan), Doris Duke Distinguished Clinical Research
Aplidin provided the rationale for the initiation of a clinical trial of Scientist Award (K.C. Anderson), Fondo de Investigación Sanitaria and European
this agent in patients with advanced MM (38). Preliminary results Commission grants FIS-FEDER 04/0843 (F. Mollinedo) and 06/0813 (C. Gajate),
Ministerio de Educación y Ciencia grant SAF2005-04293 (F. Mollinedo), Fundación de
of this ongoing clinical study indicate that Aplidin has promising Investigación Médica Mutua Madrileña (F. Mollinedo), and Fundación ‘‘la Caixa’’ grant
clinical anti-MM activity in heavily pretreated MM patients BM05-30-0 (F. Mollinedo). C. Gajate is supported by the Ramón y Cajal Program from
refractory to several conventional and novel anti-MM agents (e.g., the Ministerio de Educación y Ciencia of Spain; E.M. Ocio is supported by Carlos III-
Fondo de Investigación Sanitaria-Ministerio de Ciencia y Tecnologı́a grant CM04/0001;
bortezomib and/or lenalidomide; ref. 38). A high proportion of This work was supported by a grant from the Ministry of Education and Science of
patients in this trial had received prior high-dose therapy and Spain (BFU2006-01813/BMC), and by an Institutional Cancer Center Network program
from the ISCIII. The Cancer Research Institute receives support from the European
autologous stem cell transplant (60%), thalidomide (58%), and/or Community through the regional development funding program (FEDER). P. Maiso
bortezomib (48%). Among 26 evaluable patients, 2 (8%) achieved was supported by the FIS-FEDER through projects to J.F. San-Miguel, and a Spanish
partial response, 3 (12%) showed minor response, and 8 (31%) had Myeloma Network Program (G03/136). M. Garayoa is supported by ‘‘Instituto de Salud
Carlos III-Fondo de Investigación Sanitaria-Ministerio de Ciencia y Tecnologı́a’’ grant
stable disease. Due to the enhanced anti-MM activity observed CP 05/0279; C.S. Mitsiades is a Special Fellow of the Leukemia and Lymphoma Society
with the in vitro combination of these agents, high-dose and is supported by the ‘‘Dunkin Donuts Rising Stars’’ Program at the Dana-Farber
dexamethasone was added to Aplidin in patients not achieving Cancer Institute.
The costs of publication of this article were defrayed in part by the payment of page
response, and preliminary results of the clinical combination charges. This article must therefore be hereby marked advertisement in accordance
regimen are also very encouraging. Importantly, in the clinical trials with 18 U.S.C. Section 1734 solely to indicate this fact.
Aplidin did not cause significant toxicities that are present in We thank Joseph M. Negri, Gary Fanourakis, Supriya Rao, Despoina Sykoutri,
Montserrat Martin, and Laura San Segundo for technical assistance.
established anti-MM agents, such as neuropathy or myelosuppres- C.S. Mitsiades, E.M. Ocio, and A. Pandiella designed research, performed research,
sion (38). Furthermore, preliminary data indicate that its analyzed data, and wrote the paper; P. Maiso, C. Gajate, M. Garayoa, and F. Mollinedo
designed research, performed research, analyzed data, and contributed to the writing
combination with dexamethasone is not associated with the of the paper; N. Mitsiades, C.J. McMullan, J-C. Montero, D. Vilanova, and M.V. Mateos
thromboembolic complications observed with the combinations of designed research, performed research, and analyzed data; T. Hideshima and D.
glucocorticoids with thalidomide or lenalidomide (39, 40). Chauhan contributed research tools; N.C. Munshi, G. Otero, P. Aviles, and
G. Faircloth contributed research tools and analyzed data; P.G. Richardson, J.F. San-
In conclusion, our preclinical data on the potent anti-MM Miguel, and K.C. Anderson designed research, analyzed data, and contributed to the
activity of single-agent Aplidin and its potential in vitro synergistic/ writing of the paper.

References B (DDB), a depsipeptide isolated from Mediterranean peutic agents: therapeutic applications. Blood 2003;101:
tunicates. Cancer Lett 1996;102:31–7. 2377–80.
1. Richardson PG, Hideshima T, Mitsiades C, Anderson 7. Chauhan D, Uchiyama H, Urashima M, Yamamoto K, 11. Mitsiades N, Mitsiades CS, Poulaki V, et al. Molecular
KC. The emerging role of novel therapies for the Anderson KC. Regulation of interleukin 6 in multiple sequelae of proteasome inhibition in human multiple
treatment of relapsed myeloma. J Natl Compr Canc myeloma and bone marrow stromal cells. Stem Cells myeloma cells. Proc Natl Acad Sci U S A 2002;99:14374–9.
Netw 2007;5:149–62. 1995;13 Suppl 2:35–9. 12. Carvajal-Vergara X, Tabera S, Montero JC, et al.
2. Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. 8. Hideshima T, Richardson P, Chauhan D, et al. The Multifunctional role of Erk5 in multiple myeloma. Blood
Cancer statistics, 2007. CA Cancer J Clin 2007;57:43–66. proteasome inhibitor PS-341 inhibits growth, induces 2005;105:4492–9.
3. Newman DJ, Cragg GM. Natural products as sources apoptosis, and overcomes drug resistance in human 13. Maiso P, Carvajal-Vergara X, Ocio EM, et al. The
of new drugs over the last 25 years. J Nat Prod 2007;70: multiple myeloma cells. Cancer Res 2001;61:3071–6. histone deacetylase inhibitor LBH589 is a potent
461–77. 9. Mitsiades CS, Treon SP, Mitsiades N, et al. TRAIL/ antimyeloma agent that overcomes drug resistance.
4. Rinehart KL. Antitumor compounds from tunicates. Apo2L ligand selectively induces apoptosis and over- Cancer Res 2006;66:5781–9.
Med Res Rev 2000;20:1–27. comes drug resistance in multiple myeloma: therapeutic 14. Orfao A, Garcia-Sanz R, Lopez-Berges MC, et al. A
5. O’Hanlon LH. Scientists are searching the seas for applications. Blood 2001;98:795–804. new method for the analysis of plasma cell DNA content
cancer drugs. J Natl Cancer Inst 2006;98:662–3. 10. Mitsiades N, Mitsiades CS, Richardson PG, et al. The in multiple myeloma samples using a CD38/propidium
6. Urdiales JL, Morata P, Nunez De Castro I, Sanchez- proteasome inhibitor PS-341 potentiates sensitivity of iodide double staining technique. Cytometry 1994;17:
Jimenez F. Antiproliferative effect of dehydrodidemnin multiple myeloma cells to conventional chemothera- 332–9.

Cancer Res 2008; 68: (13). July 1, 2008 5224 www.aacrjournals.org


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research
DOI:10.1158/0008-5472.CAN-07-5725
Antimyeloma Activity of Aplidin

15. Li C, Wong WH. Model-based analysis of oligonucle- and/or refractory multiple myeloma (MM): final results myeloma: therapeutic applications. Blood 2002;99:
otide arrays: expression index computation and outlier of a multicenter phase 1 trial. Blood 2006;108:124A–A. 4079–86.
detection. Proc Natl Acad Sci U S A 2001;98:31–6. 24. Orlowski RZ. Bortezomib and its role in the 33. Romagnoli M, Trichet V, David C, et al. Significant
16. Gajate C, Mollinedo F. The antitumor ether lipid management of patients with multiple myeloma. Expert impact of survivin on myeloma cell growth. Leukemia
ET-18-OCH(3) induces apoptosis through translocation Rev Anticancer Ther 2004;4:171–9. 2007;21:1070–8.
and capping of Fas/CD95 into membrane rafts in 25. Rajkumar SV, Hayman SR, Lacy MQ, et al. Combina- 34. Mitsiades N, Mitsiades CS, Poulaki V, et al. Apoptotic
human leukemic cells. Blood 2001;98:3860–3. tion therapy with lenalidomide plus dexamethasone signaling induced by immunomodulatory thalidomide
17. Cheng PC, Dykstra ML, Mitchell RN, Pierce SK. A role (Rev/Dex) for newly diagnosed myeloma. Blood 2005; analogs in human multiple myeloma cells: therapeutic
for lipid rafts in B cell antigen receptor signaling and 106:4050–3. implications. Blood 2002;99:4525–30.
antigen targeting. J Exp Med 1999;190:1549–60. 26. Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp 35. Mitsiades N, Mitsiades CS, Poulaki V, Anderson KC,
18. Gajate C, Mollinedo F. Edelfosine and perifosine PR. Phase III clinical trial of thalidomide plus dexa- Treon SP. Intracellular regulation of tumor necrosis
induce selective apoptosis in multiple myeloma by methasone compared with dexamethasone alone in factor-related apoptosis-inducing ligand-induced apo-
recruitment of death receptors and downstream signal- newly diagnosed multiple myeloma: a clinical trial ptosis in human multiple myeloma cells. Blood 2002;99:
ing molecules into lipid rafts. Blood 2007;109:711–9. coordinated by the Eastern Cooperative Oncology 2162–71.
19. Chou TC, Talalay P. Quantitative analysis of dose- Group. J Clin Oncol 2006;24:431–6. 36. Chauhan D, Li G, Hideshima T, et al. JNK-dependent
effect relationships: the combined effects of multiple 27. Bergsagel PL, Kuehl WM. Molecular pathogenesis release of mitochondrial protein, Smac, during apopto-
drugs or enzyme inhibitors. Adv Enzyme Regul 1984;22: and a consequent classification of multiple myeloma. sis in multiple myeloma (MM) cells. J Biol Chem 2003;
27–55. J Clin Oncol 2005;23:6333–8. 278:17593–6.
20. Maroun JA, Belanger K, Seymour L, et al. Phase I 28. Gajate C, Mollinedo F. Cytoskeleton-mediated death 37. Chauhan D, Neri P, Velankar M, et al. Targeting
study of Aplidine in a dailyx5 one-hour infusion every receptor and ligand concentration in lipid rafts forms mitochondrial factor Smac/DIABLO as therapy for
3 weeks in patients with solid tumors refractory to apoptosis-promoting clusters in cancer chemotherapy. multiple myeloma (MM). Blood 2007;109:1220–7.
standard therapy. A National Cancer Institute of Canada J Biol Chem 2005;280:11641–7. 38. Mateos MV, Cibeira MT, Blade J, et al. Phase II trial
Clinical Trials Group study: NCIC CTG IND 115. Ann 29. Hideshima T, Akiyama M, Hayashi T, et al. Targeting with plitidepsin (Aplidin) alone and in combination
Oncol 2006;17:1371–8. p38 MAPK inhibits multiple myeloma cell growth in the with dexamethasone (dex) in patients with relapsed/
21. Richardson PG, Sonneveld P, Schuster MW, et al. bone marrow milieu. Blood 2003;101:703–5. refractory multiple myeloma: preliminary results. Hae-
Bortezomib or high-dose dexamethasone for relapsed 30. Bradham C, McClay DR. p38 MAPK in development matologica 2007;92:153–4.
multiple myeloma. N Engl J Med 2005;352:2487–98. and cancer. Cell Cycle 2006;5:824–8. 39. Rajkumar SV, Blood E. Lenalidomide and venous
22. Richardson PG, Mitsiades C, Hideshima T, Anderson 31. Dhillon AS, Hagan S, Rath O, Kolch W. MAP thrombosis in multiple myeloma. N Engl J Med 2006;354:
KC. Bortezomib: proteasome inhibition as an effective kinase signalling pathways in cancer. Oncogene 2007; 2079–80.
anticancer therapy. Annu Rev Med 2006;57:33–47. 26:3279–90. 40. Knight R, DeLap RJ, Zeldis JB. Lenalidomide and
23. Richardson PG, Jagannath S, Avigan DE, et al. 32. Mitsiades N, Mitsiades CS, Poulaki V, et al. Biologic venous thrombosis in multiple myeloma. N Engl J Med
Lenalidomide plus bortezomib (Rev-Vel) in relapsed sequelae of nuclear factor-nB blockade in multiple 2006;354:2079–80.

www.aacrjournals.org 5225 Cancer Res 2008; 68: (13). July 1, 2008


Downloaded from cancerres.aacrjournals.org on April 12, 2011
Copyright © 2008 American Association for Cancer Research

You might also like