ArticlePDF Available

Chen S, Dai Y, Harada H, Dent P, Grant S.. Mcl-1 downregulation potentiates ABT-737 lethality by cooperatively inducing Bak activation and Bax translocation. Cancer Res 67: 782-791

Authors:

Abstract

The Bcl-2 antagonist ABT-737 targets Bcl-2/Bcl-xL but not Mcl-1, which may confer resistance to this novel agent. Here, we show that Mcl-1 down-regulation by the cyclin-dependent kinase (CDK) inhibitor roscovitine or Mcl-1-shRNA dramatically increases ABT-737 lethality in human leukemia cells. ABT-737 induces Bax conformational change but fails to activate Bak or trigger Bax translocation. Coadministration of roscovitine and ABT-737 untethers Bak from Mcl-1 and Bcl-xL, respectively, triggering Bak activation and Bax translocation. Studies employing Bax and/or Bak knockout mouse embryonic fibroblasts (MEFs) confirm that Bax is required for ABT-737+/-roscovitine lethality, whereas Bak is primarily involved in potentiation of ABT-737-induced apoptosis by Mcl-1 down-regulation. Ectopic Mcl-1 expression attenuates Bak activation and apoptosis by ABT-737+roscovitine, whereas cells overexpressing Bcl-2 or Bcl-xL remain fully sensitive. Finally, Mcl-1 knockout MEFs are extremely sensitive to Bak conformational change and apoptosis induced by ABT-737, effects that are not potentiated by roscovitine. Collectively, these findings suggest down-regulation of Mcl-1 by either CDK inhibitors or genetic approaches dramatically potentiate ABT-737 lethality through cooperative interactions at two distinct levels: unleashing of Bak from both Bcl-xL and Mcl-1 and simultaneous induction of Bak activation and Bax translocation. These findings provide a mechanistic basis for simultaneously targeting Mcl-1 and Bcl-2/Bcl-xL in leukemia.
2007;67:782-791. Cancer Res
Shuang Chen, Yun Dai, Hisashi Harada, et al.
Translocation
Cooperatively Inducing Bak Activation and Bax
Mcl-1 Down-regulation Potentiates ABT-737 Lethality by
Updated version
http://cancerres.aacrjournals.org/content/67/2/782
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/67/2/782.full.html#ref-list-1
This article cites by 47 articles, 31 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/67/2/782.full.html#related-urls
This article has been cited by 77 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Mcl-1 Down-regulation Potentiates ABT-737 Lethality by
Cooperatively Inducing Bak Activation and
Bax Translocation
Shuang Chen,
1
Yun Dai,
1
Hisashi Harada,
1
Paul Dent,
2
and Steven Grant
1,2,3
Departments of
1
Medicine,
2
Biochemistry, and
3
Pharmacology, Virginia Commonwealth University and Massey Cancer Center,
Richmond, Virginia
Abstract
The Bcl-2 antagonist ABT-737 targets Bcl-2/Bcl-xL but not
Mcl-1, which may confer resistance to this novel agent. Here,
we show that Mcl-1 down-regulation by the cyclin-dependent
kinase (CDK) inhibitor roscovitine or Mcl-1-shRNA dramati-
cally increases ABT-737 lethality in human leukemia cells.
ABT-737 induces Bax conformational change but fails to
activate Bak or trigger Bax translocation. Coadministration
of roscovitine and ABT-737 untethers Bak from Mcl-1 and
Bcl-xL, respectively, triggering Bak activation and Bax trans-
location. Studies employing Bax and/or Bak knockout mouse
embryonic fibroblasts (MEFs) confirm that Bax is required
for ABT-737 F roscovitine lethality, whereas Bak is primarily
involved in potentiation of ABT-737–induced apoptosis by
Mcl-1 down-regulation. Ectopic Mcl-1 expression attenuates
Bak activation and apoptosis by ABT-737 + roscovitine,
whereas cells overexpressing Bcl-2 or Bcl-xL remain fully
sensitive. Finally, Mcl-1 knockout MEFs are extremely sensitive
to Bak conformational change and apoptosis induced by
ABT-737, effects that are not potentiated by roscovitine.
Collectively, these findings suggest down-regulation of Mcl-1
by either CDK inhibitors or genetic approaches dramatically
potentiate ABT-737 lethality through cooperative interactions
at two distinct levels: unleashing of Bak from both Bcl-xL and
Mcl-1 and simultaneous induction of Bak activation and Bax
translocation. These findings provide a mechanistic basis for
simultaneously targeting Mcl-1 and Bcl-2/Bcl-xL in leukemia.
[Cancer Res 2007;67(2):782–91]
Introduction
Cell death decisions are regulated by the complex interplay
between two groups of Bcl-2 family members: proapoptotic
proteins (e.g., multidomain: Bax and Bak; BH3-only: Bad, Bim,
Bid, and Noxa) and antiapoptotic proteins (e.g., Bcl-2, Bcl-xL,
Bcl-w, Mcl-1, and Bfl-1/A1; refs. 1, 2). In disorders such as
leukemia, increased expression of antiapoptotic proteins, such as
Bcl-2, is required for disease maintenance (3), confers drug
resistance (4), and is associated with poor clinical outcome (5).
These observations have prompted the development of small-
molecule Bcl-2 inhibitors (refs. 6, 7; e.g., HA14-1) that disable Bcl-2,
resulting in induction of apoptosis in leukemia cell lines (8).
Alternative strategies include the use of antisense oligodeoxynu-
cleotides (e.g., G3139; ref. 6) or stabilized forms of BH3 peptides (9),
among others. Recently, a novel inhibitor (ABT-737) of Bcl-2,
Bcl-xL, and Bcl-w, which is significantly more potent than previous
compounds of this type, has been developed. This compound acts
by mimicking the capacity of the BH3-only protein Bad to dock to
the hydrophobic groove of antiapoptotic Bcl-2 family proteins,
thereby diminishing their ability to antagonize apoptosis (10).
ABT-737 lowers the apoptotic threshold for chemotherapeutic
agents or ionizing radiation and has shown impressive precli-
nical activity against hematopoietic malignancies as well as solid
tumors in vitro and in vivo (10). Recently, ABT-737 was shown to
overcome drug resistance (e.g., toward imatinib) in Bcr/Abl
+
leukemic cells (11). However, ABT-737 has a low affinity for other
antiapoptotic Bcl-2 family proteins (e.g., Mcl-1 and A1; ref. 10)
and thus may exhibit limited cytotoxic effects in cells with high
endogenous levels of Mcl-1 (12). Moreover, ABT-737 efficiently kills
interleukin-3 (IL-3)–dependent cells (e.g., FL5.12) only after IL-3
withdrawal (13), suggesting that additional death signals may be
required for lethality. Notably, Mcl-1 is a highly expressed anti-
apoptotic protein (14) and a critical survival factor for various
malignant hematopoietic cells (14, 15). Recent evidence suggests
that more than one antiapoptotic protein (e.g., Mcl-1 and Bcl-xL)
cooperate to sequester multidomain proapoptotic proteins, such
as Bak, thereby preventing its activation (16). Thus, the impaired
capacity of ABT-737 to induce apoptosis in tumor cells expressing
high Mcl-1 levels may stem from a requirement for inhibition
of multiple antiapoptotic proteins. A corollary is that down-
regulation/inhibition of ABT-737–nontargeted proteins (e.g., Mcl-1
or A1) may enhance the lethality of this compound (12).
One candidate strategy to down-regulate/inhibit Mcl-1 involves
the use of cyclin-dependent kinase (CDK) inhibitors. In preclinical
studies, CDK inhibitors, including flavopiridol and the roscovitine
derivative CYC202 (seliciclib), are potent inducers of apoptosis in
malignant hematopoietic cells, including leukemia cells (17, 18).
Notably, results from several laboratories have established that
CDK inhibitors (e.g., flavopiridol and CYC202) act, at least in part,
by inhibiting CDK9, a kinase intimately involved in transcription
initiation and elongation through activation of the positive
transcription elongation factor-b, resulting in down-regulation of
several short-lived proteins, including Mcl-1 (17, 19).
Here, we report that Mcl-1 down-regulation by either CDK
inhibitors or a small hairpin RNA (shRNA) approach leads to a
dramatic increase in ABT-737–mediated apoptosis in human
leukemia cells. Our results also indicate that this phenomenon
stems from a mechanism involving two levels of cooperation
between antiapoptotic and multidomain proapoptotic proteins of
the Bcl-2 family: (a) simultaneous untethering of Bak from Bcl-xL
(by ABT-737) and Mcl-1 (e.g., by roscovitine) and (b) the resulting
Note: S. Chen and Y. Dai contributed equally to this work.
Requests for reprints: Steven Grant, Division of Hematology/Oncology, Virginia
Commonwealth University/Massey Cancer Center, MCV Station Box 230, Richmond,
VA 23298. Phone: 804-828-5211; Fax: 804-828-2178; E-mail: stgrant@vcu.edu.
I2007 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-06-3964
Cancer Res 2007; 67: (2). January 15, 2007
782
www.aacrjournals.org
Research Article
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
activation of both Bak and Bax, culminating in Bax mitochondrial
translocation and engagement of the apoptotic cascade. These
findings may provide a theoretical framework for combinatorial
approaches that target diverse antiapoptotic proteins that
cooperate in the efficient induction of apoptosis in malignant cells.
Materials and Methods
Cells and reagents. Human leukemia U937, HL-60, and Jurkat cells were
provided by the American Type Culture Collection (Rockville, MD) and
maintained in RPMI 1640 containing 10% FCS as previously reported (20).
U937/Bcl-2 and U937/Bcl-xL were obtained by stable transfection of cells
with full-length Bcl-2 and Bcl-xL cDNA, respectively (21). U937 cells stably
overexpressing Mcl-1 were kindly provided by Dr. Ruth Craig (Dartmouth
Medical School, Hanover, NH; ref. 22). All experiments used logarithmically
growing cells (3–5
10
5
cells/mL). Peripheral blood samples were obtained
with informed consent according to the Declaration of Helsinki from the
peripheral blood of three patients with acute myeloblastic leukemia (AML;
FAB subtype M2) undergoing routine diagnostic aspirations with approval
from the Virginia Commonwealth University Institutional Review Board.
Leukemic blasts were isolated as previously described (20). Wild-type (wt),
Bax
/
, Bak
/
, and Bax
/
/Bak
/
(double knockout) mouse embryonic
fibroblast (MEF) were kindly provided by the laboratory of Dr. Stanley
Korsmeyer (Dana-Farber Cancer Institute, Boston, MA; ref. 23). Mcl-1
/
MEFs were kindly provided by Dr. Joseph Opferman (St. Jude Children’s
Research Hospital, Memphis, TN). All experiments were initiated in cells
cultured at f60% confluence.
ABT-737 was kindly provided by Dr. Gary Gordon (Abbott Laboratories,
Abbott Park, IL; ref. 10). It was dissolved in DMSO, aliquoted, and stored at
80jC. Roscovitine and R-roscovitine were purchased from Calbiochem
(San Diego, CA), dissolved in sterile DMSO, aliquoted, and stored at 20jC.
In all experiments, the final concentration of DMSO did not exceed 0.1%.
Assessment of apoptosis. The extent of apoptosis was evaluated by flow
cytometric analysis using Annexin V-FITC staining as described previously
(20). To analyze the extent of cell death in MEFs, cells were trypsinized and
harvested together with those in the culture supernatant and then stained
with 5 Ag/mL 7-amino-actinomycin D (7AAD; Sigma, St. Louis, MO) in PBS
for 20 min at 37jC. The percentage of dead cells (7AAD
+
) was assessed by
flow cytometry using a Becton Dickinson FACScan (Becton Dickinson, San
Jose, CA).
Immunoblot. For subcellular fractionation, cells were lysed in digitonin
lysis buffer (20). After centrifugation, the supernatant (S-100, cytosolic
fraction) and pellets (organelle/membrane fractions) were collected and
subjected to immunoblot. Samples (30 Ag protein for each condition) from
whole-cell pellets or subcellular fractions were subjected to immunoblot
following procedures previously described (20). Where indicated, the blots
were reprobed with antibodies against h-actin (Sigma) or a-tubulin
(Oncogene, La Jolla, CA) to ensure equal loading and transfer of proteins.
The following antibodies were used as primary antibodies: anti-Mcl-1, anti-
caspase-9, and anti-caspase-3 (BD PharMingen, San Diego, CA); anti-Mcl-1,
anti-Bcl-2, anti-Bcl-xS/L, anti–cytochrome c, anti-AIF, anti-Bak, and anti-
Bax (Santa Cruz Biotechnology, Santa Cruz, CA); anti–cleaved caspase-3
(Asp
175
), anti–cleaved poly(ADP-ribose) polymerase (PARP; Asp
214
), and
anti-Bcl-xL (Cell Signaling, Beverly, MA); anti–RNA polymerase II (pol II)
and anti–phospho-pol II (Upstate, Lake Placid, NY); anti-human Bcl-2 onco-
protein (DAKO, Carpinteria, CA); anti-caspase-8 (Alexis, San Diego, CA);
anti-PARP (Biomol, Plymouth Meeting, PA).
Immunoprecipitation. The interaction between Bak and Mcl-1 or Bcl-
xL was evaluated by coimmunoprecipitation analysis by using ExactaCruz
kits (Santa Cruz Biotechnology) as per manufacturer’s instructions. For
these studies, CHAPS buffer was employed to avoid artifactual associations
reported with other buffers (24). Briefly, cells were lysed by syringing with a
23-gauge needle in lysis buffer [20 mmol/L Tris (pH 7.4), 135 mmol/L NaCl,
1.5 mmol/L MgCl
2
, 1 mmol/L EDTA, 10% glycerol] containing 1% CHAPS
(Pierce, Rockford, IL); 800 Ag of protein per condition was used for
immunoprecipitation with anti-Bak antibody (Santa Cruz Biotechnology),
and the immunoprecipitated protein was subjected to immunoblot analysis
using antibodies to either Mcl-1 (BD PharMingen) or Bcl-xL (Cell Signaling).
Analysis of Bak and Bax conformational change. Cells were fixed and
permeabilized using FIX & PERM Cell Permeabilization Reagents (Caltag
Lab, Burlingame, CA) as per manufacturer’s instructions. Fixed cells were
incubated with either anti-Bak (Ab-1 for U937 cells and Ab-2 for MEFs,
Calbiochem) or anti-Bax (clone 3 for U937 cells, BD Transduction Lab,
Lexington, KY; YTH-6A7 for MEFs, Trevigen, Gaithersburg, MD) on ice for
30 min and then with FITC-conjugated goat-anti-mouse IgG (Southern
Biotech, Birmingham, AL) for 30 min in the dark. After washing, the
samples were analyzed by flow cytometry. The results for each condition
were calibrated by values for cells stained with mouse IgG (Southern
Biotech) as the primary antibody. Values for untreated controls were
arbitrarily set to 100%. In parallel, cells for each condition were stained with
antibodies to total Bak (Santa Cruz Biotechnology) for comparison.
RNA interference. The pSUPER.retro.puro vector containing the human
H1 RNA promoter for expressing shRNA was obtained from Oligoengine
(Seattle, WA). DdRNAi oligonucleotides (5-GATCCCCGCGGGACTGGC-
TAGTTAAACTTCAAGAGAGTTTAACTAGCCAGTCCCGTTTTTA-3; ref. 25)
were cloned into pSUPER.retro.puro vector (pSUPER/shMcl-1). U937 cells
were transiently transfected with the pSUPER/shMcl-1 construct and its
empty vector by using the Amaxa Nucleofector device (program V-001) with
Kit V (Amaxa GmbH, Cologne, Germany) as per manufacturer’s instructions.
Reverse transcription-PCR. Total RNA was isolated using RNeasy Mini
kit with QIAshredder spin column (Qiagen, Valencia, CA) as per
manufacturer’s instructions; 1 Ag per condition of total RNA was subjected
to reverse transcription-PCR (RT-PCR) reaction using One-Step RT-PCR kit
(Qiagen) and Thermal Cycler (MJ Research, Inc., Reno, NV). The primers
forward, 5-ATCTCTCGGTACCTTCGGGAGC-3 and reverse, 5-CCTGATGC-
CACCTTCTAGGTCC-3 (26) were used for Mcl-1. PCR products of Mcl-1
(442 bp) were analyzed in 2% agarose gel with ethidium bromide staining.
Statistical analysis. The values represent the means F SD for at least
three independent experiments done in triplicate. The significance of
differences between experimental variables was determined using the
Student’s t test. Analysis of synergism was done according to median dose-
effect analysis using Calcusyn software (Biosoft, Ferguson, MO; ref. 20).
Results
CDK inhibitors transcriptionally down-regulate the expres-
sion of Mcl-1 and synergistically interact with ABT-737 to
induce apoptosis in U937 cells. Earlier reports indicated that
various CDK inhibitors, including roscovitine (27), transcriptionally
down-regulate expression of short-lived proteins, such as Mcl-1.
As shown in Fig. 1A, treatment with either CDK inhibitor at
concentrations >10 Amol/L resulted in significant declines in Mcl-1
protein levels, but no change in expression of Bcl-2 and Bcl-xL was
observed. This phenomenon was associated with inhibition of
phosphorylation of pol II CTD and a pronounced reduction in
Mcl-1 mRNA levels (Fig. 1B).
Attempts were then made to determine what effect CDK
inhibitors would have on the response of cells to ABT-737. Whereas
exposure to 12 Amol/L roscovitine alone modestly increased
apoptosis, ABT-737 at a concentration range of 150 to 750 nmol/L
had very little effect (Fig. 1C). However, combined treatment
resulted in a dramatic increase in the loss of mitochondrial
membrane potential (DW
m
; data not shown) and apoptosis.
Identical results were obtained with R-roscovitine. Moreover, lower
concentrations (V10 Amol/L) of either roscovitine or R-roscovitine,
which failed to down-regulate Mcl-1 (Fig. 1A), did not enhance
ABT-737 lethality (data not shown). Median dose-effect analysis,
employing apoptosis determined by Annexin V-FITC as an end
point, yielded combination index values <1.0 (Fig. 1C, inset),
denoting synergistic interactions.
Mcl-1 Down-regulation Enhances ABT-737 Lethality
www.aacrjournals.org
783
Cancer Res 2007; 67: (2). January 15, 2007
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Exposure of U937 cells to roscovitine F ABT-737 resulted in
marked down-regulation of Mcl-1 protein, whereas ABT-737 by itself
failed to modify Mcl-1 expression (Fig. 1D). In contrast, no change in
Bcl-2 or Bcl-xL expression was observed (data not shown). Lastly,
effects of cotreatment with ABT-737 and roscovitine were examined
in relation to mitochondrial events. Cotreatment with roscovitine
and ABT-737 triggered a pronounced increase in cytochrome c and
AIF release into the cytosolic fraction (Fig. 1D). Combined treat-
ment also induced a modest but discernible increase in caspase-9
and caspase-8 cleavage, and a marked increase in cleavage of
caspase-3, accompanied by cleavage of PARP (Fig. 1D ). However,
transfection with a dominant-negative caspase-8 construct (21)
failed to protect U937 cells from mitochondrial damage and
apoptosis induced by ABT-737/roscovitine (data not shown),
arguing against involvement of the extrinsic apoptotic pathway.
Together, these findings indicate that CDK inhibitors dramatically
increase ABT-737–mediated apoptosis in association with Mcl-1
down-regulation.
Roscovitine markedly down-regulates Mcl-1 and increases
ABT-737 lethality in various human leukemia cells, including
primary AML cells. Parallel studies were done in human HL-60
promyelocytic and Jurkat lymphoblastic leukemia cells. First, these
leukemia cells exhibited differing susceptibilities to ABT-737–
mediated lethality (IC
50
:1.3Amol/L for U937, 420 nmol/L for
Jurkat, 190 nmol/L for HL-60). Immunoblot showed that Bcl-2 and
Mcl-1 protein levels varied between the cell lines, whereas Bcl-xL
expression was equivalent. Interestingly, HL-60 cells, which were
the most sensitive of the three lines, exhibited very low Mcl-1
expression but higher levels of Bcl-2 (Fig. 2A, inset). These results
suggest that levels of Mcl-1 and/or the ratio between Mcl-1 and
Figure 1. CDK inhibitors interact with ABT-737 to induce mitochondria-related apoptotic signaling events in association with down-regulation of Mcl-1. A, U937 cells
were exposed to 7.5 to 15 Amol/L of either roscovitine (Rosc)orR-roscovitine (R-rosc ), after which immunoblot analysis was done to monitor expression of Bcl-2,
Bcl-xL, and Mcl-1. B, immunoblot analysis was conducted to assess phosphorylation status of pol II (P-pol II) CTD after 24 h of treatment with roscovitine (12 A mol/L).
Alternatively, total RNA was extracted, and RT-PCR was done to monitor Mcl-1 mRNA levels. C, U937 cells were treated with 150 to 750 nmol/L ABT-737
(ABT) F 12 Amol/L roscovitine or 300 nmol/L ABT-737 F 12 Amol/L R-roscovitine for 24 h, after which flow cytometry was conducted to determine the percentage of
apoptotic cells (Annexin V
+
). Moreover, U937 cells were exposed (24 h) to varying concentrations of ABT-737 and roscovitine alone and in combination at fixed ratio
1:100. At the end of this period, the percentage of Annexin V
+
cells was determined for each condition, and median dose-effect analysis was then employed to
characterize the nature of the interaction between these agents (inset). Combination Index (C.I.) values <1.0 denote a synergistic interaction. Representative for three
separate experiments. D, U937 cells were exposed (24 h) to 150 to 500 nmol/L ABT-737 F 12 Amol/L roscovitine, and immunoblot was then done to evaluate
expression of Mcl-1 as well as cleavage of caspases (casp ) and PARP. Alternatively, S-100 cytosolic fractions were prepared and subjected to immunoblot analysis.
Primary antibodies were employed as indicated. CF, cleavage fragment; Cyt c, cytochrome c. A, B, and D, representative results from one experiment, and two
additional studies yielded equivalent results.
Cancer Research
Cancer Res 2007; 67: (2). January 15, 2007
784
www.aacrjournals.org
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Bcl-2 expression may represent as determinants of ABT-737
sensitivity in leukemia cells.
Cotreatment with marginally toxic concentrations of ABT-737
and roscovitine markedly induced mitochondrial damage (data not
shown) and apoptosis in Jurkat and HL-60 cells (Fig. 2A). Roughly
equivalent results were obtained when R -roscovitine was coad-
ministered (data not shown). Moreover, roscovitine, administrated
alone F ABT-737 dramatically down-regulated Mcl-1 in Jurkat cells,
and combined treatment marked increased PARP cleavage (Fig. 2B).
Effects of this regimen on primary leukemia blasts isolated from
three patients with AML (Fig. 2C) were similar to those obtained in
leukemia cell lines. Furthermore, roscovitine F ABT-737 almost
completely abrogated Mcl-1 expression in AML blasts and induced
pronounced PARP (Fig. 2D), indicating that ABT-737/roscovitine
interactions occur in association with Mcl-1 down-regulation in
both continuously cultured human leukemia cell lines differing in
their sensitivity to ABT-737 as well as in primary AML blasts.
Roscovitine enhances ABT-737–mediated Bax conforma-
tional change, whereas Bak activation is induced only by
combined ABT-737 and roscovitine administration. Bax and
Bak are essential for mitochondrial outer membrane permeabiliza-
tion (28, 29), a critical cell death determinant (30). Whereas Bak is
normally localized at its site of action (i.e., organellar membranes),
Bax is monomeric and found in the cytosol of healthy cells.
Following death stimuli, Bax undergoes conformational change
and translocates to organellar membranes. Activation of both Bak
and Bax is associated with a conformational change, which can be
detected by antibodies recognizing only the active protein
conformers (16). The effect of ABT-737 F roscovitine on Bak/Bax
conformational change was then examined. When U937 cells were
exposed to ABT-737 (150–500 nmol/L) alone, a clear dose-
dependent increase in Bax conformational change was observed
(Fig. 3A). Interestingly, Bax conformational change was unaccom-
panied by Bax translocation (see below; Fig. 3C) nor did it induce
Figure 2. Roscovitine down-regulates Mcl-1 and promotes ABT-737 lethality in multiple human leukemia cell lines and primary AML cells. A, untreated U937, HL-60,
and Jurkat cells were lysed and subjected to immunoblot analysis to detect protein levels of Bcl-2, Bcl-xL, and Mcl-1 (inset ). Jurkat and HL-60 cells were exposed for
24 h to ABT-737 (Jurkat, 100–200 nmol/L; HL-60, 30–50 nmol/L) F 12 Amol/L roscovitine, after which the percentage of Annexin V
+
cells was determined by flow
cytometry. B, Jurkat cells were treated as described in (A), after which immunoblot analysis was done to monitor Mcl-1 expression and PARP cleavage. C, blasts were
isolated from the peripheral blood of three AML patients (designed as #1–3; FAB subtype M2) and then incubated (24 h) with 150 nmol/L ABT-737 F roscovitine
(#1 and #2, 10 Amol/L; #3, 12 Amol/L). At the end of this period, the percentage of Annexin V
+
cells was determined by flow cytometry. Columns, means for an
experiment done in triplicate; bars, SD. Representative experiment (patient #3). D, the blasts (patient #3 ) were incubated (24 h) with 150 and 300 nmol/L
ABT-737 F 12 Amol/L roscovitine and then lysed for immunoblot using indicated primary antibodies. C-PARP, cleaved PARP. A (inset), B , and D, representative for
three separate experiments.
Mcl-1 Down-regulation Enhances ABT-737 Lethality
www.aacrjournals.org
785
Cancer Res 2007; 67: (2). January 15, 2007
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
apoptosis (Fig. 1C). On the other hand, roscovitine (12 Amol/L)
alone minimally induced Bax conformational change. Notably, cells
coexposed to roscovitine and ABT-737 displayed a further increase
in Bax conformational change compared with cells treated ABT-737
alone (Fig. 3A), an effect accompanied by marked increases in Bax
translocation (see below; Fig. 3C ) and lethality. These findings
suggest that ABT-737–induced Bax conformational change by itself
may not be sufficient to trigger Bax translocation and apoptosis.
Parallel studies were then done in U937 cells to assess Bak
conformational change. In sharp contrast to results involving
Bax, ABT-737 by itself had little or no effect on Bak activation
(Fig. 3A), whereas roscovitine also failed to induce Bak activation.
However, combined treatment with ABT-737 and roscovitine
induced pronounced Bak activation. No change in total Bak was
noted for any condition (data not shown). Together, these results
indicate that (a) roscovitine enhances ABT-737–mediated Bax
conformational change and cooperates to trigger Bax transloca-
tion, and that (b) ABT-737 alone is ineffective in triggering Bak
conformational change; instead, roscovitine coadministration is
required for ABT-737–mediated Bak activation.
Coadministration of ABT-737 and roscovitine disrupts the
association of Bak with both Bcl-xL and Mcl-1 and induces Bax
translocation. One of the mechanisms by which Mcl-1 opposes
apoptosis is by binding/sequestering Bak and preventing its
activation (31). Furthermore, there is evidence that Bcl-xL, but
not Bcl-2, Bcl-w, or A1, acts analogously to block Bak activation
(16), and that both Mcl-1 and Bcl-xL must be neutralized (e.g., by
Noxa and Bad, respectively) to displace Bak in order to trigger cell
death (16, 32). Consequently, the effects of ABT-737 and roscovitine
on interactions between Bak and Bcl-xL or Mcl-1 were assessed.
No change in total Bak levels was observed with any treatment
(see below; Fig. 3C). U937 cells exposed to ABT-737 F roscovitine
were lysed in CHAPS buffer, and associations between Bak and
Mcl-1 or Bcl-xL were assessed (Fig. 3B). Treatment with ABT-737
Figure 3. Bax is necessary for induction of cell death by both ABT-737 alone and in combination with roscovitine, whereas Bak activation is only required for synergistic
interactions between these agents. A, U937 cells were treated (24 h) with 150 to 500 nmol/L ABT-737 F 12 Amol/L roscovitine, after which cells were stained with
anti–conformationally changed Bax (clone 3)/FITC-conjugated goat-anti-mouse IgG and subjected to flow cytometry. In parallel, Bak activation was monitored by
flow cytometry after staining with anti–conformationally changed Bak (Ab-1)/FITC-conjugated goat-anti-mouse IgG. Representative histograms (dotted, untreated
controls). Values, mean FSD for three separate experiments done in triplicate. B, U937 cells were exposed for 24 h to 150 to 300 nmol/L ABT-737 F 12 Amol/L
roscovitine, after which cells were lysed in CHAPS buffer and subjected to immunoprecipitation (IP) using anti-Bak and then immunoblotted (IB) with either anti-Mcl-1 or
anti-Bcl-xL antibody. For comparison, the right lanes (designed as C) were loaded with whole-cell lysates. C, U937 cells were treated as described in (A ), after
which S-100 cytosolic and pelleted organellar membrane fractions were prepared and subjected to immunoblot analysis using an anti-Bax antibody. Alternatively, total
levels of Bax and Bak were monitored in whole-cell lysates. D, untreated wt, Bak
/
, Bax
/
, and Bax
/
/Bak
/
[double knockout (DKO)] MEFs were lysed and
subjected to immunoblot to detect expression of both proapoptotic (Bak and Bax) and antiapoptotic (Mcl-1, Bcl-2, and Bcl-xL) proteins (inset). Various MEFs were
exposed (24 h) to 300 to 500 nmol/L ABT-737 F 12 Amol/L roscovitine, after which the cells, including those in the culture supernatant, were collected for each condition.
Cells were then stained with 7AAD and subjected to flow cytometry to determine the percentage of 7AAD
+
cells. *, P < 0.01, significantly greater than values for
treatment with each concentration of ABT-737 alone. B, C, and D (inset), representative results from one experiment, and two additional studies yielded equivalent
results.
Cancer Research
Cancer Res 2007; 67: (2). January 15, 2007
786
www.aacrjournals.org
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
alone modestly but discernibly increased the amount of Mcl-1
associating with Bak. Notably, this effect was largely abrogated by
roscovitine treatment, presumably due to Mcl-1 down-regulation
(Fig. 1). Reciprocal effects were noted when the amount of Bcl-xL
coimmunoprecipitating with Bak was monitored (i.e., roscovitine
substantially increased the amount of Bcl-xL associating with Bak,
whereas ABT-737 essentially reversed this phenomenon). Together,
these findings suggest that cotreatment with roscovitine and ABT-
737 antagonizes interactions of Bak with both Mcl-1 and Bcl-xL.
Lastly, the effect of coexposure to roscovitine and ABT-737 on
intracellular Bax localization was examined. Treatment with either
ABT-737 or roscovitine alone had little or no effect on the
intracellular disposition of Bax (Fig. 3C). However, ABT-737/
roscovitine coadministration induced a major translocation of
Bax from the cytosolic compartment to the pelleted organellar
membrane fraction. This suggests that roscovitine and ABT-737
cooperate to untether Bak from Mcl-1 and Bcl-xL, leading to Bak
activation and accompanying Bax translocation to organellar
membranes. These findings also suggest that concomitant
activation of Bax and Bak may be responsible for the dramatic
induction of apoptosis in cells coexposed to roscovitine and
ABT-737.
Bax knockout in MEFs substantially diminishes the lethality
of ABT-737 F roscovitine, whereas Bak knockout primarily
blocks synergistic interactions between these agents. To
evaluate further the functional roles of Bax and Bak in the lethality
of ABT-737 and its interactions with roscovitine, Bax
/
, Bak
/
,
and Bax
/
/Bak
/
(double knockout) MEFs were employed
(Fig. 3D, inset). Immunoblot analysis revealed that antiapoptotic
Bcl-2 family protein levels (e.g., Mcl-1, Bcl-2, and Bcl-xL) were
roughly equivalent in each of the cell types. Coadministration of
roscovitine clearly increased the lethality of ABT-737 in wt MEFs
(P < 0.01, compared with cells exposed to ABT-737 alone; Fig. 3D).
However, the lethality of ABT-737 F roscovitine was substantially
blunted in Bax
/
MEFs, indicating that Bax is critical for this
phenomenon. In marked contrast, ABT-737 was still able to induce
cell death in Bak
/
MEFs; in fact, these cells were slightly more
sensitive than wt cells. Significantly, coadministration of roscovi-
tine failed to increase the lethality of ABT-737 in Bak
/
MEFs.
Finally, Bax/Bak double knockout MEFs displayed essentially no
response to any of these treatments. Together, these findings sug-
gest that Bax is required for induction of cell death by both ABT-
737 F roscovitine, whereas Bak activation, although dispensable
for ABT-737 lethality, is nevertheless required for ABT-737/rosco-
vitine synergism. They also support the notion that cooperation
between Bak and Bax is critical for ABT-737/roscovitine lethality.
Roscovitine down-regulates Mcl-1 expression and attenu-
ates ABT-737 resistance in leukemic cells ectopically express-
ing Bcl-2 or Bcl-xL. Because ABT-737 targets Bcl-2 and Bcl-xL (10),
it might be predicted that the relative abundance of antiapoptotic
proteins, such as Bcl-2, would be related to ABT-737 sensitivity.
Consequently, the effect of Bcl-2 or Bcl-xL expression on the
susceptibility of cells to ABT-737 F roscovitine was examined using
U937 cells ectopically expressing Bcl-2 or Bcl-xL. Ectopic expres-
sion of Bcl-2 or Bcl-xL provided significant protection from the
lethal effects of etoposide (VP-16), a potent inhibitor of DNA
topoisomerase (Fig. 4A and B). Notably, Bcl-2 or Bcl-xL over-
expression attenuated ABT-737–mediated lethality but did not
affect roscovitine cytotoxicity. However, Bcl-2 or Bcl-xL over-
expression failed to protect cells from mitochondrial damage
(i.e., loss of DW
m
; data not shown) and apoptosis induced by
roscovitine and ABT-737 coadministration (P > 0.05, for each ABT-
737 concentration, compared with empty vector controls U937/
pCEP or U937/3.1). Cotreatment with roscovitine/ABT-737 induced
an equivalent decline in Mcl-1 expression and enhanced PARP
cleavage in U937/Bcl-2, U937/Bcl-xL, and controls but did not
modify Bcl-2 or Bcl-xL expression (Fig. 4C and D). Collectively,
these findings indicate that although ectopic expression of either
Bcl-2 or Bcl-xL reduces human leukemia cell sensitivity to ABT-737,
they are unable to prevent the ABT-737/roscovitine regimen from
diminishing Mcl-1 levels and inducing apoptosis.
Ectopic expression of Mcl-1 but not Bcl-2 blocks Bak
activation and apoptosis triggered by combined exposure of
leukemia cells to ABT-737 and roscovitine. Because Bcl-2/Bcl-xL
and Mcl-1 may play disparate functional roles in blocking apoptosis
(32), studies were done employing U937 cells ectopically expressing
Mcl-1 (22). As in cells overexpressing Bcl-2 or Bcl-xL, ectopic
expression of Mcl-1 substantially protected cells from VP-16– and
ABT-737–mediated lethality (Fig. 5A). However, in striking contrast
to the former cells, enforced Mcl-1 expression significantly
diminished mitochondrial damage (loss of DW
m
; data not shown)
and apoptosis (Fig. 5A) induced by the ABT-737/roscovitine
regimen (P < 0.001, versus U937/pCEP).
Immunoblot revealed that combined treatment with ABT-737/
roscovitine clearly diminished Mcl-1 expression in U937/pCEP cells
but failed to do so in their U937/Mcl-1 counterparts (Fig. 5B ).
Moreover, PARP cleavage induced by ABT-737/roscovitine was
almost completely abrogated in U937/Mcl-1 cells. Basal Bcl-2
expression was somewhat lower in U937/Mcl-1 cells compared
with controls but was not modified appreciably in either line
with any treatment. Together, these findings argue strongly that
down-regulation of Mcl-1 plays a functional role in potentiation
of ABT-737–mediated lethality by roscovitine.
Finally, the effects of ectopic expression of Bcl-2 or Mcl-1 were
compared with respect to conformational change of Bax and Bak
induced by the ABT-737/roscovitine regimen. Consistent with their
inability to block ABT-737/roscovitine–mediated lethality, Bcl-2
overexpression failed to attenuate conformational change of Bax or
Bak in cells exposed to ABT-737 and roscovitine in combination
(Fig. 5C), although it did reduce Bax conformational change
induced by ABT-737 alone (data not shown). Similar results were
obtained in cells ectopically expressing Bcl-xL (data not shown).
However, in sharp contrast, ectopic expression of Mcl-1, which also
attenuated Bax conformational change mediated by ABT-737 alone
(data not shown), essentially abrogated Bak activation triggered by
the ABT-737/roscovitine regimen and also partially reduced Bax
conformational change after exposure to the combination (Fig. 5D).
These findings provide further evidence that disruption of Mcl-1
function plays a critical role in ABT-737/ roscovitine interactions
associated with Bax/Bak activation and apoptosis.
RNA interference or gene knockout of Mcl-1 dramatically
sensitizes cells to ABT-737 but abrogates the capacity of
roscovitine to potentiate Bak activation and lethality. To
evaluate further the functional role of Mcl-1 in Bax/ Bak activation
as well as apoptosis mediated by ABT-737 F roscovitine, a shRNA
strategy and Mcl-1
/
MEFs were employed. First, U937 cells were
transiently transfected with a construct encoding shRNA against
Mcl-1 mRNA, and immunoblot analysis documented Mcl-1 down-
regulation (Fig. 6A, inset). Mcl-1 down-regulation by this approach
dramatically sensitized human leukemia cells to ABT-737 lethality
(P < 0.02–0.001, for each ABT-737 concentration, compared with
those transfected with empty vector; Fig. 6A ).
Mcl-1 Down-regulation Enhances ABT-737 Lethality
www.aacrjournals.org
787
Cancer Res 2007; 67: (2). January 15, 2007
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Further studies were done in wt and Mcl-1
/
MEFs. Immuno-
blots revealed that levels of Bcl-2, Bcl-xL, Bak, and Bax in Mcl-1
/
cells were roughly equivalent to those in wt MEFs, whereas Mcl-1
was absent (Fig. 6B, inset ). Mcl-1
/
cells exhibited a dramatic
increase in sensitivity to ABT-737 (50–100 nmol/L) compared with
wt cells (P < 0.001; Fig. 6B). Significantly, roscovitine was unable
to enhance cell killing mediated by ABT-737 in Mcl-1
/
cells
(P > 0.05). Moreover, Bak conformational change was observed only
after coexposure of wt MEFs to ABT-737 and roscovitine, consistent
with preceding findings in human leukemia cells. In striking
contrast, ABT-737 alone dramatically induced Bak activation in
Mcl-1
/
cells, a phenomenon that was not further enhanced by
roscovitine (Fig. 6C). Analogously, Mcl-1
/
cells displayed greater
Bax conformational change following ABT-737 exposure compared
with their wt counterparts (Fig. 6C). However, as in the case of Bak,
roscovitine failed to increase ABT-737–mediated Bax conforma-
tional change in Mcl-1
/
cells. Together, these results provide clear
evidence that Mcl-1 is a critical determinant of both ABT-737
actions as well as the capacity of roscovitine to potentiate ABT-737
lethality through cooperative activation of Bak and Bax.
Discussion
ABT-737, a novel small-molecule Bcl-2/Bcl-xL/Bcl-w inhibitor
currently in development as an anticancer agent, has a relatively
low affinity for the more divergent antiapoptotic Bcl-2 family
proteins (e.g., Mcl-1 and A1; ref. 10). ABT-737 is less efficient in
killing tumor cells exhibiting relatively high levels of Mcl-1 (12).
Because Mcl-1 has a short half-life (i.e., <2 h; ref. 33), it is
particularly susceptible to down-regulation by agents that disrupt
its de novo synthesis. Consequently, attention has recently focused
on the capacity of several clinical relevant CDK inhibitors (e.g.,
flavopiridol and the roscovitine derivative CYC202) to transcrip-
tionally down-regulate Mcl-1 through CDK9 inhibition (17, 19).
Moreover, we previously reported that flavopiridol enhanced the
lethality of HA14-1, although the mechanism underlying this
interaction was not determined (34). Consistent with these
findings, roscovitine and its R enantiomer R-roscovitine inhibited
phosphorylation of RNA polymerase II CTD, diminished Mcl-1
mRNA levels, and markedly down-regulated protein levels.
Significantly, such actions correlated closely with synergistic
interactions with ABT-737. Although the possibility that other
Figure 4. Overexpression of Bcl-2 or Bcl-xL fails to protect cells from roscovitine/ABT-737–mediated lethality. A and B, U937/Bcl-2 (A) and U937/Bcl-xL (B ), as well
as their empty-vector controls (U937/pCEP and U937/pcDNA3.1), were treated (24 h) with 150 to 500 nmol/L ABT-737 F 12 Amol/L roscovitine or 25 Amol/L VP-16 (6 h)
for comparison, after which the percentage of Annexin V
+
cells was assessed by flow cytometry. **, P < 0.001, significantly less than values for empty vector controls.
C, U937/Bcl-2 and U937/pCEP cells were treated as described in (A), after which cells were lysed and subjected to immunoblot for expression of Bcl-2 and Mcl-1 as
well as PARP cleavage. D, U937/Bcl-xL and U937/pcDNA3.1 cells were exposed for 24 h to ABT-737 (top, 300 nmol/L; bottom, 150–500 nmol/L) F 12 Amol/L
roscovitine, after which immunoblot analysis was done to monitor protein levels of Bcl-xL and Mcl-1 as well as PARP cleavage. B and D, representative for three
separate experiments.
Cancer Research
Cancer Res 2007; 67: (2). January 15, 2007
788
www.aacrjournals.org
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
activities (e.g., CDK disruption) contribute to the lethality of
combination regimens involving CDK inhibitors cannot be com-
pletely excluded (35), it is noteworthy that Mcl-1 down-regulation
plays an important role in apoptosis in malignant hematopoietic
cells (17, 19, 36). On the other hand, in certain transformed cells,
Mcl-1 down-regulation, although required, seems to be incapable
by itself of initiating apoptosis (31, 37), indicating that other
cooperating factors may be necessary for cell death. These findings
suggest that interventions targeting more than one antiapoptotic
protein may be required for optimal cell killing.
Whereas resistance to ABT-737 may reflect the compensatory
actions of Mcl-1 (12), the present investigation was prompted by
recent evidence that that Bak activation requires simultaneous
disruption of its associations with Mcl-1 (e.g., by Noxa) and Bcl-xL
(e.g., by Bad; ref. 16). Moreover, increased production of Noxa can
oppose Mcl-1 antiapoptotic functions, leading to simultaneous
activation of Bax and Bak (38). The present results suggest that
coadministration of ABT-737 and roscovitine recapitulate the
actions of more physiologic proapoptotic BH3-only proteins.
Specifically, ABT-737, by binding to hydrophobic groove within
the Bcl-xL BH3 domain (10), untethers Bak from Bcl-xL, analogous
to the actions of BH3-only proteins such as Bad (39). On the other
hand, Mcl-1 down-regulation by roscovitine, mimicking the actions
of Noxa in displacing Bak from Mcl-1 (16), reciprocally released Bak
from Mcl-1 sequestration. Thus, coadministration of ABT-737 and
roscovitine markedly diminished the association of Bak with both
Bcl-xL and Mcl-1, inducing Bak activation. In support of this
hypothesis, Bak activation was observed only when roscovitine
and ABT-737 were administrated concomitantly, but not after
ABT-737 alone. The notion of cooperativity in the regulation of
ABT-737 lethality is further supported by the results obtained in
Mcl-1 knockout MEFs, in which ABT-737 alone, in contrast to its
actions in wt cells, markedly induced Bak activation. Significantly,
roscovitine was unable to enhance ABT-737 lethality further in
these cells presumably because Mcl-1 was absent and levels could
not be reduced further. These findings provide strong support for
the concept that disruption of more than one antiapoptotic protein
of the Bcl-2 family (i.e., Mcl-1 and Bcl-2/Bc-xL) represents a highly
potent apoptotic stimulus (12).
The finding that ABT-737 induced Bax conformational change but
did not trigger apoptosis by itself suggests that the lethality of the
ABT-737/roscovitine regimen involves not simply activation of
either Bax or Bak, but cooperativity between these proteins.
Untethering of Bak from both Mcl-1 and Bcl-xL allows Bak confor-
mational change, homo-oligomerization (16), as well as possible
associations with Bax (40). Nevertheless, there is evidence that
Bax and Bak may interact to promote apoptosis (41). Results
obtained with Bax and Bak knockout MEFs are fully compatible with
a model in which Bax and Bak cooperate to trigger cell death. For
example, Bax knockout cells displayed marked resistance to ABT-
737 given alone or in combination with roscovitine. This suggests
that the presence of Bax is essential for both ABT-737 lethality and
synergistic interactions with roscovitine. In striking contrast, Bak
knockout cells remained fully sensitive to ABT-737–mediated cell
Figure 5. Ectopic expression of Mcl-1, but not Bcl-2, diminishes Bak activation and potentiation of apoptosis in cells coexposed to ABT-737 and roscovitine. A, U937/
Mcl-1 and its empty-vector control (U937/pCEP) cells were exposed to 150 to 500 nmol/L ABT-737 F 12 Amol/L roscovitine (24 h) or 25 Amol/L VP-16 (6 h) for
comparison, after which the percentage of Annexin V
+
cells was determined by flow cytometry. **, P < 0.001, significantly less than values for U937/pCEP cells.
B, U937/Mcl-1 and U937/pCEP cells were treated (24 h) with 150 to 500 nmol/L ABT-737 F 12 Amol/L roscovitine, and then immunoblot analysis was done to monitor
expression of Mcl-1 and Bcl-2 as well as PARP cleavage. Representative for three separate experiments. C and D, U937/Bcl-2 (C ) and U937/Mcl-1 (D) cells,
as well as their empty-vector controls (U937/pCEP), were treated for 24 h with 300 nmol/L ABT-737 + 12 Amol/L roscovitine, after which conformational change of Bax
or Bak was monitored by flow cytometry using anti-Bax (clone 3) or anti-Bak (Ab-1) antibody, respectively.
Mcl-1 Down-regulation Enhances ABT-737 Lethality
www.aacrjournals.org
789
Cancer Res 2007; 67: (2). January 15, 2007
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
killing, indicating that Bak is not absolutely required for the lethality
of an agent targeting Bcl-2/Bcl-xL. However, it is significant that
roscovitine failed to potentiate ABT-737 lethality in these cells,
arguing strongly that Bak is required for full engagement of the
apoptotic program following disruption of the Bcl-2/Bcl-xL axis.
Moreover, Bax translocation and cell death occurred only in human
leukemia cells coexposed to ABT-737 and roscovitine (Fig. 3C ).
Although the mechanisms responsible for potentiation of ABT-737–
mediated Bax translocation by roscovitine are presently unclear,
there are several plausible explanations. These include the
possibilities that (a) Mcl-1 may be involved in Bax regulation, either
through a process involving ‘‘activator’’ BH3-only proteins such as
Bim and/or tBid (13, 42–45), or directly by itself (46); or that (b)
activated Bak may promote Bax translocation in an as yet to be
defined way. Collectively, the present results support the notion
that simultaneous interruption of Mcl-1 and Bcl-xL function frees
and activates Bak, which, in the setting of Bax conformational
change, results in Bax translocation, leading in turn to full engage-
ment of the apoptotic machinery.
It is noteworthy that overexpression of Bcl-2 or Bcl-xL failed
to protect leukemia cells from the ABT-737/roscovitine regimen,
reflecting the important contribution of Mcl-1 down-regulation
to the lethality of this regimen. The finding that ectopic expression
of Mcl-1 diminished potentiation of ABT-737 lethality by rosco-
vitine highlights the central role of Mcl-1 down-regulation in
synergism between these agents. This interpretation is further
supported by results showing that roscovitine was unable to
enhance ABT-737–mediated apoptosis in Mcl-1 knockout
MEFs. Moreover, overexpression of Mcl-1, but not Bcl-2/Bcl-xL,
essentially abrogated Bak activation following exposure to ABT-
737/roscovitine, strongly arguing that Mcl-1 plays a major role in
regulating Bak. This concept is consistent with previous findings
indicating that Mcl-1 binds with considerably greater affinity
to Bak compared with Bcl-xL (IC
50
< 10 versus < 100 nmol/L;
ref. 16).
Whereas recent studies suggest that ABT-737 and the more
specific Bcl-xL inhibitor A-385358 increase the antitumor activity
of conventional cytotoxic drugs (10, 47), this phenomenon may
reflect a generic lowering of the apoptotic threshold. On the other
hand, the present results suggest that a mechanism-based
approach combining agents that target distinct antiapoptotic
molecules (e.g., CDK inhibitors that down-regulate Mcl-1 expres-
sion and small-molecule Bcl-2/Bcl-xL inhibitors like ABT-737)
deserve attention. These findings also highlight the importance of
Figure 6. Knockout or down-regulation of Mcl-1 sensitize cells to cell death induced by ABT-737. A, U937 cells were transiently transfected with either empty
vector (EV) or shMcl-1/pSUPER for 24 h, after which cells were lysed and subjected to immunoblot analysis (inset ). After transfection with empty vector or shMcl-1,
cells were incubated for 6 h and then exposed to the indicated concentrations of ABT-737 for an additional 24 h, after which the percentage of viable cells (7AAD
)
was determined by flow cytometry after stained with 7AAD. *, P < 0.02; **, P < 0.001, significantly less than values for cells transfected with empty vector.
B, immunoblot analysis was done to monitor levels of antiapoptotic (Mcl-1, Bcl-2, and Bcl-xL) and multidomain proapoptotic proteins (Bax and Bak) in untreated wt
and Mcl-1 knockout MEFs (inset ). MEFs were exposed to 50 to 100 nmol/L ABT-737 alone or in the presence of 12 Amol/L roscovitine for 24 h, after which cells,
including those in the culture supernatant, were harvested together for flow cytometry using 7AAD staining. **, P < 0.001, significantly greater than values for wt cells.
C, MEFs (wt and Mcl-1
/
) were incubated (24 h) with 100 nmol/L ABT-737 F 12 Amol/L roscovitine, after which cells were stained with anti–conformationally changed
Bak (Ab-2) or Bax (YTH-6A7)/FITC-conjugated goat-anti-mouse IgG and subjected to flow cytometry.
Cancer Research
Cancer Res 2007; 67: (2). January 15, 2007
790
www.aacrjournals.org
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
cooperative interactions between such agents at two separate but
interrelated levels in cell death induction: (a) release of Bak from
both Bcl-xL and Mcl-1 and (b) simultaneous activation of both
Bax and Bak, which may be essential for Bax translocation and
ABT-737 lethality. Whether a strategy combining CDK inhibitors,
or other transcriptional repressors capable of down-regulating
Mcl-1, with Bcl-2/Bcl-xL antagonists will result in enhanced
therapeutic efficacy will depend upon multiple factors, including
the capacity of such agents to diminish Mcl-1 expression in vivo,
and whether the therapeutic index is enhanced. In this context, it
is noteworthy that ABT-737 displays in vivo antitumor selectivity
in preclinical studies (10). In any case, the present findings
suggest that in addition to combining Bcl-2/Bcl-xL antagonists
with conventional cytotoxic drugs, combination strategies involv-
ing targeted agents that down-regulate Mcl-1, a protein that can
compensate for the loss of Bcl-2/Bcl-xL function, represents a
potentially useful alternative approach.
Acknowledgments
Received 10/25/2006; accepted 11/28/2006.
Grant support: National Cancer Institute grants CA63753, CA 93738, and CA
100866; Leukemia and Lymphoma Society of America award 6045-03; V Foundation;
and Department of Defense.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
We thank Dr. Joseph T. Opferman for Mcl-1 knockout MEFs.
Mcl-1 Down-regulation Enhances ABT-737 Lethality
www.aacrjournals.org
791
Cancer Res 2007; 67: (2). January 15, 2007
References
1. Green DR. At the gates of death. Cancer Cell 2006;9:
328–30.
2. Letai A. BCL-2: found bound and drugged! Trends Mol
Med 2005;11:442–4.
3. Letai A, Sorcinelli MD, Beard C, Korsmeyer SJ.
Antiapoptotic BCL-2 is required for maintenance of a
model leukemia. Cancer Cell 2004;6:241–9.
4. Konopleva M, Zhao S, Hu W, et al. The anti-apoptotic
genes Bcl-X(L) and Bcl-2 are over-expressed and con-
tribute to chemoresistance of non-proliferating leukae-
mic CD34
+
cells. Br J Haematol 2002;118:521–34.
5. Del Poeta G, Venditti A, Del Principe MI, et al. Amount
of spontaneous apoptosis detected by Bax/Bcl-2 ratio
predicts outcome in acute myeloid leukemia (AML).
Blood 2003;101:2125–31.
6. Reed JC, Pellecchia M. Apoptosis-based therapies for
hematologic malignancies. Blood 2005;106:408–18.
7. Green DR, Kroemer G. Pharmacological manipulation
of cell death: clinical applications in sight? J Clin Invest
2005;115:2610–7.
8. Wang JL, Liu D, Zhang ZJ, et al. Structure-based
discovery of an organic compound that binds Bcl-2
protein and induces apoptosis of tumor cells. Proc Natl
Acad Sci U S A 2000;97:7124–9.
9. Walensky LD, Kung AL, Escher I, et al. Activation of
apoptosis in vivo by a hydrocarbon-stapled BH3 helix.
Science 2004;305:1466–70.
10. Oltersdorf T, Elmore SW, Shoemaker AR, et al. An
inhibitor of Bcl-2 family proteins induces regression of
solid tumours. Nature 2005;435:677–81.
11. Kuroda J, Puthalakath H, Cragg MS, et al. Bim and
Bad mediate imatinib-induced killing of Bcr/Abl
+
leukemic cells, and resistance due to their loss is
overcome by a BH3 mimetic. Proc Natl Acad Sci U S A
2006;103:14907–12.
12. Cory S, Adams JM. Killing cancer cells by flipping the
Bcl-2/Bax switch. Cancer Cell 2005;8:5–6.
13. Certo M, Moore VG, Nishino M, et al. Mitochondria
primed by death signals determine cellular addiction to
antiapoptotic BCL-2 family members. Cancer Cell 2006;
9:351–65.
14. Kaufmann SH, Karp JE, Svingen PA, et al. Elevated
expression of the apoptotic regulator Mcl-1 at the time
of leukemic relapse. Blood 1998;91:991–1000.
15. Derenne S, Monia B, Dean NM, et al. Antisense
strategy shows that Mcl-1 rather than Bcl-2 or Bcl-x(L)
is an essential survival protein of human myeloma cells.
Blood 2002;100:194–9.
16. Willis SN, Chen L, Dewson G, et al. Proapoptotic Bak
is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until
displaced by BH3-only proteins. Genes Dev 2005;19:
1294–305.
17. Chen R, Keating MJ, Gandhi V, Plunkett W.
Transcription inhibition by flavopiridol: mechanism of
chronic lymphocytic leukemia cell death. Blood 2005;
106:2513–9.
18. Alvi AJ, Austen B, Weston VJ, et al. A novel CDK
inhibitor, CYC202 (R -roscovitine), overcomes the defect
in p53-dependent apoptosis in B-CLL by down-regula-
tion of genes involved in transcription regulation and
survival. Blood 2005;105:4484–91.
19. MacCallum DE, Melville J, Frame S, et al. Seliciclib
(CYC202, R -roscovitine) induces cell death in multiple
myeloma cells by inhibition of RNA polymerase II-
dependent transcription and down-regulation of Mcl-1.
Cancer Res 2005;65:5399–407.
20. Dai Y, Rahmani M, Pei XY, et al. Farnesyltransferase
inhibitors interact synergistically with the Chk1 inhib-
itor UCN-01 to induce apoptosis in human leukemia
cells through interruption of both Akt and MEK/ERK
pathways and activation of SEK1/JNK. Blood 2005;105:
1706–16.
21. Dai Y, Dent P, Grant S. Tumor necrosis factor-related
apoptosis-inducing ligand (TRAIL) promotes mitochon-
drial dysfunction and apoptosis induced by 7-hydrox-
ystaurosporine and mitogen-activated protein kinase
kinase inhibitors in human leukemia cells that ectop-
ically express Bcl-2 and Bcl-xL. Mol Pharmacol 2003;64:
1402–9.
22. Zhou P, Qian L, Kozopas KM, Craig RW. Mcl-1, a
Bcl-2 family member, delays the death of hematopoietic
cells under a variety of apoptosis-inducing conditions.
Blood 1997;89:630–43.
23. Wei MC, Zong WX, Cheng EH, et al. Proapoptotic
BAX and BAK: a requisite gateway to mitochondrial
dysfunction and death. Science 2001;292:727–30.
24. Hsu YT, Youle RJ. Nonionic detergents induce
dimerization among members of the Bcl-2 family. J Biol
Chem 1997;272:13829–34.
25. Taniai M, Grambihler A, Higuchi H, et al. Mcl-1
mediates tumor necrosis factor-related apoptosis-
inducing ligand resistance in human cholangiocarci-
noma cells. Cancer Res 2004;64:3517–24.
26. Hartley PS, Bayne RA, Robinson LL, Fulton N,
Anderson RA. Developmental changes in expression of
myeloid cell leukemia-1 in human germ cells during
oogenesis and early folliculogenesis. J Clin Endocrinol
Metab 2002;87:3417–27.
27. Hahntow IN, Schneller F, Oelsner M, et al. Cyclin-
dependent kinase inhibitor roscovitine induces apopto-
sis in chronic lymphocytic leukemia cells. Leukemia
2004;18:747–55.
28. Reed JC. Proapoptotic multidomain Bcl-2/Bax-family
proteins: mechanisms, physiological roles, and thera-
peutic opportunities. Cell Death Differ 2006;13:1378–86.
29. Lucken-Ardjomande S, Martinou JC. Newcomers in
the process of mitochondrial permeabilization. J Cell Sci
2005;118:473–83.
30. Green DR. Apoptotic pathways: ten minutes to dead.
Cell 2005;121:671–4.
31. Cuconati A, Mukherjee C, Perez D, White E. DNA
damage response and MCL-1 destruction initiate apoptosis
in adenovirus-infected cells. Genes Dev 2003;17:2922–32.
32. Chen L, Willis SN, Wei A, et al. Differential targeting
of prosurvival Bcl-2 proteins by their BH3-only ligands
allows complementary apoptotic function. Mol Cell
2005;17:393–403.
33. Yang-Yen HF. Mcl-1: a highly regulated cell death and
survival controller. J Biomed Sci 2006;13:201–4.
34. Pei XY, Dai Y, Grant S. The small-molecule Bcl-2
inhibitor HA14–1 interacts synergistically with flavopir-
idol to induce mitochondrial injury and apoptosis in
human myeloma cells through a free radical-dependent
and Jun NH2-terminal kinase-dependent mechanism.
Mol Cancer Ther 2004;3:1513–24.
35. Cai D, Latham VM, Jr., Zhang X, Shapiro GI.
Combined depletion of cell cycle and transcriptional
cyclin-dependent kinase activities induces apoptosis in
cancer cells. Cancer Res 2006;66:9270–80.
36. Wittmann S, Bali P, Donapaty S, et al. Flavopiridol
down-regulates antiapoptotic proteins and sensitizes
human breast cancer cells to epothilone B-induced
apoptosis. Cancer Res 2003;63:93–9.
37. Nijhawan D, Fang M, Traer E, et al. Elimination of
Mcl-1 is required for the initiation of apoptosis following
ultraviolet irradiation. Genes Dev 2003;17:1475–86.
38. Qin JZ, Xin H, Sitailo LA, Denning MF, Nickoloff BJ.
Enhanced killing of melanoma cells by simultaneously
targeting Mcl-1 and NOXA. Cancer Res 2006;66:9636–45.
39. Kelekar A, Chang BS, Harlan JE, Fesik SW, Thompson
CB. Bad is a BH3 domain-containing protein that forms
an inactivating dimer with Bcl-XL. Mol Cell Biol 1997;17:
7040–6.
40. MikhailovV,MikhailovaM,DegenhardtK,
Venkatachalam MA, White E, Saikumar P. Association
of Bax and Bak homo-oligomers in mitochondria. Bax
requirement for Bak reorganization and cytochrome c
release. J Biol Chem 2003;278:5367–76.
41. Lindsten T, Ross AJ, King A, et al. The combined
functions of proapoptotic Bcl-2 family members bak
and bax are essential for normal development of
multiple tissues. Mol Cell 2000;6:1389–99.
42. Clohessy JG, Zhuang J, de Boer J, Gil-Gomez G,
Brady HJ. Mcl-1 interacts with truncated Bid and
inhibits its induction of cytochrome c release and its
role in receptor-mediated apoptosis. J Biol Chem 2006;
281:5750–9.
43. Han J, Goldstein LA, Gastman BR, Rabinowich H.
Interrelated roles for Mcl-1 and BIM in regulation of
TRAIL-mediated mitochondrial apoptosis. J Biol Chem
2006;281:10153–63.
44. Gomez-Bougie P, Bataille R, Amiot M. Endogenous
association of Bim BH3-only protein with Mcl-1, Bcl-xL
and Bcl-2 on mitochondria in human B cells. Eur J
Immunol 2005;35:971–6.
45. Kuwana T, Bouchier-Hayes L, Chipuk JE, et al. BH3
domains of BH3-only proteins differentially regulate Bax-
mediated mitochondrial membrane permeabilization
both directly and indirectly. Mol Cell 2005;17:525–35.
46. Sedlak TW, Oltvai ZN, Yang E, et al. Multiple Bcl-2
family members demonstrate selective dimerizations
with Bax. Proc Natl Acad Sci U S A 1995;92:7834–8.
47. Shoemaker AR, Oleksijew A, Bauch J, et al. A small-
molecule inhibitor of Bcl-XL potentiates the activity
of cytotoxic drugs in vitro and in vivo . Cancer Res 2006;
66:8731–9.
Research.
on May 30, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
... Venetoclax displayed modest monotherapy efficacy in AML (8), but was subsequently approved when administered in combination with hypomethylating agents or low-dose cytarabine, providing an additional 5 months overall survival based on the results of a phase III clinical trial for patients not fit for intensive induction chemotherapy (9). As in the case of other targeted agents, de novo or acquired resistance to venetoclax occurs through various mechanisms, including upregulation of other anti-apoptotic proteins (10) and downregulation of pro-apoptotic proteins (11), raising the possibility that combination strategies circumventing these events may be necessary to enhance the therapeutic efficacy of this agent in AML. ...
... Apoptosis was evaluated by flow cytometry using Annexin V-FITC/ PI staining as before (10). Loss of mitochondrial membrane potential and cell death were assessed by double staining with 7-AAD as before (10). ...
... Apoptosis was evaluated by flow cytometry using Annexin V-FITC/ PI staining as before (10). Loss of mitochondrial membrane potential and cell death were assessed by double staining with 7-AAD as before (10). Drug concentrations used in cytotoxicity studies were selected on the basis of relatively modest (e.g., generally <20%) singleagent induction of cell death. ...
Article
Purpose: Acute myeloid leukemia (AML) is an aggressive disease with a poor outcome. We investigated mechanisms by which the anti-AML activity of ABT-199 (venetoclax) could be potentiated by dual mTORC1/TORC2 inhibition. Methods: Venetoclax/INK128 synergism was assessed in various AML cell lines and primary patient AML samples in vitro. AML cells over-expressing MCL-1, constitutively active AKT, BAK and/or BAX knock-out, and acquired venetoclax resistance were investigated to define mechanisms underlying interactions. The antileukemic efficacy of this regimen was also examined in xenograft and patient-derived xenograft (PDX) models. Results: Combination treatment with venetoclax and INK128 (but not the mTORC1 inhibitor Rapamycin) dramatically enhanced cell death in AML cell lines. Synergism was associated with p-AKT and p-4EBP1 down-regulation and dependent upon MCL-1 downregulation and BAK/BAX up-regulation as MCL-1 overexpression and BAX/BAK knock out abrogated cell death. Constitutive AKT activation opposed synergism between venetoclax and PI3K or AKT inhibitors, but not INK128. Combination treatment also synergistically induced cell death in venetoclax- resistant AML cells. Similar events occurred in primary patient-derived leukemia samples but not normal CD34+ cells. Finally, venetoclax and INK128 co-treatment displayed increased anti-leukemia effects in in vivo xenograft and PDX models. Conclusions: The venetoclax/INK128 regimen exerts significant anti-leukemic activity in various preclinical models through mechanisms involving MCL-1 down-regulation and BAK/BAX activation, and offers potential advantages over PI3K or AKT inhibitors in cells with constitutive AKT activation. This regimen is active against primary and venetoclax resistant AML cells, and in in vivoAML models. Further investigation of this strategy appears warranted.
... 17,18 MM cells with high Mcl-1 expression are less sensitive to venetoclax, 13 but downregulation can overcome this. [19][20][21][22][23][24] These findings show that earlier therapies or clonal selection may have shifted cellular plasticity toward Mcl-1 dependency during disease development. 25 The research shows that Mcl-1 contributes to treatment resistance and illness progression. ...
Preprint
Despite a record number of clinical studies investigating various anti-cancer drugs, the 5-year survival rate for multiple myeloma (MM) patients in the United States is only 55%, and nearly all patients relapse. Poor patient outcomes demonstrate that myeloma cells are “born to survive,” which means they can adapt and evolve following treatment. As a result, new therapeutic approaches to combat this survival mechanism and target treatment-resistant malignant cells are required. Mcl-1, an anti-apoptotic protein, is required for the development of MM and resistance to therapy. This study looks at the possibility of KS18, a Mcl-1 inhibitor derived from pyoluteorin, to treat resistant MM. We show that KS18 inhibits Mcl-1 selectively and promotes post-translational modifications, resulting in UPS-dependent Mcl-1 degradation. Our findings show that KS18-induced Mcl-1 degradation results in caspase-dependent apoptosis. Importantly, KS18 triggered apoptosis in MM patient samples and bortezomib-resistant cells, synergizing with venetoclax to boost apoptosis. Furthermore, KS18 inhibits colony formation in bortezomib-resistant cells. KS18 treated NSG mice displayed significant tumor shrinkage without significant toxicity after four weeks of therapy with a single acceptable dose each week, indicating its powerful anti-neoplastic and anti-resistance characteristics. This study strongly implies that KS18 may treat MM and provide new hope to patients who are experiencing recurrence or resistance. Key points Given that KS18 is a robust Mcl-1 inhibitor that targets Mcl-1 efficiently, it has the potential to be a novel treatment for multiple myeloma. KS18 has shown promise in re-sensitizing myeloma cells to chemotherapy as well as in overcoming resistance to bortezomib, venetoclax, and ABT-737.
... Nevertheless, in our different model systems used in this study, the IC50 for ALX toxicity was only 2.5 µM, which is four times lower than the doses reported to inhibit Bcl-xL-Bak interactions. Furthermore, Bcl-XL is preferentially required for CML progression [34], while the anti-apoptotic MCL1 is essential for the survival of normal and Ph+ leukemic stem cell populations [35,36], suggesting that ALX must act on additional signaling pathways to target CSLs. ...
Article
Full-text available
Background & aims: In CML, Leukemic Stem Cells (LSCs) that are insensitive to Tyrosine Kinase Inhibitors are responsible for leukemia maintenance and relapses upon TKI treatment arrest. We previously showed that downregulation of the BMI1 polycomb protein that is crucial for stem/progenitor cells self-renewal induced a CCNG2/dependent proliferation arrest leading to elimination of Chronic Myeloid Leukemia (CML) cells. Unfortunately, as of today, pharmacological inhibition of BMI1 has not made its way to the clinic. Methods: We used the Connectivity Map bioinformatic database to identify pharmacological molecules that could mimick BMI1 silencing, to induce CML cell death. We selected the bis-biguanide Alexidin (ALX) that produced a transcriptomic profile positively correlating with the one obtained after BMI silencing in K562 CML cells. We then evaluated the efficiency of ALX in combination with TKI on CML cells. Results: Here we report that cell growth and clonogenic activity of K562 and LAMA-84 CML cell lines were strongly inhibited by ALX. ALX didn't modify BCR::ABL1 phosphorylation and didn't affect BMI1 expression but was able to increase CCNG2 expression leading to autophagic processes that preceed cell death. Besides, ALX could enhance the apoptotic response induced by any Tyrosine Kinase Inhibitors (TKI) of the three generations. We also noted a strong synergism between ALX and TKIs to increase expression of caspase-9 and caspase-3 and induce PARP cleavage, Bad expression and significantly decreased Bcl-xL family member expression. We also observed that the blockage of the mitochondrial respiratory chain by ALX can be associated with inhibition of glycolysis by 2-DG to achieve an enhanced inhibition of K562 proliferation and clonogenicity. ALX specifically affected the differentiation of BCR::ABL1-transduced healthy CD34+ cells but not of mock-infected healthy CD34+ control cells. Importantly, ALX strongly synergized with TKIs to inhibit clonogenicity of primary CML CD34+ cells from diagnosed patients. Long Term Culture of Initiating Cell (LTC-IC) and dilution of the fluorescent marker CFSE allowed us to observe that ALX and Imatinib (IM) partially reduced the number of LSCs by themselves but that the ALX/IM combination drastically reduced this cell compartment. Using an in vivo model of NSG mice intravenously injected with K562-Luciferase transduced CML cells, we showed that ALX combined with IM improved mice survival. Conclusions: Collectively, our results validate the use of ALX bis-biguanide to potentiate the action of conventional TKI treatment as a potential new therapeutic solution to eradicate CML LSCs.
... Cell death was evaluated by flow cytometry and assessed by double staining with Annexin V-FITC/Propidium Iodide (PI), as it has been before [10]. ...
Article
Full-text available
Interactions between the novel hypomethylating agent (HMA) thio-deoxycytidine (T-dCyd) and the BCL-2 antagonist ABT-199 (venetoclax) have been examined in human myelodysplastic syndrome (MDS) cells. The cells were exposed to agents alone or in combination, after which apoptosis was assessed, and a Western blot analysis was performed. Co-administration of T-dCyd and ABT-199 was associated with the down-regulation of DNA methyltransferase 1 (DNMT1) and synergistic interactions documented by a Median Dose Effect analysis in multiple MDS-derived lines (e.g., MOLM-13, SKM-1, and F-36P). Inducible BCL-2 knock-down significantly increased T-dCyd’s lethality in MOLM-13 cells. Similar interactions were observed in the primary MDS cells, but not in the normal cord blood CD34+ cells. Enhanced killing by the T-dCyd/ABT-199 regimen was associated with increased reactive oxygen species (ROS) generation and the down-regulation of the anti-oxidant proteins Nrf2 and HO-1, as well as BCL-2. Moreover, ROS scavengers (e.g., NAC) reduced lethality. Collectively, these data suggest that combining T-dCyd with ABT-199 kills MDS cells through an ROS-dependent mechanism, and we argue that this strategy warrants consideration in MDS therapy.
... However, ABT-737 demonstrated little or no activity towards Mcl-1, rendering it refractory. Targeted downregulation of Mcl-1 reinstated the sensitivity of cancer cells towards ABT-737, suggesting that Mcl-1 is a major resistance factor against ABT-737 and its derivatives and that suppressing Mcl-1 expression or its pharmacological inhibition would yield a much better clinical outcome [72][73][74]. The clinical efficacy of a similar BH3 mimetic that specifically targets Bcl-2, called ABT-199 (venetoclax) was limited due to Mcl-1 overexpression in AML. ...
Article
Full-text available
The antiapoptotic B cell lymphoma-2 (Bcl-2) family members are apical regulators of the intrinsic pathway of apoptosis that orchestrate mitochondrial outer membrane permeabilization (MOMP) through interactions with their proapoptotic counterparts. Overexpression of antiapoptotic Bcl-2 family proteins has been linked to therapy resistance and poor prognosis in diverse cancers. Among the antiapoptotic Bcl-2 family members, predominant overexpression of the prosurvival myeloid cell leukemia-1 (Mcl-1) has been reported in a myriad of hematological malignancies and solid tumors, contributing to therapy resistance and poor outcomes, thus making it a potential druggable target. The unique structure of Mcl-1 and its complex regulatory mechanism makes it an adaptive prosurvival switch that ensures tumor cell survival despite therapeutic intervention. This review focusses on diverse mechanisms adopted by tumor cells to maintain sustained elevated levels of Mcl-1 and how high Mcl-1 levels contribute to resistance in conventional as well as targeted therapies. Moreover, recent developments in the Mcl-1-targeted therapeutics and the underlying challenges and considerations in designing novel Mcl-1 inhibitors are also discussed.
... Consequently, the activity of Bcl-2 inhibitors is inversely related to expression of Mcl-1 in tumor cells (83,84). A corollary of this notion is that agents or interventions capable of down-regulating or inhibiting Mcl-1 could increase the activity of Bcl-2 inhibitors (85)(86)(87). Indeed, multiple such agents have been demonstrated to synergistically interact with Bcl-2 inhibitors in various hematologic malignancies. ...
Article
Full-text available
The introduction of various targeted agents into the armamentarium of cancer treatment has revolutionized the standard care of patients with cancer. However, like conventional chemotherapy, drug resistance, either preexisting (primary or intrinsic resistance) or developed following treatment (secondary or acquired resistance), remains the Achilles heel of all targeted agents with no exception, via either genetic or non-genetic mechanisms. In the latter, emerging evidence supports the notion that intracellular signaling pathways for tumor cell survival act as a mutually interdependent network via extensive cross-talks and feedback loops. Thus, dysregulations of multiple signaling pathways usually join forces to drive oncogenesis, tumor progression, invasion, metastasis, and drug resistance, thereby providing a basis for so-called “bypass” mechanisms underlying non-genetic resistance in response to targeted agents. In this context, simultaneous interruption of two or more related targets or pathways (an approach called dual-targeted therapy, DTT), via either linear or parallel inhibition, is required to deal with such a form of drug resistance to targeted agents that specifically inhibit a single oncoprotein or oncogenic pathway. Together, while most types of tumor cells are often addicted to two or more targets or pathways or can switch their dependency between them, DTT targeting either intrinsically activated or drug-induced compensatory targets/pathways would efficiently overcome drug resistance caused by non-genetic events, with a great opportunity that those resistant cells might be particularly more vulnerable. In this review article, we discuss, with our experience, diverse mechanisms for non-genetic resistance to targeted agents and the rationales to circumvent them in the treatment of cancer, emphasizing hematologic malignancies.
... We could restore sensitivity to ABT-737 whether downregulating of MCL1. MCL1 is a remarkably significant resistance to ABT-737, and is also resistance to decrease cytotoxicity of anticancer drug [92]. SIRT1mediated overview in chemotherapeutics resistance mechanism in cancer is shown in Figure 2. ...
Article
Cancer is a big group of diseases and one of the leading causes of mortality worldwide. Despite enormous studies and efforts are being carried out in understanding the cancer and developing drugs against tumorigenesis, drug resistance is the main obstacle in cancer treatments. Chemotherapeutic treatment is an important part of cancer treatment and drug resistance is getting gradually multidimensional with the advancement of studies in cancer. The underlying mechanisms of drug resistance are largely unknown. Sirtuin1 (SIRT1) is a type of the Class III histone deacetylase family that is distinctively dependent on nicotinamide adenine dinucleotide (NAD+) for catalysis reaction. SIRT1 is a molecule which upon upregulation directly influences tumor progression, metastasis, tumor cell apoptosis, autophagy, DNA repair, as well as other interlinked tumorigenesis mechanism. It is involved in drug metabolism, apoptosis, DNA damage, DNA repair, and autophagy, which are key hallmarks of drug resistance and may contribute to multidrug resistance. Thus, understanding the role of SIRT1 in drug resistance could be important. This study focuses on the SIRT1 based mechanisms that might be a potential underlying approach in the development of cancer drug resistance and could be a potential target for drug development.
Article
Full-text available
Varicella zoster virus (VZV) establishes lifelong infection after primary disease and can reactivate. Several drugs are approved to treat VZV diseases, but new antivirals with greater potency are needed. Previously, we identified β-l-5-((E)-2-bromovinyl)-1-((2S,4S)-2-(hydroxymethyl)-1,3-(dioxolane-4-yl))uracil (l-BHDU, 1), which had significant anti-VZV activity. In this communication, we report the synthesis and evaluation of numerous l-BHDU prodrugs: amino acid esters (14-26), phosphoramidates (33-34), long-chain lipids (ODE-l-BHDU-MP, 38, and HDP-l-BHDU-MP, 39), and phosphate ester prodrugs (POM-l-BHDU-MP, 41, and POC-l-BHDU-MP, 47). The amino acid ester l-BHDU prodrugs (l-phenylalanine, 16, and l-valine, 17) had a potent antiviral activity with EC50 values of 0.028 and 0.030 μM, respectively. The phosphate ester prodrugs POM-l-BHDU-MP and POC-l-BHDU-MP had a significant anti-VZV activity with EC50 values of 0.035 and 0.034 μM, respectively, and no cellular toxicity (CC50 > 100 μM) was detected. Out of these prodrugs, ODE-l-BHDU-MP (38) and POM-l-BHDU-MP (41) were selected for further evaluation in future studies.
Article
Avoidance of apoptosis is critical for the development and sustained growth of tumors. The pro-survival protein myeloid cell leukemia 1 (Mcl-1) is an anti-apoptotic member of the Bcl-2 family of proteins which is overexpressed in many cancers. Upregulation of Mcl-1 in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy. Therefore, pharmacological inhibition of Mcl-1 is regarded as an attractive approach to treating relapsed or refractory malignancies. Herein, we disclose the design, synthesis, optimization, and early preclinical evaluation of a potent and selective small-molecule inhibitor of Mcl-1. Our exploratory design tactics focused on structural modifications which improve the potency and physicochemical properties of the inhibitor while minimizing the risk of functional cardiotoxicity. Despite being in the "non-Lipinski" beyond-Rule-of-Five property space, the developed compound benefits from exquisite oral bioavailability in vivo and induces potent pharmacodynamic inhibition of Mcl-1 in a mouse xenograft model.
Article
In view of the crucial role of Mcl-1 in tumor survival and resistance as well as the importance of Bcl-XL in platelets homeostasis, an appropriate tuning of binding selectivity between Mcl-1 and Bcl-XL is crucial to discover safer anticancer candidates. In this respect, comprehensive computational approaches such as protein comparison, molecular docking, molecular dynamics simulations, mutagenesis, and density functional theory (DFT) calculation were applied to elucidate the different binding modes between highly selective Mcl-1 and Bcl-XL inhibitors. It was found that although Mcl-1 and Bcl-XL shared high sequence homology in their active pockets, the P2 pocket of Mcl-1 merged with P1 and P3, whereas the P3 pocket bridged through the P2 and P4 pocket of Bcl-XL. Accordingly, the key residues that determined the selectivity between Mcl-1 and Bcl-XL included Asn260, Arg263, and Phe270 in Mcl-1, and Glu96, Ser106, Leu108, Asn136, and Arg139 in Bcl-XL. In particular, Phe270 served as a major contributor to the π-π stacking interactions between Mcl-1 and its selective inhibitor, whereas Tyr195 was the key residue establishing the π−π stacking between Bcl-XL and its selective inhibitor. Collectively, these data shed promising light in depicting the selective mechanisms between Mcl-1 and Bcl-XL, which would lay a solid foundation for the future designing of the selective inhibitors in the discovery and optimization of a compound on the path toward developing future anticancer drugs.
Article
Full-text available
Members of the Bcl-2 family (including Bcl-2, Bcl-XL, and Bax) play key roles in the regulation of apoptosis. These proteins are believed to be membrane-associated and have been proposed to regulate apoptosis through both homodimerization and heterodimerization. We have found that whereas Bcl-2 is predominantly membrane-associated as previously reported, significant amounts of Bcl-XL and most of the Bax proteins are not membrane-associated and thus appear in the cytosolic fraction of thymocyte and splenocyte extracts. This finding allows the study of the dimerization properties and conformation of these proteins in the absence of detergent perturbation. For this analysis, we have produced monoclonal antibodies that are specific for known epitopes of Bax, Bcl-2, and Bcl-XL. An antibody to an N-terminal epitope (α uBax 6A7) between amino acids 12 and 24 fails to bind the soluble cytosolic form of Bax, indicating that this epitope is normally buried. Nonionic detergents alter the Bax conformation to expose this epitope. In the presence of nonionic detergent, the 6A7 antibody avidly binds the monomeric form of Bax, but not Bax complexed with either Bcl-XL or Bcl-2. In contrast, a monoclonal antibody to an adjacent epitope of Bax (α mBax 5B7) within amino acids 3–16 binds the soluble and detergent-altered forms of Bax and also binds the Bax·Bcl-XL or the Bax·Bcl-2 complex. Surprisingly, in the absence of detergent Bax fails to form homodimers or heterodimers with Bcl-XL. These results demonstrate a novel conformational state of members of the Bcl-2 family under a physiological condition that is distinct from the detergent-altered state that forms dimers and is currently believed to regulate apoptosis.
Article
Full-text available
Bcl-2, Bcl-xL, and Mcl-1 are three related intracellular polypeptides that have been implicated as negative regulators of apoptosis. In contrast, the partner protein Bax acts as a positive regulator of apoptosis. Based on the observation that all four of these polypeptides are expressed in a variety of acute myelogenous leukemia (AML) and acute lymphocytic leukemia (ALL) cell lines, cellular levels of these polypeptides were examined by immunoblotting in bone marrow samples harvested from 123 adult AML patients and 36 adult ALL patients before initial antileukemic therapy. Levels of Bcl-2, Mcl-1, Bcl-xL, and Bax each varied over a more than 10-fold range in different pretreatment leukemia specimens. When the 54 AML and 23 ALL samples that contained greater than 80% malignant cells were examined in greater detail, it was observed that pretreatment levels of Bcl-2 and Mcl-1 correlated with each other (R = .44, P < .001 for AML and R = .79, P < .0001 for ALL). In addition, a weak negative correlation between Bax expression and age was observed in AML samples (R = -0.35, P < .02) but not ALL samples. There was no relationship between pretreatment levels of these polypeptides and response to initial therapy. However, examination of 19 paired samples (the first harvested before chemotherapy and the second harvested 23 to 290 days later at the time of leukemic recurrence) revealed a greater than or equal to twofold increase in Mcl-1 levels in 10 of 19 pairs (7 of 15 AML and 3 of 4 ALL) at recurrence. In contrast, 2 of 19 pairs contained twofold less Mcl-1 at the time of recurrence. Approximately equal numbers of samples showed twofold increases and decreases in Bcl-2 (5 increases, 3 decreases) and Bcl-xL (1 increase, 4 decreases) at recurrence. Bax levels did not show a twofold decrease in any patient. these results, coupled with recent observations that cells overexpressing Mcl-1 are resistant to a variety of chemotherapeutic agents, raise the possibility that some chemotherapeutic regimens might select for leukemia cells with elevated levels of this particular apoptosis inhibitor.
Article
Full-text available
Bcl-2 and related proteins are key regulators of apoptosis or programmed cell death implicated in human disease including cancer. We recently showed that cell-permeable Bcl-2 binding peptides could induce apoptosis of human myeloid leukemia in vitro and suppress its growth in severe combined immunodeficient mice. Here we report the discovery of HA14-1, a small molecule (molecular weight = 409) and nonpeptidic ligand of a Bcl-2 surface pocket, by using a computer screening strategy based on the predicted structure of Bcl-2 protein. In vitro binding studies demonstrated the interaction of HA14-1 with this Bcl-2 surface pocket that is essential for Bcl-2 biological function. HA14-1 effectively induced apoptosis of human acute myeloid leukemia (HL-60) cells overexpressing Bcl-2 protein that was associated with the decrease in mitochondrial membrane potential and activation of caspase-9 followed by caspase-3. Cytokine response modifier A, a potent inhibitor of Fas-mediated apoptosis, did not block apoptosis induced by HA14-1. Whereas HA14-1 strongly induced the death of NIH 3T3 (Apaf-1+/+) cells, it had little apoptotic effect on Apaf-1-deficient (Apaf-1−/−) mouse embryonic fibroblast cells. These data are consistent with a mechanism by which HA14-1 induces the activation of Apaf-1 and caspases, possibly by binding to Bcl-2 protein and inhibiting its function. The discovery of this cell-permeable molecule provides a chemical probe to study Bcl-2-regulated apoptotic pathways in vivo and could lead to the development of new therapeutic agents.
Article
Full-text available
Proapoptotic Bcl-2 family members have been proposed to play a central role in regulating apoptosis. However, mice lacking bax display limited phenotypic abnormalities. As presented here, bak(-/-) mice were found to be developmentally normal and reproductively fit and failed to develop any age-related disorders. However, when Bak-deficient mice were mated to Bax-deficient mice to create mice lacking both genes, the majority of bax(-/-)bak(-/-) animals died perinatally with fewer than 10% surviving into adulthood. bax(-/-)bak(-/-) mice displayed multiple developmental defects, including persistence of interdigital webs, an imperforate vaginal canal, and accumulation of excess cells within both the central nervous and hematopoietic systems. Thus, Bax and Bak have overlapping roles in the regulation of apoptosis during mammalian development and tissue homeostasis.
Article
Full-text available
Multiple death signals influence mitochondria during apoptosis, yet the critical initiating event for mitochondrial dysfunction in vivo has been unclear. tBID, the caspase-activated form of a “BH3-domain–only” BCL-2 family member, triggers the homooligomerization of “multidomain” conserved proapoptotic family members BAK or BAX, resulting in the release of cytochrome c from mitochondria. We find that cells lacking both Bax andBak, but not cells lacking only one of these components, are completely resistant to tBID-induced cytochrome c release and apoptosis. Moreover, doubly deficient cells are resistant to multiple apoptotic stimuli that act through disruption of mitochondrial function: staurosporine, ultraviolet radiation, growth factor deprivation, etoposide, and the endoplasmic reticulum stress stimuli thapsigargin and tunicamycin. Thus, activation of a “multidomain” proapoptotic member, BAX or BAK, appears to be an essential gateway to mitochondrial dysfunction required for cell death in response to diverse stimuli.
Article
Engagement of death receptors such as tumor necrosis factor-R1 and Fas brings about the cleavage of cytosolic Bid to truncated Bid (tBid), which translocates to mitochondria to activate Bax/Bak, resulting in the release of cytochrome c. The mechanism underlying the activation, however, is not fully understood. Here, we have identified the anti-apoptotic Bcl-2 family member Mcl-1 as a potent tBid-binding partner. Site-directed mutagenesis reveals that the Bcl-2 homology (BH) 3 domain of tBid is essential for binding to Mcl-1, whereas all threeBHdomains (BH1, BH2, and BH3) of Mcl-1 are required for interaction with tBid. In vitro studies using isolated mitochondria and recombinant proteins demonstrate that Mcl-1 strongly inhibits tBid-induced cytochrome c release. In addition to its ability to interact directly with Bax and Bak, tBid also binds Mcl-1 and displaces Bak from the Mcl-1-Bak complex. Importantly, overexpression of Mcl-1 confers resistance to the induction of apoptosis by both TRAIL and tumor necrosis factor-alpha in HeLa cells, whereas targeting Mcl-1 by RNA interference sensitizes HeLa cells to TRAIL-induced apoptosis. Therefore, our study demonstrates a novel regulation of tBid by Mcl-1 through protein-protein interaction in apoptotic signaling from death receptors to mitochondria.
Article
Using a Bax-dependent membrane-permeabilization assay, we show that peptides corresponding to the BH3 domains of Bcl-2 family “BH3-only” proteins have dual functions. Several BH3 peptides relieved the inhibition of Bax caused by the antiapoptotic Bcl-xL and/or Mcl-1 proteins, some displaying a specificity for either Bcl-xL or Mcl-1. Besides having this derepression function, the Bid and Bim peptides activated Bax directly and were the only BH3 peptides tested that could potently induce cytochrome c release from mitochondria in cultured cells. Furthermore, Bax activator molecules (cleaved Bid protein and the Bim BH3 peptide) synergistically induced cytochrome c release when introduced into cells along with derepressor BH3 peptides. These observations support a unified model of BH3 domain function, encompassing both positive and negative regulation of other Bcl-2 family members. In this model, the simple inhibition of antiapoptotic functions is insufficient to induce apoptosis unless a direct activator of Bax or Bak is present.
Article
A family of Bcl-2-related proteins regulates cell death and shares highly conserved BH1 and BH2 domains. BH1 and BH2 domains of Bcl-2 were required for it to heterodimerize with Bax and to repress apoptosis. A yeast two-hybrid assay accurately reproduced this interaction and defined a selectivity and hierarchy of further dimerizations. Bax also heterodimerizes with Bcl-xL, Mcl-1, and A1. A Gly-159-->Ala substitution in BH1 of Bcl-xL disrupted its heterodimerization with Bax and abrogated its inhibition of apoptosis in mammalian cells. This suggests that the susceptibility to apoptosis is determined by multiple competing dimerizations in which Bax may be a common partner.
Article
Mcl-1 is a member of the Bcl-2 family that was identified based on increased expression in myeloblastic leukemia cells undergoing differentiation. Mcl-1 was previously found to be similar to Bcl-2 in causing a delay in apoptotic cell death in Chinese hamster ovary cells. The work described here was aimed at determining whether Mcl-1 could also exert such an effect in hematopoietic cells, because endogenous Mcl-1 expression is prominent in the hematopoietic system. A further aim was to assess the effects of Mcl-1 in cells exposed to a variety of cytotoxic stimuli, because Bcl-2 is known to have a broad spectrum of activity. To approach these aims, FDC-P1 murine myeloid progenitor cells were transfected with vectors driving either constitutive or inducible expression of Mcl-1. The introduced Mcl-1 gene was found to cause a prolongation of viability under various conditions that cause apoptotic cell death, including exposure to cytotoxic agents (the chemotherapeutic drug etoposide, calcium ionophore, or UV irradiation) and the withdrawal of required growth factors. In addition, Mcl-1 was found to interact with Bax, a member of the Bcl-2 family that promotes cell death as a homodimer but that can heterodimerize with Bcl-2 to promote cell viability. Although Mcl-1 prolonged cell viability, it did not prevent eventual cell death upon continuous exposure to a cytotoxic agent. Prolongation of viability was maximal when expression of Mcl-1 was induced before the application of the apoptotic stimulus, although some increase occurred if Mcl-1 was induced shortly thereafter and before overt apoptosis. Taken as a whole, these findings provide further parallels between Mcl-1 and Bcl-2, showing that Mcl-1 can interact with Bax in hematopoietic FDC-P1 cells and can prolong cell viability under a variety of cytotoxic conditions.
Article
The Bcl-2 related protein Bad is a promoter of apoptosis and has been shown to dimerize with the anti-apoptotic proteins Bcl-2 and Bcl-XL. Overexpression of Bad in murine FL5.12 cells demonstrated that the protein not only could abrogate the protective capacity of coexpressed Bcl-XL but could accelerate the apoptotic response to a death signal when it was expressed in the absence of exogenous Bcl-XL. Using deletion analysis, we have identified the minimal domain in the murine Bad protein that can dimerize with Bcl-xL. A 26-amino-acid peptide within this domain, which showed significant homology to the alpha-helical BH3 domains of related apoptotic proteins like Bak and Bax, was found to be necessary and sufficient to bind Bcl-xL. To determine the role of dimerization in regulating the death-promoting activity of Bad and the death-inhibiting activity of Bcl-xL, mutations within the hydrophobic BH3-binding pocket in Bcl-xL that eliminated the ability of Bcl-xL to form a heterodimer with Bad were tested for the ability to promote cell survival in the presence of Bad. Several of these mutants retained the ability to impart protection against cell death regardless of the level of coexpressed Bad protein. These results suggest that BH3-containing proteins like Bad promote cell death by binding to antiapoptotic members of the Bcl-2 family and thus inhibiting their survival promoting functions.