Caspase Control: Protagonists of Cancer Cell Apoptosis
Emergence of castration-resistant metastatic prostate cancer is due to activation of survival pathways, including apoptosis suppression and anoikis resistance, and increased neovascularization. Thus targeting of apoptotic players is of critical significance in prostate cancer therapy since loss of a...
Збережено в:
Дата: | 2012 |
---|---|
Автори: | , |
Формат: | Стаття |
Мова: | English |
Опубліковано: |
Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України
2012
|
Назва видання: | Experimental Oncology |
Теми: | |
Онлайн доступ: | http://dspace.nbuv.gov.ua/handle/123456789/139061 |
Теги: |
Додати тег
Немає тегів, Будьте першим, хто поставить тег для цього запису!
|
Назва журналу: | Digital Library of Periodicals of National Academy of Sciences of Ukraine |
Цитувати: | Caspase Control: Protagonists of Cancer Cell Apoptosis / M.V. Fiandalo, N. Kyprianou // Experimental Oncology. — 2012. — Т. 34, № 3. — С. 165-175. — Бібліогр.: 150 назв. — англ. |
Репозитарії
Digital Library of Periodicals of National Academy of Sciences of Ukraineid |
irk-123456789-139061 |
---|---|
record_format |
dspace |
spelling |
irk-123456789-1390612018-06-20T03:11:52Z Caspase Control: Protagonists of Cancer Cell Apoptosis Fiandalo, M.V. Kyprianou, N. Reviews Emergence of castration-resistant metastatic prostate cancer is due to activation of survival pathways, including apoptosis suppression and anoikis resistance, and increased neovascularization. Thus targeting of apoptotic players is of critical significance in prostate cancer therapy since loss of apoptosis and resistance to anoikis are critical in aberrant malignant growth, metastasis and conferring therapeutic failure. The majority of therapeutic agents act through intrinsic mitochondrial, extrinsic death receptor pathways or endoplasmic reticulum stress pathways to induce apoptosis. Current therapeutic strategies target restoring regulatory molecules that govern the pro-survival pathways such as PTEN which regulates AKT activity. Other strategies focus on reactivating the apoptotic pathways either by down-regulating anti-apoptotic players such as BCL-2 or by up-regulating pro-apoptotic protein families, most notably, the caspases. Caspases are a family of cystine proteases which serve critical roles in apoptotic and inflammatory signaling pathways. During tumorigenesis, significant loss or inactivation of lead members in the caspase family leads to impairing apoptosis induction, causing a dramatic imbalance in the growth dynamics, ultimately resulting in aberrant growth of human cancers. Recent exploitation of apoptosis pathways towards re-instating apoptosis induction via caspase re-activation has provided new molecular platforms for the development of therapeutic strategies effective against advanced prostate cancer as well as other solid tumors. This review will discuss the current cellular landscape featuring the caspase family in tumor cells and their activation via pharmacologic intervention towards optimized anti-cancer therapeutic modalities. This article is part of a Special Issue entitled “Apoptosis: Four Decades Later”. 2012 Article Caspase Control: Protagonists of Cancer Cell Apoptosis / M.V. Fiandalo, N. Kyprianou // Experimental Oncology. — 2012. — Т. 34, № 3. — С. 165-175. — Бібліогр.: 150 назв. — англ. 1812-9269 http://dspace.nbuv.gov.ua/handle/123456789/139061 en Experimental Oncology Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України |
institution |
Digital Library of Periodicals of National Academy of Sciences of Ukraine |
collection |
DSpace DC |
language |
English |
topic |
Reviews Reviews |
spellingShingle |
Reviews Reviews Fiandalo, M.V. Kyprianou, N. Caspase Control: Protagonists of Cancer Cell Apoptosis Experimental Oncology |
description |
Emergence of castration-resistant metastatic prostate cancer is due to activation of survival pathways, including apoptosis suppression and anoikis resistance, and increased neovascularization. Thus targeting of apoptotic players is of critical significance in prostate cancer therapy since loss of apoptosis and resistance to anoikis are critical in aberrant malignant growth, metastasis and conferring therapeutic failure. The majority of therapeutic agents act through intrinsic mitochondrial, extrinsic death receptor pathways or endoplasmic reticulum stress pathways to induce apoptosis. Current therapeutic strategies target restoring regulatory molecules that govern the pro-survival pathways such as PTEN which regulates AKT activity. Other strategies focus on reactivating the apoptotic pathways either by down-regulating anti-apoptotic players such as BCL-2 or by up-regulating pro-apoptotic protein families, most notably, the caspases. Caspases are a family of cystine proteases which serve critical roles in apoptotic and inflammatory signaling pathways. During tumorigenesis, significant loss or inactivation of lead members in the caspase family leads to impairing apoptosis induction, causing a dramatic imbalance in the growth dynamics, ultimately resulting in aberrant growth of human cancers. Recent exploitation of apoptosis pathways towards re-instating apoptosis induction via caspase re-activation has provided new molecular platforms for the development of therapeutic strategies effective against advanced prostate cancer as well as other solid tumors. This review will discuss the current cellular landscape featuring the caspase family in tumor cells and their activation via pharmacologic intervention towards optimized anti-cancer therapeutic modalities. This article is part of a Special Issue entitled “Apoptosis: Four Decades Later”. |
format |
Article |
author |
Fiandalo, M.V. Kyprianou, N. |
author_facet |
Fiandalo, M.V. Kyprianou, N. |
author_sort |
Fiandalo, M.V. |
title |
Caspase Control: Protagonists of Cancer Cell Apoptosis |
title_short |
Caspase Control: Protagonists of Cancer Cell Apoptosis |
title_full |
Caspase Control: Protagonists of Cancer Cell Apoptosis |
title_fullStr |
Caspase Control: Protagonists of Cancer Cell Apoptosis |
title_full_unstemmed |
Caspase Control: Protagonists of Cancer Cell Apoptosis |
title_sort |
caspase control: protagonists of cancer cell apoptosis |
publisher |
Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України |
publishDate |
2012 |
topic_facet |
Reviews |
url |
http://dspace.nbuv.gov.ua/handle/123456789/139061 |
citation_txt |
Caspase Control: Protagonists of Cancer Cell Apoptosis / M.V. Fiandalo, N. Kyprianou // Experimental Oncology. — 2012. — Т. 34, № 3. — С. 165-175. — Бібліогр.: 150 назв. — англ. |
series |
Experimental Oncology |
work_keys_str_mv |
AT fiandalomv caspasecontrolprotagonistsofcancercellapoptosis AT kyprianoun caspasecontrolprotagonistsofcancercellapoptosis |
first_indexed |
2025-07-10T07:32:27Z |
last_indexed |
2025-07-10T07:32:27Z |
_version_ |
1837244359777452032 |
fulltext |
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
CASPASE CONTROL: PROTAGONISTS OF CANCER CELL
APOPTOSIS
M.V. Fiandalo, N. Kyprianou*
Department of Molecular and Cellular Biochemistry and the Markey Cancer Center, University
of Kentucky College of Medicine, Department of Urology, Lexington, KY 40536, USA
Emergence of castration-resistant metastatic prostate cancer is due to activation of survival pathways, including apoptosis sup-
pression and anoikis resistance, and increased neovascularization. Thus targeting of apoptotic players is of critical significance
in prostate cancer therapy since loss of apoptosis and resistance to anoikis are critical in aberrant malignant growth, metastasis
and conferring therapeutic failure. The majority of therapeutic agents act through intrinsic mitochondrial, extrinsic death receptor
pathways or endoplasmic reticulum stress pathways to induce apoptosis. Current therapeutic strategies target restoring regulatory
molecules that govern the pro-survival pathways such as PTEN which regulates AKT activity. Other strategies focus on reactivat-
ing the apoptotic pathways either by down-regulating anti-apoptotic players such as BCL-2 or by up-regulating pro-apoptotic
protein families, most notably, the caspases. Caspases are a family of cystine proteases which serve critical roles in apoptotic and
inflammatory signaling pathways. During tumorigenesis, significant loss or inactivation of lead members in the caspase family leads
to impairing apoptosis induction, causing a dramatic imbalance in the growth dynamics, ultimately resulting in aberrant growth
of human cancers. Recent exploitation of apoptosis pathways towards re-instating apoptosis induction via caspase re-activation
has provided new molecular platforms for the development of therapeutic strategies effective against advanced prostate cancer
as well as other solid tumors. This review will discuss the current cellular landscape featuring the caspase family in tumor cells and
their activation via pharmacologic intervention towards optimized anti-cancer therapeutic modalities. This article is part of a Spe-
cial Issue entitled “Apoptosis: Four Decades Later”.
Key Words: apoptosis, caspase-8, proteasome inhibitors.
THE CHALLENGE IN CANCER TREATMENT
In the year ����� the challenge for clinicians� onco
lo gists and �asic scientists remains the development
of effective therapeutic strategies that �lock malignant
cell growth� without impairing normal healthy cells.
Investigative efforts focus on successful exploitation
of cancer specific characteristics acquired during
malignant transformation via a series of genetic and
epigenetic mutations resulting in uncontrolled growth
and evasion of apoptosis mechanisms [�]. Losing
critical regulatory mechanisms that control normal
tissue homeostasis ena�les tumor cells to acquire
new characteristics such as tissue invasion� metasta
sis and angiogenesis. �ome of the control pathways
are activated such as cell proliferation� cell cycle pro
gression and prosurvival pathways� while others are
downregulated� like the cell death pathways including
apoptosis and anoikis.
A�errant cell proliferation during cancer initiation
and progression to metastasis is controlled �y cell
cycle progression. The cell cycle consists of several
phases; G�� G�� �� G�� and Mitosis regulated �y vari
ous cyclins and CDK �cyclin dependant kinases� and
progression from one phase to another is dependent
on specific checkpoints [�]. A very critical player at this
checkpoint is p�� �notoriously known as the guardian
of the genome� due to its role in rescuing damaged
DNA� via upregulation of downstream effectors� such
as p�� �induces cell cycle arrest� and PUMA which
�locks antiapoptotic players leading to apoptosis
induction [�]. Cell cycle regulation via p�� can prima
rily �e a�rogated �y lossoffunction mutations� losing
p�� activity allows for the cell to replicate regardless
of DNA integrity and increases apoptosis resistance
[�]. Additional mechanisms of p�� downregulation
involve the overexpression of MDM�� an E� ligase
that mediates the polyu�iquitination of p�� resulting
in its degradation. Overexpression of MDM� ensures
rapid degradation of p�� leading to diminished if not
a�olished p�� activity and unregulated cell cycle
progression [�]. Proteasome inhi�itors� agents that
�lock protein degradation mediated �y ��� protea
some� have �een shown to sta�ilize p�� and restore
p�� mediated apoptosis [�]. Esta�lished chemo
therapeutic agents such as mitomycin C or doxoru
�icin are used to induce cell cycle arrest in a variety
of epithelial cancers. These agents work to either
cross link DNA �mitomycin C� or �ind directly to the
DNA intercalating into the dou�lehelix strands caus
ing the DNA to �ecome rigid and �reak �doxoru�icin�
[�]. The major limitation however is that these drugs
damage surrounding healthy normal cells� tissues�
and organs such as kidney �mitomycin C� or the heart
�doxoru�icin� [8� 9]. Treatment of MCF� �reast cancer
xenografts with a com�ination of mitomycin C with
Received: July 6, 2012
*Correspondence: E-mail: nkypr2@uky.edu
Abbreviations used: APC — allophycocyanin; ATCC — American type
culture collection; CRPC — castration-recurrent prostate cancer;
DISC — death inducing signaling complex; FADD — Fas associated
death domain; GST — glutathione S-transferase; HDAC7 — histone
deacetylase 7; IP — immunoprecipitation; MTT — 3-(4,5-dimethylthi-
azol-2-yl)-2,5-diphenyltetrazolium bromide; NB7 — neuroblastoma
7; PI — propidium iodide; TNF-α — tumor necrosis factor-α; TRAIL —
TNF-alpha related apoptosis inducing ligand.
Exp Oncol ����
��� �� �������
INVITED REVIEW
��� Experimental Oncology ��� �������� ���� ��eptem�er�
curcumin significantly decreased mitomycin C related
side effects [��].
�trategies involved with overcoming the toxic
side effects of doxoru�icin �y changing the mode
of doxoru�icin delivery �y encapsulating the drug
in titanium nanoparticals which showed promising
results [��]. Circumventing the caveats associated
with systemic toxicities of these chemotherapeutics
involved examining other prosurvival pathways� such
as the AKT signaling pathway which can also impact
cell cycle regulation and growth arrest. The AKT path
way is activated through �inding of growth factors
to their cognate tyrosine kinase receptors which then
carry out the signal transduction through the interplay
�etween �RC� phosphatidylinositol� kinase �PI�K�
and phosphotidylinositol�� ��isphosphate �PIP���
and phosphotidylinositol�� �� �triphosphate �PIP��
and a critical regulatory molecule� PTEN [��]. PTEN�
a phosphatase� regulates AKT activation �ecause
it dephosphorylates and converts PIP� to PIP� thus
preventing PIP��AKT interaction [��]. AKT is a kinase
that phosphorylates several different targets such
as mTOR� IKK �an inhi�itory �inding protein that pre
vents the nuclear factor of kappa B �NFκB� activation��
and cell cycle inhi�itors �p��� p���. In cancer activation
of the AKT pathway can �ecome a�errant �ecause
of a variety of mutations that can occur within PI3K�
AKT� and PTEN [��]. One of the most detrimental and
oncogenic potential promoting mutations are those
that render these molecules constitutively active. Con
stitutively active PI�K can lead to the increase in the
conversion rate of PIP� to PIP� favoring PIP� produc
tion leading to increased AKT activation [��]. A spe
cific AKT mutation� E��K in either AKT� or AKT� leads
to constitutive activation and promotes increased
trafficking to the plasma mem�rane [��]. PI3K and
AKT activating mutations are deleterious for the cell;
however� another mechanism that can elevate AKT
activity to supraphysiological levels and contri�ute
to oncogenesis is the loss of PTEN� a critical regula
tor of AKT activation. Interrogation of the signaling
events dictated �y AKT� mTOR� and PI�K has lead to the
development of a powerful class of pharmacologic
inhi�itors. The most promising class of AKT inhi�itors
are the lipid �ased inhi�itors which essentially inhi�it
AKT �inding to the plasma mem�rane. Perifosine has
emerged as one of the most promising AKT inhi�itors
and has �een through several phase II clinical tri
als [����9]. The mTOR inhi�itors include rapamycin
and its derivatives� such as CCI��9� �locks mTOR
function through similar mechanisms that primarily
involve �inding to the cofactor FKBP and together�
rapamycin and FBKP �ind to mTOR and inhi�it activity
[��]. PI�K inhi�itors like wortmannin or its derivative�
LY�9���� �ind covalently to PI�K to inhi�it its kinase
activity [��]. These agents however� lack specificity
and new carefully designed inhi�itors such as CAL
��� �Calistoga� have shown promising results in clini
cal trials [��] which are ongoing at Clearview Cancer
institute ������. PI�K inhi�itors exhi�it higher efficacy
com�ination with existing chemotherapeutic agents
such as� an AKT or mTOR inhi�itors �ecause these
com�inations �lock two pathways� eliminating indi
vidual pathway activity as well as preventing activation
of alternate or redundant nonAKT mediated pathways
activated through PI�K [��]. PTEN is the direct inhi�i
tor of AKT �ecause it converts AKT activating PIP� into
PIP�� which does not activate AKT. PTEN studies have
revealed that PTEN may �e downregulated either
through inactivating mutations� gene deletions� and
phosphorylation of PTEN �y CK� have the same result�
persistent AKT signaling that contri�utes to tumor
formation [��]. PTEN loss has �een associated with
several cancers at the advanced stage of disease�
including prostate cancer [��]. PTEN mutations have
�een linked with aggressive androgen dependant
or androgen independent �termed castration recur
rent� prostate cancer [��� ��] and evidence suggests
that oncogenesis results due to the loss of AKT and
cell cycle regulation [��� �8].
Prostate cancer is one of the most prevalent
causes of cancer related death in males with several
risk factors� such as age� race� and diet contri�uting
towards prostate cancer development and progression
[�9]. Regulation of androgen signaling via the andro
gen receptor �AR� is critical to maintaining prostate
homeostasis. The androgen axis involves conver
sion of testosterone into �αdihydrotestosterone
�y �αreductase� the active meta�olite that �inds
to AR and the ligandreceptor complex is translocated
to the nucleus to activate su�sequent signaling path
ways [��]. When prostate cells undergo tumorgenesis
they take on different molecular characteristics� one
of the more prominent changes is the upregulation
of androgen receptor either through gene amplifica
tion or through other processes leading to AR over
expression [��]. �uch an event prominently activates
AR pathways leading to increased proliferation and
reduced apoptosis� or may further sensitize prostate
cancer cells to growth factors stimuli such as EGFR
[��]. Currently the most promising therapy for treating
castrationrecurrent prostate cancer �CRPC�� involves
chemically depleting androgens in the prostate �y in
hi�itors of androgen axis such as a�iraterone [��].
Additional agents are currently �eing tested in clinical
trials such as MDV���� which is a competitive inhi�i
tor �locking AR�androgen signaling with therapeutic
promise in prostate cancer patients [��].
Another class of chemotherapeutics are protea
some inhi�itors capa�le of inducing apoptosis and thus
with great potential as anticancer agents [��]. There
are two types of proteasome inhi�itors� natural inhi�i
tors �lactacystin and epoxomicin� and synthetic inhi�i
tors such as MG��� and velcade [��]. MG��� inhi�its
the chymotrypsin like activity of the ��� proteasome
[��]. Velcade� �P����/�ortizomi�� is an FDA ap
proved proteasome inhi�itor used in treating multiple
myeloma [�8]. Velcade is a dipeptide �oronic acid
small molecule that �locks the chymotrypsinlike
activity of the ��� particle [�9]. Investigators have
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
reported that velcade has an impact on several key
cellular processes such as inhi�iting cell cycle and
NFκB activation [��]. Velcade sensitizes cancer
cells to apoptosis through several mechanisms� such
as the downregulation of cFLIP� which inhi�its cas
pase8 activation at the DI�C [��]. �everal in vitro
and in vivo studies have shown that velcade induces
apoptosis in multiple myeloma cells [��]. However
multiple myeloma patients are either initially resistant
or acquired resistance to velcade during the course
of treatment [��]. Attempts to overcome velcade
resistance have led to development of various com
�inations of velcade with different chemotherapeutic
agents such as� PCI���8� �an HDAC inhi�itor� which
was found to synergize with velcade to induce reactive
oxygen species damage as well as caspase8 activa
tion in nonHodgkins lymphoma [��]. �ignificantly
enough� Mitsiades and colleagues [��]� showed that
velcade in com�ination with doxoru�icin can overcome
velcade resistance in multiple myeloma. Another anti
cancer strategy involved using velcade in com�ination
with TNFα related apoptosis inducing ligand �TRAIL�.
Recent studies �y Christian et al. suggests the a�ility
of velcade to sensitize TRAILresistant prostate can
cer cell lines in vitro and in vivo to TRAILmediated
apoptosis and together TRAIL and velcade sta�ilize
caspase8 p�8 su�unit [��� ��].
A major caveat for using velcade as an anticancer
strategy is the lack of cell type and cell signaling
specificity. Velcade was designed to �lock the protea
some and not to discriminate �etween a malignant
or a healthy cell therefore� all cells are impacted �y vel
cade treatment. Velcade treatment can lead to side
effects such as throm�ocytopenia and peripheral
neuropathy [�8]. To �ypass the caveats associated
with velcade� while achieving apoptosis induction
investigators are pursuing the activity of E� ligases
in an attempt to achieve and their involvement with
the extrinsic pathway of apoptosis.
For CRPC� taxanes provide the only clinically ef
fective chemotherapeutic approach. These agents
target microtu�ules and the cellular cytoskeleton� thus
sta�ilizing microtu�ules and preventing microtu�ule
reorganization� towards disruption of kinetochore
formation during mitosis [�9]. Proposed mechanisms
conferring taxane resistance involve either microtu�ule
mutations that prevent drug �inding or the cell itself
pumps out the taxane through Pglycoprotein pumps
[��]. Taxanes have �een used against other solid
tumors such as �reast� lung� ovarian� and esopha
geal cancers [��� ��]. Alternative approaches involve
inducing or restoring the apoptotic pathways through
a variety of other agents such as staurosporin� etopo
side� and a new emerging class of apoptosis inducing
agents� the death ligands such as TRAIL.
MECHANISMS OF APOPTOSIS
REGULATION
Apoptosis �programmed cell death� plays a critical
role in regulating cell growth and tissue development.
�ince loss of apoptosis leads to tumor initiation�
growth� and progression [��]� exploitation of apoptosis
mechanisms can lead to developing new anticancer
strategies� that can effectively impair the tumorigenic
process. Each pathway of apoptosis is activated �y dif
ferent triggers such as celldetachment �anoikis� mito
chondrial signals �intrinsic pathway�� or death ligands
�extrinsic pathway� �Figs. � and ��.
Cellular insult ECM
Cytosol
AKT
BAX
BADBCL2
MOMP
opening
Caspase-2
Caspase-9
Caspase-4
Caspase 3, 7
Caspase-12
Endoplasmic
reticulum
tBID
XIAP
SMAC
Diablo
Apoptosome
Processed caspase-9
Apoptosis
APAF-1
Cleaves downstream targets
Cytochrome C release
Mitochondria
Fig. 1. Intrinsic and ER pathways of apoptosis. The intrin
sic pathway of apoptosis activated �y various stimuli� leads
to a downregulation of antiapoptotic BCL� family mem�ers�
allowing proapoptotic mem�ers to pertur� the mitochondria.
Cytochrome c release from the mitochondria leads to APAF� and
procaspase9 recruitment forming the apoptosome; cas
pase9 is then activated� cleaving downstream targets� such
as executioner caspase� and �. The ER stress pathway induces
the activation of caspase� or �� leading to caspase�� activa
tion� which then activates the intrinsic pathway of apoptosis
Cleaves downstream targets
Caspase 3, 7
Apoptosis
ECM
Cytosol
TRAIL
TRAIL Receptor
c-Flip FADD
Initiator
Pro-caspase-8
Processed
caspase-8
processed
HDAC7 BID
Intrinsic Pathway
activated
Death Inducing
Signaling Complex
(DISC)p43/41
Pro
Pro
18
18
18
18
10
10
10
10
Fig. 2. TRAILmediated extrinsic pathway of apoptosis. Medi
ated through the �inding of TRAIL to its cognate receptor� upon
�inding the receptors oligomerize within the mem�rane. Fas
associated death domain �FADD� is recruited� followed �y pro
caspase8 which is then processed into its active p�8 and
p�� su�units which then can oligomerize into a heterotetramer
A mechanism designed to protect against cellular
metastasis is anoikis� which is an apoptosis pro
gram that is induced upon the loss of critical protein
interactions �etween the cell and the extracellular
matrix. The major players and pathways involved with
anoikis are integrin� focal adhesion� and growth fac
tors �IGF�� interactions as well as the JNK pathway�
and caspase activation signaling events [��]. Anoikis
plays a role in all tissue development and regenera
tion and in preventing epithelial cell detachment and
migration in normal and tumor cells [��]� including
��8 Experimental Oncology ��� �������� ���� ��eptem�er�
shedding of colon epithelial cells [��] and mammary
gland reduction [��� �8]. Anoikis is initiated when
adherent cells detach from the �asement mem�rane�
more specifically� the loss of integrin �either α� or β��
signaling with the focal adhesion points [�9]. Apoptotic
pathways are activated upon cell detachment and
loss of integrin signaling in normal cells; cancer cells
develop resistance to anoikis through diverse mecha
nisms such as overcoming the loss of focal adhesion
kinase �FAK�� overexpression of talin �integrin partner/
focal adhesion player�� acquiring mutations in FAK
that trigger anoikis inhi�itory mechanisms or navigat
ing stimuli from the microenvironment signaling loss
of apoptotic pathways [��].
The intrinsic pathway of apoptosis is under
heavy regulation �y several different types of mo
lecules that can �e separated into two main classes�
antiapoptotic proteins such as the XIAP �inhi�itors
of apoptosis�� BCL� family proteins such as BCL��
BCLxL or the proapoptotic proteins� which include
BCL� family mem�ers; BAX� BAD� BID� �MAC� and
Dia�lo are activated through signaling events that
lead to mitochondrial outer mem�rane permea�iliza
tion �MOMP�. Cytochrome c is released� �inds with
APAF� and caspase9 to form the apoptosome [��].
Upon apoptosome formation� caspase9 �ecomes
catalytically active and acts on downstream targets
including caspase� and � �Fig. �� [��]. Tumor cells
can inactivate apoptotic signaling programs �y engag
ing antiapoptotic mechanisms involving the upregu
lation of apoptotic suppressors �Bcl�� BclxL� and/
or through the downregulation of critical apoptosis in
ducing players such as the caspase family �caspase��
�� �� 8� 9� ��� ��� [��]. Mechanisms that inhi�it
the intrinsic pathway of apoptosis are interconnected
with activities of the AKT �Fig. �� and NFκB pathways.
Therefore� activated AKT pathway inhi�its the intrin
sic pathway of apoptosis as AKT signaling promotes
BCL� and BCLxL activity while inhi�iting BAX and
BAD players involved with inducing apoptosis [��].
Blocking BAX and BAD activity can prevent MOMP
from opening� thus preventing cytochrome c release�
and consequentially inhi�iting apoptosome forma
tion. Another family of antiapoptotic proteins that can
inhi�it �oth the intrinsic and extrinsic pathways is the
inhi�itors of apoptosis �IAP� which have two arms� the
cIAP or Xlinked IAP �XIAP�. The IAP family consists
of E� ligases that can �lock apoptosome formation
through �inding directly to APAF� or caspase9� thus
inhi�iting caspase9 activation [��]. XIAP also �ind
directly to caspase� preventing its activation� and
inaddition to �locking activation XIAP can facilitate
the transfer of u�iquitin� there�y tagging the caspases
for degradation �y the ��� proteasome [��]. There
are also mutations acquired in the proapoptotic ma
chinery itself� the most nota�le mutations �eing those
occurring in the caspase family. To that end� �rinvisula
et al.� identified caspase9β� a caspase9 mutant that
lacks the large active su�unit and esta�lished that
caspase9β acts in a dominant negative fashion pre
venting caspase� activation [��]. Moreover� Park and
colleagues identified several gene polymorphisms
that give rise to altered forms of caspase9 that
impair caspase9 activity and there�y �lock apop
tosis induction [�8]. Posttranslational modification
of caspase9 phosphorylation at Thr ��9 mediated
as a result of CDK� and cyclin B in cell cycle [�9] also
prevents caspase9 recruitment to the apoptosome
�locking caspase9 activation. Regardless of how
caspase9 is modified� if this caspase fails to �e
come active then the su�sequently executioner cas
pase�/� activation is inhi�ited� ultimately impairing
the intrinsic pathway of apoptosis activation [��� ��].
The extrinsic pathway �also referred to as the death
receptor pathway� involves the induction of apoptosis
through the activation of death receptors via death
ligands such as tumor necrosis factorα �TNFα��
FA�L� and TRAIL [��]. While FA�L and TRAIL strictly
activate the extrinsic pathway mediated apoptosis�
TNFα can play two different roles� although this mole
cule is capa�le of inducing apoptosis� TNFα is also
capa�le of activating prosurvival pathway. TNFacti
vation impacts several critical cellular pathways some
of which include cellular proliferation� differentiation�
and apoptosis [��]. �pecifically� TNFα �inding to its
cognate receptor can lead to the formation of two
separate complexes� complex � which can lead to the
induction of either the NFκB pathway �prosurvival�
[��] or complex � which activates the apoptotic
pathway mediated primarily through Fas associated
death domain �FADD� and caspase8 activation [��].
Complex � mediated NFκB induction is initiated
through the �inding of TNFα to its cognate receptor
TNFR� which then leads to the recruitment of two
adaptor proteins TNF receptorassociated protein
with a death domain �TRADD� and receptorinteracting
protein � �RIP�� [��]. Upon �inding of TRADD another
adaptor molecule� TNF associated factor� �TRAF��
followed �y the recruitment of cIAP �cellular inhi�itors
of apoptosis�� as well as U�c� and U�c�� �E� u�iq
uitin conjugating enzymes� [��] to form complex �.
Once complex � is formed� TRAF� is phosphorylated
�y PKC resulting in K�� link polyu�iquitination [�8]
that leads to proteasomal degradation [�9]. TRAF��
cIAP� and U�c�� function in concert to facilitate
K�� linked polyu�iquitination of RIP� [8�]. K�� linked
polyu�iquitination of RIP� leads to activation of Tak�/
TAB complex to activate the IKK complex [8�]. The
IKK complex consists of several components� IKK α�
IKK β� and IKK γ� this kinase complex phosphorylates
the inhi�itor of kappa B molecule �IκBα� [8�]. IκB α�
is a �ound inhi�itor of NFκB that functions to prevent
nuclear import of NFκB into the nucleus. Nuclear
translocation of NFκB results in �inding to its re
spective DNA �inding sites and gene activation [8�].
NFκB upregulates several different gene types�
such as inflammatory response prosurvival genes�
BCL� family� caspase8 inhi�itor cFLIP� cIAP and
angiogenesis players and proliferation genes� cyclin
D� and MYC [8�]. Interestingly enough� proteasome
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ��9
inhi�ition can impede the NFκB pathway as it prevents
proteasome degradation of IκBα thus leading to de
creased activation of NFκB [8�].
Apoptosis induction through complex � of the
TNFα pathway proceeds via depletion of cFLIP and/
or cIAP expression� as well RIP� kinase phosphoryla
tion [8�� 8�]. Once phosphorylated� RIP�� FADD and
initiator caspase8 are recruited thus assem�ling com
plex �. Once complex � is formed� caspase8 is pro
cessed and can then cleave its downstream targets�
caspase� and �� towards apoptosis execution [88].
Recent evidence identified the ripoptosome� a �mD
apoptosis signaling complex composed of RIP��
FADD� and caspase8m as a key player in apoptosis
activation [89]. This complex assem�les when cIAP
expression levels are depleted� either through upreg
ulation of �MAC� a direct inhi�itor of cIAP� or through
�MAC mimetics or other chemotherapeutics such
as etoposide� which is a topoisomerase II inhi�itor
used to treat solid tumors. In addition to inducing DNA
�reaks� etoposide can downregulate cIAPs [89�9�].
The a�ility to form this apoptosis inducing complex�
can serve as a powerful tool for developing anti
cancer strategies �ecause an agent �or com�ination
of agents� could induce apoptosis� while �ypassing
the normal requirements for apoptosis induction.
Interestingly the ripoptosome triggers necroptopsis
�programmed necrosis� mediated through RIP� sig
naling [���].
The extrinsic pathway of apoptosis is a�rogated
through several mechanisms� including the upregula
tion of the inhi�itors of apoptosis proteins such as cIAP
or XIAP. Upregulation of these inhi�itors of apopto
sis molecules will drive the TNFα pathway towards
NFκB activation in the same manner as descri�ed
a�ove. Apart from inhi�ition �y the IAP family� recent
data indicate that IL�/�TAT� signaling can override
apoptotic signals �y activating prosurvival proteins
�BCL�� BCLxL� as well as cyclin D [9�]. TRAIL and
FA�mediated apoptosis pathways are very similar
to one another in that these trimeric ligands �ind their
specific cognate receptors towards apoptosis induc
tion. TRAIL �inds to the DR �/DR� receptors which leads
to receptor oligomerization in the plasma mem�rane�
some groups suggest that these receptors oligomerize
in the lipid rafts of the plasma mem�rane [9�]. Once
the receptors oligomerize there is recruitment of adap
tor protein FADD. FADD �inding to the TRAIL receptor
leads to initiator caspase8 recruitment to form the
death inducing signaling complex �DI�C�. Following
DI�C formation procaspase8 �ecomes autocata
lytically active� once active caspase8 is processed
into the active p�8 and p�� su�units via two cleavage
events. Once processed the p�8 and p�� dimers
can oligomerize with other p�8/p�� dimers to form
active heterotetramers� that cleaves specific targets
such as HDAC� [9�]� and executioner caspase� and
� which fully induce the apoptotic response [9�].
Tumor cells utilize various mechanisms to inactivate
the extrinsic pathway of apoptosis; that �e down
regulation of death receptors or upregulation of decoy
receptors [9�]. For TRAIL� its cognate receptors con
sist of death receptor� or � �DR�� ��� as a protec
tive measure� the cell also expresses decoy receptors
Dcr� and Dcr� as an effort to prevent unintended
apoptosis induction through TRAIL �inding to the
death receptors [98]. In addition to receptor or decoy
mediated inhi�ition� the extrinsic pathway is inhi�ited
�y overexpression of BCL�� BCLxL� cIAP and XIAP
antiapoptotic proteins [99� ���]. �tudies in mouse
models demonstrated that TRAIL targets cancer
cells and not healthy nonneoplastic cells [���]. This
specificity renders TRAIL a valua�le chemotherapeutic
agent due to the limited side effects and TRAIL protein
can �e synthesized via standard protein purification
methods [��]. TRAIL Cterminal conjugation can ex
tend TRAIL halflife �y �� hours allowing it to �e used
for in vitro and in vivo experiments. For clinical trial
and treatment applications companies� like Human
Genome �ciences �Rockville MD� U�A� have gene
rated TRAIL receptor activating anti�odies with the
intent to extend TRAIL halflife. These companies were
successful in generating TRAIL receptor anti�odies
as evidenced �y several in vitro and in vivo studies that
analyzed TRAIL anti�odies� mapatumuma� and lexa
tumuma� indicating their a�ility to induce apoptosis
[���� ���]. Additional preclinical studies have com
�ined TRAIL with existing chemotherapeutic agents�
including phytoshingosine �impacts sphingolipid me
ta�olism� [���]� doxoru�icin [���]� docetaxel [���]
and paclitaxel [���] all of which with much promise�
suggesting that TRAIL should �e investigated further
as a chemotherapeutic strategy. There is a clinical trial
in progress involving the com�ination of TRAIL and
VEGF inhi�itor� �evacizuma� �Clinical trial identifier�
NCT����8����.
CASPASES IN CONTROL OF APOPTOSIS
Caspases are a family of cysteine proteases which
contain cysteine residue at their active site and cleave
their su�strate at position next to aspartate residue.
A very unique family of enzymes which remain active
right from early stages of em�ryonic development
till the death of organism. The entire group of mam
malian caspases is divided into three different groups
on the �asis of their prodomains and specific func
tion they play in the in several different pathways�
including� inflammatory� development� and apoptotic
pathways. Although each caspase serves a different
purpose there are several similarities in cleavage�
the proform is cleaved into a large catalytically ac
tive su�unit and a small su�unit as shown for the
critical apoptotic caspase8. Caspase� and � play
a role in inflammation [��8� ��9]. The endoplasmic
reticulum �ER� stress response pathways� unfolded
protein response �UPR�� or ER associated degradation
�ERAD� are mediated through caspase�� eventu
ally leading apoptosis induction through the intrinsic
pathway. ER is a critical cellular organelle whose
primary function is to ensure proteins are properly
��� Experimental Oncology ��� �������� ���� ��eptem�er�
folded �efore export into the Golgi apparatus [���].
The protein folding machinery consists of several
components that work in concert to ensure proper
protein folding. However� when the ER is overwhelmed
with polypeptides that are incapa�le of �eing folded
correctly� three sensors� IRE�� PERK� and ATF� trig
ger the UPR [���]. As high ER stress persists� CHOP
expression is markedly increased� downregulating the
prosurvival BCL� family mem�ers� BCL� and BCL
xL allowing for BAX and BAD expression and activity
to increase [���]. Alternative intrinsic pathway induc
tion mechanisms involve the activation of caspase��
�� and �� [���]� contri�uting to caspase9 process
ing either through disrupting the mitochondria [���]
or through APAF� independent mechanisms [���].
Caspase� induces the intrinsic pathway of apoptosis
either through Bid cleavage thus leading to intrinsic
pathway activation via the opening of the MOMP re
leasing cytochrome c facilitating apoptosome forma
tion or through caspase�� PIDD� and adaptor protein
RAIDD �ind and form PIDDosome [���]. Compared
to healthy cells� tumor cells have a marked increase
in protein synthesis as well as an overproduction
of misfolded or unfolded protein in the ER� resulting
in extremely high ER stress levels. Therefore treatment
strategies focus on apoptosis induction via elevating
ER stress using chemotherapeutics such as velcade
[���]. Velcade is designed to �lock the ��� protea
some and once inhi�ited� UPR response mechanisms
�ecome impaired leading to apoptosis induction [��8].
To circumvent proteasome inhi�ition� cells activate
lysosomal pathways as an alternate mechanism for
protein clearance. Exploitation of this mechanism
as an anticancer strategy involves the use of vel
cade in com�ination with lysosomal inhi�itor� tu�acin
�an HDAC� inhi�itor that �lock aggresome formation��
that results apoptosis [��9]. The major players involved
with the intrinsic pathway of apoptosis induction� are
cytochrome c� APAF�� and caspase9 which form the
apoptosome. Caspase9 is su�sequently processed
into its active p�� and p�9 su�units [���] and capa�le
of cleaving executioner caspase�/�. Cancer circum
vents the intrinsic pathway activation �y engaging vari
ous mechanisms� such as loss of caspase9 activation
via the involvement of the BCL� family mem�ers� BCL
�� BCLxL or XIAP �inding� or �y �locking apoptosome
formation through prosurvival signals� preventing the
opening of the mitochondria and �locking the release
of cytochrome c [���]. Of direct clinical significance
is evidence that tumors from patients with colorectal�
lung� or gastric cancer� har�or different point muta
tions in caspase9� that render it inactive and incapa�le
of inducing apoptosis [���].
The mechanistic landscape of caspase activation
during the tumor cellular demise� takes intriguing
functional turns during cancer initiation and progres
sion �Fig. ��. Executioner caspase� and � �effector
caspases� are processed into active su�units and re
sponsi�le for the execution of the apoptosis program
through the cleavage of caspaseactivated DNase
which then translocates to the nucleus and cleaves
DNA [���]. Caspase� propagates and amplifies the
apoptosis signal through a loop that leads to cas
pase9 cleavage thus further propagating the apop
totic cascade [���]. Loss of caspase� expression pro
motes tumorigenesis [���]� while caspase� is down
regulated in cancer [���]. Caspase�� is an initiator
caspase recruited to the DI�C like caspase8 and is ca
pa�le of inducing apoptosis in certain caspase8 de
ficient tumor cells [���] however� caspase�� is not
sufficient to induce apoptosis in the a�sence of cas
pase8 in other cancer types [��8].
Fig. 3. General cleavage events for critical apoptotic related
caspases. Caspase8 and �� are larger than other caspases
�ecause they contain DED domains responsi�le for �inding
to the DI�C complex
CASPASE-8 IN DEATH RECEPTOR
INDEPENDENT AND DEPENDENT
PATHWAYS
Intriguing new evidence supports a role for cas
pase8 in nonapoptotic signaling pathways. �tupack
et al. ����9� reported that caspase8 in neuro�las
toma cell lines plays a role in mediating focal adhe
sion complex formation and cellular migration [��9].
Earlier studies defined a pathway� similar to anoikis
phenomenon� termed intergin mediated death [���].
Caspase8 is phosphorylated on tyrosine residue
�8� via �RC kinase and is associated with FAK and
CNB� and upon its recruitment activates the calpain
family of proteases that cleave talin [���]. �ignificantly
enough� the Nterminal cleavage product of talin�
the FERM� is an integrin �inding domain which facili
tates cell migration [��9]� indirectly implicating cas
pase8 in mediating metastasis via the focal adhesion
complex [��9]. Moreover� Ra��� a critical modulator
of caspase8 action in cell migration [���]� is function
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
ally involved in migration� either through lamellipodia
formation [���]� β� integrin �inding� or through actin
cytoskeleton [���].
�everal lines of evidence support the involvement
of caspase8 in EGF signaling pathways inducing
ERK activation through the incorporation of cas
pase8 in �RC containing complexes. This work iden
tified through a RXDLL motif found within the DED
of caspase8 prodomain� this allows caspase8 to as
sociate with �RC although the data did not show any
evidence that caspase8 phosphorylation was required
for �RC association and EGF pathway activation [���].
Caspase8 as a critical player for extrinsic pathway
activation has long �een considered a tumor sup
pressor molecule. Indeed caspase8 deficient cells
are insensitive to death ligand stimulus and cannot
induce apoptosis through the extrinsic pathway� thus
facilitating tumorigenic transformation and conferring
therapeutic resistance [���]. Human cancer cells
regulate caspase8 activity through a variety of mecha
nisms� one mechanism is caspase8 partial or whole
gene deletion� [���] or gene methylation. For example�
medullo�lastoma pediatric neuro�lastoma tumors
downregulate caspase8 expression through methy
lation of the caspase8 promoter there�y inhi�iting
caspase8 transcription thus preventing protein trans
lation and expression [��8]. �tudies involving a screen
across multiple cancer types identified frame shift and
missense mutations in caspase8 [��9]� which altered
amino acid compositions in the DED domain� a domain
a�solutely critical for caspase8 recruitment to the
DI�C and initiating cleavage events [��9]. Moreover�
mutations were found in the p�8 catalytically active
su�unit and the p�� regions validation of the screen
results revealed that most of the mutants severely
diminished apoptosis induction in gastric carcinomas
[��9]. Upon recruitment to the DI�C caspase8 un
dergoes two cleavage events� the first cleavage event
occurs at aspartic acid residue �8� in the p�� su�unit�
giving rise to the p��/�� intermediate which is �ound
at the DI�C. This cleavage event is followed �y a se
cond cleavage at aspartic acid residues ���� ��� which
then release caspase8 from the DI�C into the cy
tosol �Fig. ��. Pioneering work �y Dr. Marcus Peter
defined how the DI�C components were assem�led
at the plasma mem�rane through TRAIL and/or FA�
receptor and FADD palmitylation [���� ���]. Further
studies provided evidence towards DI�C mediated
caspase8 processing [���� ���]. Additional studies
focusing on DI�C formation �y Marcus Peter’s la�
identified that cFLIP was a specific inhi�itor of cas
pase8 DI�C recruitment and activation [���]. �u�se
quent work identified cFLIP isoforms that �lock gene
induction as well as processing of caspase8 [���].
Besides cFLIP� XIAP and cIAP are also capa�le
of �locking caspase8 activation [���]. Emerging
evidence �y two independent groups� Jin et al. [���]�
and Peng et al. [��8]� provided new insights regard
ing caspase8 polyu�iquitination. Jin and colleagues
provided evidence indicating that E� ligase CUL� me
diated polyu�iquitination led to caspase8 incorpora
tion into an aggresome. Caspase8 polyu�iquitination
however in the context of EGR signaling� is mediated
through R�K� activity [��8]� engaging two E� ligases�
�iah� and PO�H� in prostate cancer cells [��9]� thus
exerting a regulatory role on caspase8 activity down
stream of DI�C and caspase8 processing [��9]. The
com�ination of proteasome inhi�ition with TRAIL takes
an alllethal impact� as it induces apoptosis in TRAIL
resistant prostate cancer cells in vitro and in vivo [��].
Moreover� com�ination of TRAIL and velcade leads
to caspase8 p�8 su�unit sta�ilization [��� ��� ���]�
implicating caspase8 degradation �eing controlled
�y the ��� proteasome.
In summary� strategies to circumvent therapeutic
resistance �y restoration of apoptotic pathways� utiliz
ing single apoptosis inducing agents such as TRAIL�
separately or in com�ination with other chemothera
peutics� provide new promise in the clinical manage
ment of cancer patients. These apoptosis inducing
agents may also �e capa�le of inducing apoptosis�
regardless of the tumor hormonal milieu and driven
�y the cellular interactions with the tumor microenvi
ronment. In that regard com�ination of proteasome
inhi�itor and TRAIL is capa�le cleaving and activating
caspase8 in either androgendependent� or androgen
independent prostate cancer �CRPC�. The clinical
knowledge of microtu�ulintargeted chemotherapy
�taxanes� as the only effective treatment for CRPC�
calls for the need to understand the mechanisms
of action of this drug in order to augment its therapeu
tic efficacy and overcome the therapeutic resistance
to its antitumor actions in a large num�er of prostate
cancer patients. Profiling the caspase activation status
in response to taxane�ased chemotherapy �in com
�ination with apoptosisdriven agents� and in the
context of cytoskeleton organization� provides excit
ing new platforms for therapeutic optimization driving
apoptosis to its full execution in a su�set of tumors and
ultimately impacting patient survival.
ACKNOWLEDGEMENTS
The authors wish to acknowledge Drs. �teven
�chwarze� Vivek Rangnekar and �tephen �trup for
useful discussions and the administrative assistance
of Lorie Howard. The studies have �een supported
�y grants from the Department of Defense �U�AMRMC
PC�������� the James F. Hardymon Endowment at the
University of Kentucky College of Medicine and the
Markey Cancer Foundation.
REFERENCES
1. Hanahan D, Weinberg RA. The hallmarks of cancer.
Cell 2000; 100: 57–70.
2. Vermeulen K, van Bockstaele DR, Berneman ZN. The
cell cycle: a review of regulation, deregulation and therapeutic
targets in cancer. Cell Prolif 2003; 36: 131–49.
3. Malumbres M, Barbacid M. Cell cycle, CDKs and can-
cer: a changing paradigm. Nat Rev Cancer 2009; 9: 153–66.
4. Wang X, Di Pasqua AJ, Govind S, et al. Selective deple-
tion of mutant p53 by cancer chemopreventive isothiocyanates
��� Experimental Oncology ��� �������� ���� ��eptem�er�
and their structure-activity relationships. J Med Chem 2011;
54: 809–16.
5. Wasylyk C, Salvi R, Argentini M, et al. p53 medi-
ated death of cells overexpressing MDM2 by an inhibitor
of MDM2 interaction with p53. Oncogene 1999; 18: 1921–34.
6. Williams SA, McConkey DJ. The proteasome inhibitor
bortezomib stabilizes a novel active form of p53 in human
LNCaP-Pro5 prostate cancer cells. Cancer Res 2003;
63: 7338–44.
7. Celli CM, Jaiswal AK. Role of GRP58 in mitomycin C-
induced DNA cross-linking. Cancer Res 2003; 63: 6016–25.
8. Bugger H, Guzman C, Zechner C, et al. Uncoupling
protein downregulation in doxorubicin-induced heart failure
improves mitochondrial coupling but increases reactive oxy-
gen species generation. Cancer Chemother Pharmacol 2011;
67: 1381–8.
9. Kumari R, Sharma A, Ajay AK, Bhat MK. Mitomycin
C induces bystander killing in homogeneous and heteroge-
neous hepatoma cellular models. Mol Cancer 2009; 8: 87.
10. Zhou QM, Zhang H, Lu YY, et al. Curcumin reduced
the side effects of mitomycin C by inhibiting GRP58-mediated
DNA cross-linking in MCF-7 breast cancer xenografts. Cancer
Sci 2009; 100: 2040–5.
11. Chen Y, Wan Y, Wang Y, et al. Anticancer efficacy
enhancement and attenuation of side effects of doxorubicin
with titanium dioxide nanoparticles. Int J Nanomedicine
2011; 6: 2321–6.
12. Hennessy BT, Smith DL, Ram PT, et al. Exploiting the
PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug
Discov 2005; 4: 988–1004.
13. Sansal I, Sellers WR. The biology and clinical relevance
of the PTEN tumor suppressor pathway. J Clin Oncol 2004;
22: 2954–63.
14. Skeen JE, Bhaskar PT, Chen CC, et al. Akt deficiency
impairs normal cell proliferation and suppresses oncogenesis
in a p53-independent and mTORC1-dependent manner.
Cancer Cell 2006; 10: 269–80.
15. Kang S, Bader AG, Vogt PK. Phosphatidylinositol
3-kinase mutations identified in human cancer are oncogenic.
Proc Natl Acad Sci USA 2005; 102: 802–7.
16. Davies MA, Stemke-Hale K, Tellez C, et al. A novel
AKT3 mutation in melanoma tumours and cell lines. Br J Can-
cer 2008; 99: 1265–8.
17. Brachmann S, Fritsch C, Maira SM, Garcia-Echever-
ria C. PI3K and mTOR inhibitors: a new generation of targeted
anticancer agents. Curr Opin Cell Biol 2009; 21: 194–8.
18. Ernst DS, Eisenhauer E, Wainman N, et al. Phase
II study of perifosine in previously untreated patients with
metastatic melanoma. Invest New Drugs 2005; 23: 569–75.
19. Rahmani M, Reese E, Dai Y, et al. Coadministration
of histone deacetylase inhibitors and perifosine synergistically
induces apoptosis in human leukemia cells through Akt and
ERK1/2 inactivation and the generation of ceramide and reac-
tive oxygen species. Cancer Res 2005; 65: 2422–32.
20. Faivre S, Kroemer G, Raymond E. Current develop-
ment of mTOR inhibitors as anticancer agents. Nat Rev Drug
Discov 2006; 5: 671–88.
21. Maira SM, Stauffer F, Schnell C, Garcia-Echeverria C.
PI3K inhibitors for cancer treatment: where do we stand?
Biochem Soc Trans 2009; 37: 265–72.
22. Maira SM, Furet P, Stauffer F. Discovery of novel an-
ticancer therapeutics targeting the PI3K/Akt/mTOR pathway.
Future Med Chem 2009; 1: 137–55.
23. Courtney KD, Corcoran RB, Engelman JA. The PI3K
pathway as drug target in human cancer. J Clin Oncol 2010;
28: 1075–83.
24. Vazquez F, Grossman SR, Takahashi Y, et al. Phos-
phorylation of the PTEN tail acts as an inhibitory switch
by preventing its recruitment into a protein complex. J Biol
Chem 2001; 276: 48627–30.
25. Stahl JM, Cheung M, Sharma A, et al. Loss of PTEN
promotes tumor development in malignant melanoma. Cancer
Res 2003; 63: 2881–90.
26. Bismar TA, Yoshimoto M, Duan Q, et al. Interac-
tions and relationships of PTEN, ERG, SPINK1 and
AR in castration-resistant prostate cancer. Histopathology
2012; 60: 645–52.
27. Yoshimoto M, Ludkovski O, DeGrace D, et al. PTEN
genomic deletions that characterize aggressive prostate cancer
originate close to segmental duplications. Genes Chromo-
somes Cancer 2012; 51: 149–60.
28. Vlietstra RJ, van Alewijk DC, Hermans KG, et al. Fre-
quent inactivation of PTEN in prostate cancer cell lines and
xenografts. Cancer Res 1998; 58: 2720–3.
29. Jemal A, Center MM, DeSantis C, Ward EM. Global
patterns of cancer incidence and mortality rates and trends.
Cancer Epidemiol Biomarkers Prev 2010; 19: 1893–907.
30. Heinlein CA, Chang C. The roles of androgen recep-
tors and androgen-binding proteins in nongenomic androgen
actions. Mol Endocrinol 2002; 16: 2181–7.
31. George D, Moul JW. Emerging treatment options for
patients with castration-resistant prostate cancer. Prostate
2012; 72: 338–49.
32. Stanbrough M, Bubley GJ, Ross K, et al. Increased
expression of genes converting adrenal androgens to testo-
sterone in androgen-independent prostate cancer. Cancer Res
2006; 66: 2815–25.
33. Nelson WG, Haffner MC, Yegnasubramanian S. Beef-
ing up prostate cancer therapy with performance-enhancing
(anti-) steroids. Cancer Cell 2011; 20: 7–9.
34. Scher HI, Beer TM, Higano CS, et al. Antitumour
activity of MDV3100 in castration-resistant prostate can-
cer: a phase 1-2 study. Lancet 2010; 375: 1437–46.
35. Vu HY, Juvekar A, Ghosh C, et al. Proteasome in-
hibitors induce apoptosis of prostate cancer cells by inducing
nuclear translocation of IkappaBalpha. Arch Biochem Biophys
2008; 475: 156–63.
36. Almond JB, Cohen GM. The proteasome: a novel target
for cancer chemotherapy. Leukemia 2002; 16: 433–43.
37. Li W, Zhang X, Olumi AF. MG-132 sensitizes TRAIL-
resistant prostate cancer cells by activating c-Fos/c-Jun
heterodimers and repressing c-FLIP(L). Cancer Res 2007;
67: 2247–55.
38. Crawford LJ, Walker B, Ovaa H, et al. Compara-
tive selectivity and specificity of the proteasome inhibitors
BzLLLCOCHO, PS-341, and MG-132. Cancer Res 2006;
66: 6379–86.
39. Berkers CR, Verdoes M, Lichtman E, et al. Activity
probe for in vivo profiling of the specificity of proteasome
inhibitor bortezomib. Nat Methods 2005; 2: 357–62.
40. Yu C, Rahmani M, Dent P, Grant S. The hierarchical
relationship between MAPK signaling and ROS generation
in human leukemia cells undergoing apoptosis in response
to the proteasome inhibitor Bortezomib. Exp Cell Res 2004;
295: 555–66.
41. Sayers TJ, Brooks AD, Koh CY, et al. The protea-
some inhibitor PS-341 sensitizes neoplastic cells to TRAIL-
mediated apoptosis by reducing levels of c-FLIP. Blood 2003;
102: 303–10.
42. Balsas P, Lopez-Royuela N, Galan-Malo P, et al. Co-
operation between Apo2L/TRAIL and bortezomib in multiple
myeloma apoptosis. Biochem Pharmacol 2009; 77: 804–12.
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
43. Nencioni A, Hua F, Dillon CP, et al. Evidence for
a protective role of Mcl-1 in proteasome inhibitor-induced
apoptosis. Blood 2005; 105: 3255–62.
44. Bhalla S, Balasubramanian S, David K, et al. PCI-
24781 induces caspase and reactive oxygen species-dependent
apoptosis through NF-kappaB mechanisms and is synergistic
with bortezomib in lymphoma cells. Clin Cancer Res 2009;
15: 3354–65.
45. Mitsiades N, Mitsiades CS, Richardson PG, et al. The
proteasome inhibitor PS-341 potentiates sensitivity of multiple
myeloma cells to conventional chemotherapeutic agents: thera-
peutic applications. Blood 2003; 101: 2377–80.
46. Thorpe JA, Christian PA, Schwarze SR. Proteasome
inhibition blocks caspase-8 degradation and sensitizes prostate
cancer cells to death receptor-mediated apoptosis. Prostate
2008; 68: 200–9.
47. Christian PA, Thorpe JA, Schwarze SR. Velcade sensi-
tizes prostate cancer cells to TRAIL induced apoptosis and sup-
presses tumor growth in vivo. Cancer Biol Ther 2009; 8: 73–80.
48. Richardson PG, Xie W, Mitsiades C, et al. Single-agent
bortezomib in previously untreated multiple myeloma: efficacy,
characterization of peripheral neuropathy, and molecular cor-
relations with response and neuropathy. J Clin Oncol 2009;
27: 3518–25.
49. Giannakakou P, Gussio R, Nogales E, et al. A common
pharmacophore for epothilone and taxanes: molecular basis
for drug resistance conferred by tubulin mutations in human
cancer cells. Proc Natl Acad Sci USA 2000; 97: 2904–9.
50. Madan RA, Pal SK, Sartor O, Dahut WL. Overcoming
chemotherapy resistance in prostate cancer. Clin Cancer Res
2011; 17: 3892–902.
51. Ehrlichova M, Koc M, Truksa J, et al. Cell death
induced by taxanes in breast cancer cells: cytochrome C is re-
leased in resistant but not in sensitive cells. Anticancer Res
2005; 25: 4215–24.
52. Ramalingam S, Belani CP. Taxanes for advanced
non-small cell lung cancer. Exp Opin Pharmacother 2002;
3: 1693–709.
53. Lowe SW, Lin AW. Apoptosis in cancer. Carcinogenesis
2000; 21: 485–95.
54. Zhan M, Zhao H, Han ZC. Signalling mechanisms
of anoikis. Histol Histopathol 2004; 19: 973–83.
55. Frisch SM, Screaton RA. Anoikis mechanisms. Curr
Opin Cell Biol 2001; 13: 555–62.
56. Strater J, Wedding U, Barth TF, et al. Rapid onset
of apoptosis in vitro follows disruption of beta 1-integrin/matrix
interactions in human colonic crypt cells. Gastroenterology
1996; 110: 1776–84.
57. Lund LR, Romer J, Thomasset N, et al. Two distinct
phases of apoptosis in mammary gland involution: proteinase-
independent and -dependent pathways. Development 1996;
122: 181–93.
58. Haenssen KK, Caldwell SA, Shahriari KS, et al.
ErbB2 requires integrin alpha5 for anoikis resistance via Src
regulation of receptor activity in human mammary epithelial
cells. J Cell Sci 2010; 123: 1373–82.
59. Vachon PH. Integrin signaling, cell survival, and
anoikis: distinctions, differences, and differentiation. J Signal
Transduct 2011; 2011: 738137.
60. Rennebeck G, Martelli M, Kyprianou N. Anoikis and
survival connections in the tumor microenvironment: is there
a role in prostate cancer metastasis? Cancer Res 2005;
65: 11230–5.
61. Siu WP, Pun PB, Latchoumycandane C, Boelster-
li UA. Bax-mediated mitochondrial outer membrane permea-
bilization (MOMP), distinct from the mitochondrial perme-
ability transition, is a key mechanism in diclofenac-induced
hepatocyte injury: Multiple protective roles of cyclosporin A.
Toxicol Appl Pharmacol 2008; 227: 451–61.
62. Ohtsuka T, Buchsbaum D, Oliver P, et al. Synergistic
induction of tumor cell apoptosis by death receptor antibody
and chemotherapy agent through JNK/p38 and mitochondrial
death pathway. Oncogene 2003; 22: 2034–44.
63. Elmore S. Apoptosis: a review of programmed cell
death. Toxicol Pathol 2007; 35: 495–516.
64. Tait SW, Green DR. Mitochondria and cell death: outer
membrane permeabilization and beyond. Nat Rev Mol Cell
Biol 2010; 11: 621–32.
65. Deveraux QL, Roy N, Stennicke HR, et al. IAPs block
apoptotic events induced by caspase-8 and cytochrome c by di-
rect inhibition of distinct caspases. EMBO J 1998; 17: 2215–23.
66. Schile AJ, Garcia-Fernandez M, Steller H. Regulation
of apoptosis by XIAP ubiquitin-ligase activity. Genes Dev
2008; 22: 2256–66.
67. Srinivasula SM, Ahmad M, Guo Y, et al. Identifica-
tion of an endogenous dominant-negative short isoform
of caspase-9 that can regulate apoptosis. Cancer Res 1999;
59: 999–1002.
68. Park JY, Park JM, Jang JS, et al. Caspase 9 promoter
polymorphisms and risk of primary lung cancer. Hum Mol
Genet 2006; 15: 1963–71.
69. Allan LA, Clarke PR. Apoptosis and autopha-
gy: Regulation of caspase-9 by phosphorylation. FEBS J 2009;
276: 6063–73.
70. Janssen K, Pohlmann S, Janicke RU, et al. Apaf-1 and
caspase-9 deficiency prevents apoptosis in a Bax-controlled
pathway and promotes clonogenic survival during paclitaxel
treatment. Blood 2007; 110: 3662–72.
71. Bratton SB, Salvesen GS. Regulation of the Apaf-1–
caspase-9 apoptosome. J Cell Sci 2010; 123: 3209–14.
72. Wallach D, Kang TB, Kovalenko A. The extrinsic cell
death pathway and the elan mortel. Cell Death Differ 2008;
15: 1533–41.
73. Naude PJ, den Boer JA, Luiten PG, Eisel UL. Tumor
necrosis factor receptor cross-talk. FEBS J 2011; 278: 888–98.
74. Wajant H, Scheurich P. TNFR1-induced activa-
tion of the classical NF-kappaB pathway. FEBS J 2011;
278: 862–76.
75. Wang L, Du F, Wang X. TNF-alpha induces two dis-
tinct caspase-8 activation pathways. Cell 2008; 133: 693–703.
76. Chen ZJ. Ubiquitin signalling in the NF-kappaB
pathway. Nat Cell Biol 2005; 7: 758–65.
77. Wu CJ, Conze DB, Li X, et al. TNF-alpha induced
c-IAP1/TRAF2 complex translocation to a Ubc6-containing
compartment and TRAF2 ubiquitination. EMBO J 2005;
24: 1886–98.
78. Li S, Wang L, Dorf ME. PKC phosphorylation
of TRAF2 mediates IKKalpha/beta recruitment and K63-
linked polyubiquitination. Mol Cell 2009; 33: 30–42.
79. Habelhah H, Takahashi S, Cho SG, et al. Ubiquiti-
nation and translocation of TRAF2 is required for activa-
tion of JNK but not of p38 or NF-kappaB. EMBO J 2004;
23: 322–32.
80. Wicovsky A, Henkler F, Salzmann S, et al. Tumor
necrosis factor receptor-associated factor-1 enhances proin-
flammatory TNF receptor-2 signaling and modifies TNFR1–
TNFR2 cooperation. Oncogene 2009; 28: 1769–81.
81. Habelhah H. Emerging complexity of protein ubiq-
uitination in the NF-kappaB pathway. Genes Cancer 2010;
1: 735–47.
��� Experimental Oncology ��� �������� ���� ��eptem�er�
82. Ea CK, Deng L, Xia ZP, et al. Activation of IKK
by TNFalpha requires site-specific ubiquitination of RIP1 and
polyubiquitin binding by NEMO. Mol Cell 2006; 22: 245–57.
83. Mathes E, O’Dea EL, Hoffmann A, Ghosh G. NF-
kappaB dictates the degradation pathway of IkappaBalpha.
EMBO J 2008; 27: 1357–67.
84. Papa S, Bubici C, Zazzeroni F, et al. The NF-kappaB-
mediated control of the JNK cascade in the antagonism
of programmed cell death in health and disease. Cell Death
Differ 2006; 13: 712–29.
85. Li XH, Fang X, Gaynor RB. Role of IKKgamma/nemo
in assembly of the Ikappa B kinase complex. J Biol Chem 2001;
276: 4494–500.
86. Rangamani P, Sirovich L. Survival and apoptotic
pathways initiated by TNF-alpha: modeling and predictions.
Biotechnol Bioeng 2007; 97: 1216–29.
87. Biton S, Ashkenazi A. NEMO and RIP1 control cell
fate in response to extensive DNA damage via TNF-alpha
feedforward signaling. Cell 2011; 145: 92–103.
88. Yuan J, Kroemer G. Alternative cell death mechanisms
in development and beyond. Genes Dev 2010; 24: 2592–602.
89. Tenev T, Bianchi K, Darding M, et al. The Ripopto-
some, a signaling platform that assembles in response to geno-
toxic stress and loss of IAPs. Mol Cell 2011; 43: 432–48.
90. Bertrand MJ, Vandenabeele P. The Ripoptosome: death
decision in the cytosol. Mol Cell 2011; 43: 323–5.
91. Imre G, Larisch S, Rajalingam K. Ripoptosome: a novel
IAP-regulated cell death-signalling platform. J Mol Cell Biol
2011; 3: 324–6.
92. Feoktistova M, Geserick P, Kellert B, et al. cIAPs block
Ripoptosome formation, a RIP1/caspase-8 containing intra-
cellular cell death complex differentially regulated by cFLIP
isoforms. Mol Cell 2011; 43: 449–63.
93. Li S, Wang N, Brodt P. Metastatic cells can escape the
proapoptotic effects of TNF-alpha through increased auto-
crine IL-6/STAT3 signaling. Cancer Res 2012; 72: 865–75.
94. Song JH, Tse MC, Bellail A, et al. Lipid rafts and non-
rafts mediate tumor necrosis factor related apoptosis-inducing
ligand induced apoptotic and nonapoptotic signals in non
small cell lung carcinoma cells. Cancer Res 2007; 67: 6946–55.
95. Scott FL, Fuchs GJ, Boyd SE, et al. Caspase-8 cleaves
histone deacetylase 7 and abolishes its transcription repressor
function. J Biol Chem 2008; 283: 19499–510.
96. Diessenbacher P, Hupe M, Sprick MR, et al. NF-
kappaB inhibition reveals differential mechanisms of TNF
versus TRAIL-induced apoptosis upstream or at the level
of caspase-8 activation independent of cIAP2. J Invest Der-
matol 2008; 128: 1134–47.
97. Matsuda T, Almasan A, Tomita M, et al. Resistance
to Apo2 ligand (Apo2L)/tumor necrosis factor-related apopto-
sis-inducing ligand (TRAIL)-mediated apoptosis and consti-
tutive expression of Apo2L/TRAIL in human T-cell leukemia
virus type 1-infected T-cell lines. J Virol 2005; 79: 1367–78.
98. LeBlanc HN, Ashkenazi A. Apo2L/TRAIL and its
death and decoy receptors. Cell Death Differ 2003; 10: 66–75.
99. Hinz S, Trauzold A, Boenicke L, et al. Bcl-xL protects
pancreatic adenocarcinoma cells against CD95- and TRAIL-
receptor-mediated apoptosis. Oncogene 2000; 19: 5477–86.
100. Fulda S, Meyer E, Debatin KM. Inhibition of TRAIL-
induced apoptosis by Bcl-2 overexpression. Oncogene 2002;
21: 2283–94.
101. Mitsiades CS, Treon SP, Mitsiades N, et al. TRAIL/
Apo2L ligand selectively induces apoptosis and overcomes
drug resistance in multiple myeloma: therapeutic applications.
Blood 2001; 98: 795–804.
102. Belyanskaya LL, Marti TM, Hopkins-Donaldson S,
et al. Human agonistic TRAIL receptor antibodies Mapatu-
mumab and Lexatumumab induce apoptosis in malignant
mesothelioma and act synergistically with cisplatin. Mol
Cancer 2007; 6: 66.
103. Luster TA, Carrell JA, McCormick K, et al. Mapatumum-
ab and lexatumumab induce apoptosis in TRAIL-R1 and TRAIL-
R2 antibody-resistant NSCLC cell lines when treated in combina-
tion with bortezomib. Mol Cancer Ther 2009; 8: 292–302.
104. Choi SY, Kim MJ, Chung HY, et al. Phytosphingosine
in combination with TRAIL sensitizes cancer cells to TRAIL
through synergistic up-regulation of DR4 and DR5. Oncol
Rep 2007; 17: 175–84.
105. Guo L, Fan L, Pang Z, et al. TRAIL and doxorubicin
combination enhances anti-glioblastoma effect based on passive
tumor targeting of liposomes. J Control Release 2011; 154: 93–102.
106. Yoo J, Park SS, Lee YJ. Pretreatment of docetaxel
enhances TRAIL-mediated apoptosis in prostate cancer cells.
J Cell Biochem 2008; 104: 1636–46.
107. Mielgo A, Torres VA, Clair K, et al. Paclitaxel promotes
a caspase 8-mediated apoptosis through death effector domain
association with microtubules. Oncogene 2009; 28: 3551–62.
108. Yu HB, Finlay BB. The caspase-1 inflamma-
some: a pilot of innate immune responses. Cell Host Microbe
2008; 4: 198–208.
109. Bian ZM, Elner SG, Khanna H, et al. Expression
and functional roles of caspase-5 in inflammatory responses
of human retinal pigment epithelial cells. Invest Ophthalmol
Vis Sci 2011; 52: 8646–56.
110. Anelli T, Sitia R. Protein quality control in the early
secretory pathway. EMBO J 2008; 27: 315–27.
111. Shuda M, Kondoh N, Imazeki N, et al. Activation
of the ATF6, XBP1 and grp78 genes in human hepatocellular
carcinoma: a possible involvement of the ER stress pathway
in hepatocarcinogenesis. J Hepatol 2003; 38: 605–14.
112. Lai E, Teodoro T, Volchuk A. Endoplasmic reticulum
stress: signaling the unfolded protein response. Physiology
(Bethesda) 2007; 22: 193–201.
113. Bian ZM, Elner SG, Elner VM. Dual involvement
of caspase-4 in inflammatory and ER stress-induced apoptotic
responses in human retinal pigment epithelial cells. Invest
Ophthalmol Vis Sci 2009; 50: 6006–14.
114. Jing G, Wang JJ, Zhang SX. ER stress and apopto-
sis: a new mechanism for retinal cell death. Exp Diabetes Res
2012; 2012: 589589.
115. Rao RV, Castro-Obregon S, Frankowski H, et al. Cou-
pling endoplasmic reticulum stress to the cell death program.
An Apaf-1-independent intrinsic pathway. J Biol Chem 2002;
277: 21836–42.
116. Tinel A, Tschopp J. The PIDDosome, a protein
complex implicated in activation of caspase-2 in response
to genotoxic stress. Science 2004; 304: 843–6.
117. Verfaillie T, Salazar M, Velasco G, Agostinis P. Link-
ing ER stress to autophagy: potential implications for cancer
therapy. Int J Cell Biol 2010; 2010:930509.
118. Dong H, Chen L, Chen X, et al. Dysregulation of un-
folded protein response partially underlies proapoptotic activity
of bortezomib in multiple myeloma cells. Leuk Lymphoma
2009; 50: 974–84.
119. Hideshima T, Bradner JE, Wong J, et al. Small-
molecule inhibition of proteasome and aggresome function
induces synergistic antitumor activity in multiple myeloma.
Proc Natl Acad Sci USA 2005; 102: 8567–72.
120. Rodriguez J, Lazebnik Y. Caspase-9 and APAF-1 form
an active holoenzyme. Genes Dev 1999; 13: 3179–84.
Experimental Oncology ��� �������� ���� ��eptem�er���� �������� ���� ��eptem�er� ��eptem�er� ���
121. Shiozaki EN, Chai J, Rigotti DJ, et al. Mechanism
of XIAP-mediated inhibition of caspase-9. Mol Cell 2003;
11: 519–27.
122. Soung YH, Lee JW, Kim SY, et al. Mutational analysis
of proapoptotic caspase-9 gene in common human carcino-
mas. APMIS 2006; 114: 292–7.
123. Larsen BD, Rampalli S, Burns LE, et al. Caspase 3/
caspase-activated DNase promote cell differentiation by inducing
DNA strand breaks. Proc Natl Acad Sci USA 2010; 107: 4230–5.
124. Fujita E, Egashira J, Urase K, et al. Caspase-9 pro-
cessing by caspase-3 via a feedback amplification loop in vivo.
Cell Death Differ 2001; 8: 335–44.
125. Devarajan E, Sahin AA, Chen JS, et al. Down-regu-
lation of caspase 3 in breast cancer: a possible mechanism for
chemoresistance. Oncogene 2002; 21: 8843–51.
126. Palmerini F, Devilard E, Jarry A, et al. Caspase
7 downregulation as an immunohistochemical marker of co-
lonic carcinoma. Hum Pathol 2001; 32: 461–7.
127. Kischkel FC, Lawrence DA, Tinel A, et al. Death receptor
recruitment of endogenous caspase-10 and apoptosis initiation
in the absence of caspase-8. J Biol Chem 2001; 276: 46639–46.
128. Sprick MR, Rieser E, Stahl H, et al. Caspase-10 is recruit-
ed to and activated at the native TRAIL and CD95 death-inducing
signalling complexes in a FADD-dependent manner but can not
functionally substitute caspase-8. EMBO J 2002; 21: 4520–30.
129. Barbero S, Mielgo A, Torres V, et al. Caspase-8 as-
sociation with the focal adhesion complex promotes tumor
cell migration and metastasis. Cancer Res 2009; 69: 3755–63.
130. Stupack DG, Teitz T, Potter MD, et al. Potentiation
of neuroblastoma metastasis by loss of caspase-8. Nature
2006; 439: 95–9.
131. Barbero S, Barila D, Mielgo A, et al. Identification
of a critical tyrosine residue in caspase 8 that promotes cell
migration. J Biol Chem 2008; 283: 13031–4.
132. Torres VA, Mielgo A, Barila D, et al. Caspase 8 pro-
motes peripheral localization and activation of Rab5. J Biol
Chem 2008; 283: 36280–9.
133. Spaargaren M, Bos JL. Rab5 induces Rac-indepen-
dent lamellipodia formation and cell migration. Mol Biol Cell
1999; 10: 3239–50.
134. Pellinen T, Tuomi S, Arjonen A, et al. Integrin traf-
ficking regulated by Rab21 is necessary for cytokinesis. Dev
Cell 2008; 15: 371–85.
135. Finlay D, Vuori K. Novel noncatalytic role for cas-
pase-8 in promoting SRC-mediated adhesion and Erk signal-
ing in neuroblastoma cells. Cancer Res 2007; 67: 11704–11.
136. Krelin Y, Zhang L, Kang TB, et al. Caspase-8 defi-
ciency facilitates cellular transformation in vitro. Cell Death
Differ 2008; 15: 1350–5.
137. Teitz T, Wei T, Valentine MB, et al. Caspase 8 is deleted
or silenced preferentially in childhood neuroblastomas with
amplification of MYCN. Nat Med 2000; 6: 529–35.
138. Gonzalez-Gomez P, Bello MJ, Inda MM, et al. Dele-
tion and aberrant CpG island methylation of Caspase 8 gene
in medulloblastoma. Oncol Rep 2004; 12: 663–6.
139. Soung YH, Lee JW, Kim SY, et al. CASPASE-8 gene
is inactivated by somatic mutations in gastric carcinomas.
Cancer Res 2005; 65: 815–21.
140. Medema JP, Scaffidi C, Kischkel FC, et al. FLICE
is activated by association with the CD95 death-inducing
signaling complex (DISC). EMBO J 1997; 16: 2794–804.
141. Feig C, Tchikov V, Schutze S, Peter ME. Palmi-
toylation of CD95 facilitates formation of SDS-stable receptor
aggregates that initiate apoptosis signaling. EMBO J 2007;
26: 221–31.
142. Ganten TM, Haas TL, Sykora J, et al. Enhanced
caspase-8 recruitment to and activation at the DISC is criti-
cal for sensitisation of human hepatocellular carcinoma cells
to TRAIL-induced apoptosis by chemotherapeutic drugs. Cell
Death Differ 2004; 11 Suppl 1: S86–96.
143. Martin DA, Siegel RM, Zheng L, L enar-
do MJ. Membrane oligomerization and cleavage activates
the caspase-8 (FLICE/MACHalpha1) death signal. J Biol
Chem 1998; 273: 4345–9.
144. Scaffidi C, Schmitz I, Krammer PH, Peter ME. The
role of c-FLIP in modulation of CD95-induced apoptosis.
J Biol Chem 1999; 274: 1541–8.
145. Kavuri SM, Geserick P, Berg D, et al. Cellular
FLICE-inhibitory protein (cFLIP) isoforms block CD95- and
TRAIL death receptor-induced gene induction irrespective
of processing of caspase-8 or cFLIP in the death-inducing
signaling complex. J Biol Chem 2011; 286: 16631–46.
146. Kruidering M, Evan GI. Caspase-8 in apoptosis: the
beginning of “the end”? IUBMB Life 2000; 50: 85–90.
147. Jin Z, Li Y, Pitti R, et al. Cullin3-based polyubiquiti-
nation and p62-dependent aggregation of caspase-8 mediate
extrinsic apoptosis signaling. Cell 2009; 137: 721–35.
148. Peng C, Cho YY, Zhu F, et al. Phosphorylation of cas-
pase-8 (Thr-263) by ribosomal S6 kinase 2 (RSK2) mediates
caspase-8 ubiquitination and stability. J Biol Chem 2011;
286: 6946–54.
149. Christian PA, Fiandalo MV, Schwarze SR. Possible
role of death receptor-mediated apoptosis by the E3 ubiquitin
ligases Siah2 and POSH. Mol Cancer 2011; 10: 57.
150. Brooks AD, Jacobsen KM, Li W, et al. Bortezomib
sensitizes human renal cell carcinomas to TRAIL apoptosis
through increased activation of caspase-8 in the death-
inducing signaling complex. Mol Cancer Res 2010; 8: 729–38.
Copyright © Experimental Oncology, 2012
|