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

J. Pathol.

187: 127137 (1999)

APOPTOSIS AND THERAPY


. . *
Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, U.S.A.

SUMMARY
The dogma that antineoplastic treatments kill tumour cells by damaging essential biological functions has been countered by the
notion that treatment itself initiates a programmed cellular response. This response often produces the morphological features of
apoptosis and is determined by a network of proliferation and survival genes, some of which are differentially expressed in normal and
malignant cells. Correspondingly, mutations that interfere with the initiation or execution of apoptosis may produce tumour-cell drug
resistance. Remarkably, many of the genes that modulate apoptosis in response to cytotoxic drugs also affect apoptosis during tumour
development; hence, the process of apoptosis provides a conceptual framework for understanding how cancer genes can influence the
outcome of cancer therapy. Although the relative contribution of apoptosis to radiation and drug-induced cell death remains
controversial, clinical studies have associated anti-apoptotic mutations with treatment failure. While careful preclinical and clinical
studies will be necessary to resolve this point, our current understanding of apoptosis should facilitate the design of rational new
therapies. Copyright  1999 John Wiley & Sons, Ltd.
KEY WORDSanti-cancer treatment; resistance; DNA damage; clinical outcome; p53; programmed cell death

APOPTOSIS IS PROGRAMMED CELL DEATH Receptor-linked apoptosis


Apoptosis is a process of cell death that was initially At the molecular level, the best understood cell death
described by its morphological characteristics, including pathways involve those initiated by death receptors,
cell shrinkage, membrane blebbing, chromatin conden- including Fas/CD95, tumour necrosis factor (TNF)
sation, and nuclear fragmentation.1 Apoptosis acts in receptor type 1 (TNFR1), DR3, DR4 and DR5.8,11,12
embryonic development and tissue homeostasis,2 and For example, binding of TNF to TNFR1 results in
can also be induced by pathological insults. The fact that its trimerization and the subsequent recruitment of
apoptosis is controlled by genes reveals its clinical the signal transducing molecules TRADD (TNFR1-
relevance: as in other developmental or homeostatic associated death domain protein) and FADD
processes, cell death can be disrupted by mutations. (Fas-associating protein with death domain) through
Hence, a variety of disease states involve either hyper- conserved protein interaction regions known as death
activation or suppression of apoptosis, including domains. This complex then associates with caspase-8, a
neurodegenerative diseases and cancer, respectively.3 cell death protease that becomes activated in the
While the terms apoptosis and programmed cell death receptor/adapter complex. Activated caspase-8 initiates
are often used synonymously, cell deaths can be pro- a protease cascade that cleaves cellular targets and
grammed without undergoing the morphological char- results in apoptotic cell death.10,13,14 Hence, disruption
acteristics of apoptosis4,5 and can occur long after the of FADD can prevent activation of caspase-8, thereby
death-initiating event.6 producing defects in receptor-mediated cell death (e.g.
A comprehensive overview of apoptosis is beyond the ref. 15). Importantly, TNF-induced apoptosis can be
scope of this discussion (for review, see refs 2 and 710). modulated by TRADDs ability to bind TRAF2 (TNF
However, as a general summary, most apoptotic path- receptor-associated factor), which facilitates nuclear
ways involve a sensor that detects a death-inducing factor kappa B (NF-B)-mediated cell survival.16,17
signal, a signal transduction network, and an execution
machinery that actively carries out the process of cell DNA damage-induced apoptosis
death. The probability with which each signal activates
the execution machinery is controlled by a series of DNA damage can also induce apoptosis through a
regulators that, in turn, are influenced by cellular con- central player, p53, although p53-independent pathways
text. Hence, stimuli which induce apoptosis in one clearly exist. Current evidence suggests that DNA strand
setting may not in another. In order to lay the ground- breaks are sensed by kinases such as the DNA-
work for therapy-induced apoptosis, two different dependent protein kinase (DNA-PK) or the ataxia-
apoptotic pathways are outlined below. telangiectasia-mutated gene product (ATM) leading to
phosphorylation and activation of p53.18,19 p53 trans-
*Correspondence to: S. W. Lowe, Cold Spring Harbor Laboratory, mits the apoptotic signal by a complicated mechanism
1 Bungtown Road, P.O. Box 100, Cold Spring Harbor, NY 11724, that involves, at least in part, its ability to transactivate
U.S.A. E-mail: lowe@cshl.org target genes such as bax and a series of p53-inducible
Contract grant sponsor: Dr Mildred Scheel Cancer Foundation. genes (PIGs).20,21 The next step is poorly understood,
Contract grant sponsor: National Cancer Institute; Contract grant but may involve changes in mitochondrial membrane
number: CA13106. potential and release of cytochrome c.22 In any case,
CCC 00223417/99/01012711$17.50
Copyright  1999 John Wiley & Sons, Ltd.
128 C. A. SCHMITT AND S. W. LOWE

once in the cytosol, cytochrome c can interact in a genes should create selective pressure for disruption of
multi-protein complex with Apaf1 and pro-caspase-9, apoptosis during tumour progression. That pro- and
leading to caspase-9 activation and the initiation of a anti-apoptotic mutations co-operate during tumour
protease cascade similar to that described above.23,24 development has extensive support from animal
Concordantly, inactivation of caspase-9 disrupts apop- models.33,4749
tosis in situations where p53 is required for programmed
cell death.25,26
Apoptosis induced by DNA-damaging agents can be APOPTOSIS LINKS CANCER GENETICS WITH
dramatically affected by cellular context, including tissue CANCER THERAPY
type and genetic background (see below). One of the
most important modulators is Bcl-2, owing to its ability The importance of programmed forms of cell death is
to affect cytochrome c release from mitochondria9,27 not restricted to tumour development, but has relevance
and/or modulate the Apaf1/caspase-9 interaction.24,28,29 for cancer therapy and mechanisms of drug sensitivity
Given these complexities, the scenario described above and resistance. However, until recently, the tumour-
must be considered a conceptual framework for further specific toxicity of most anticancer agents was attributed
research and discussion. In reality, the apoptotic net- to their debilitating effects on actively proliferating cells.
work is much too complicated to be accurately described The interaction of an agent with its primary intercellular
by simple linear pathways. target was presumed to produce massive cellular dam-
age, thereby disrupting vital metabolic functions and
causing catastrophic cell death. Based on this view,
APOPTOSIS IN NEOPLASIA efforts to understand drug resistance have focused on
mutations that disrupt or prevent the drugtarget inter-
Failure to maintain an appropriate balance of cell action. More recently, it has become apparent that most
numbers is a hallmark of neoplasia. Apoptosis is the anticancer agents induce apoptosis. Thus, the effective-
counterweight to proliferation, and disruption of ness of anticancer drugs may not simply reflect their
apoptosis is a key event in tumourigenesis. For ability to cause cellular damage, but also the cells ability
example, Bcl-2 was cloned by virtue of its high frequency to detect and respond to this damage.1,50 Conversely,
of constitutive activation following translocation in since drugs with distinct primary targets can induce
follicular lymphomas.30 Indeed, several key players in apoptosis through similar mechanisms, mutations
apoptosis were originally identified by their role in apoptotic programmes can produce multi-drug
in oncogenic transformation or as genes mutated in resistance.50,51
human tumours.31,32 Moreover, inactivation of p53 Cytotoxic anticancer agents are effective only when
is an extremely common event in human tumours: tumour cells are more readily killed than the surround-
while p53 functions in several cellular processes, its ing normal tissue. If the efficacy of these agents is partly
role in apoptosis clearly contributes to tumour determined by their ability to induce apoptosis, then
development.33,34 tumour cells must be more susceptible to apoptosis than
Paradoxically, the forces of tumour development can the normal tissue from which they arose. Interestingly,
actually promote apoptotic cell death, owing largely to pro-oncogenes such as E1A and c-myc dramatically
cellular mechanisms that couple excessive proliferation increase cellular radio- and chemosensitivity in cultured
to apoptosis. Specifically, studies using cultured cells cells and xenografts.5153 Since changes induced by these
demonstrate that many mitogenic oncogenes (e.g. myc, oncogenes are unique to malignant cells, it has been
E1A, E2F-1, cdc25, ras) force uncontrolled proliferation suggested that this effect contributes to the therapeutic
but at the same time can enhance apoptosis.2 Of note, index of cytotoxic agents.51,54 Conversely, disruption of
apoptosis in oncogene-expressing cells is markedly p53 or overexpression of Bcl-2 can produce pleiotropic
potentiated by growth inhibitory signals prevalent in resistance to anticancer agents by preventing apoptosis
developing tumours, including low concentrations of (see below).
survival/growth factors35,36 and low oxygen (hypoxia).37 It is remarkable that the same genetic alterations that
Hence, these developing tumour cells strike a balance influence apoptosis during tumour development also
between proliferation and apoptosis which is dependent modulate drug sensitivity and resistance. For example,
on environmental cues. c-myc enhances apoptosis in low concentrations of sur-
Promotion of apoptosis by oncogenes may function as vival factors or oxygen and following treatment with
a cellular fail-safe mechanism that limits tumour devel- diverse cytotoxic agents;35,37,53 conversely, loss of p53
opment. Consistent with this view, molecules that modu- and overexpression of Bcl-2 suppress apoptosis induced
late oncogene-induced cell death in cultured cells are by oncogenes and depletion of survival factors, hypoxia,
commonly mutated in tumours. For example, the p53 and cytotoxic drugs.35,37,39,51 As such, the concept of
tumour suppressor is activated by a variety of mitogenic apoptosis provides an attractive link between the forces
oncogenes. p53 is necessary for oncogene-induced apop- of tumour development and factors influencing drug
tosis in a variety of settings,3841 and recent studies have sensitivity and resistance. Indeed, this association may
demonstrated that yet another tumour suppressor, explain the phenomenon of de novo drug resistance in
p19ARF, mediates this effect.4246 Moreover, oncogene- advanced solid tumours: selection against apoptosis
induced cell death is efficiently blocked by Bcl-2.2,9 during tumour development can simultaneously produce
Importantly, these data predict that pro-apoptotic onco- drug-resistant cells.
Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
APOPTOSIS AND THERAPY 129

APOPTOSIS AND THE CLASSIC VIEW OF engineered cells to ask whether controlled alterations in
DRUG TOXICITY the expression of apoptotic genes can increase or
decrease cellular sensitivity to radiation or various
Although the role of apoptosis in treatment is a recent chemotherapeutic agents. Although these systems are
concept, the predictions arising from this model can be artificial, they clearly demonstrate that defects in apop-
integrated with classic views of drug action to explain tosis can produce resistance to anticancer agents. A
previous observations. The classic view is based on the sampling of some apoptotic genes and their relationship
drugtarget interaction and its relation to a distinct to drug toxicity under preclinical conditions are
cell-cycle phase. For example, pharmacological S-phase discussed below.
synchronization was thought to sensitize tumour cells to
anticancer drugs most toxic to DNA synthesis or the
replication machinery.55,56 Apoptosis links the cell-cycle The p53 tumour suppressor
phase preference of anticancer agents to a DNA The most well-studied gene in terms of its impact on
damage-checkpoint-initiated cell death programme. DNA damage-induced cell death is p53. Early studies
Given an intact checkpoint control, drug-mediated cell linking p53 loss with radiation and drug resistance came
death is now understood as a checkpoint response to from model systems using p53 knockout mice. For
DNA damage. example, loss of p53 markedly attenuates radiation
DNA damage-induced cell death can occur immedi- and drug-induced apoptosis in a variety of murine
ately or be delayed. The majority of drugs target DNA tissues.6065 Importantly, oncogenically transformed
synthesis, whereas others are more related to cell metab- cells derived from p53-deficient fibroblasts are cross-
olism and division. Hence, some drugtarget interactions resistant to a variety of cytotoxic agents both in cell
mediate immediate genomic injury while others cause culture and in nude mice.51,66 In addition, studies using
delayed genotoxicity, shedding light on the phenomenon the more complicated genetic background of tumour-
of acute versus delayed apoptosis.6 The acuteness and derived lines also found a high dependency of most
severity of the drug-induced DNA damage are not only clinically established anticancer drugs on functional
related to the drugtarget interaction, but also depend p53.67,68 Moreover, many studies document an increase
on the dose and the time [the area-under-the-curve in radiation and chemosensitivity following transfer of
(AUC)]. While low doses resulting in minor, repairable p53 genes into p53-deficient tumour cells in culture and
damage may only delay cell-cycle progression, higher in xenografts.69,70
doses leading to severe DNA damage can initiate
apoptosis or even necrosis. Furthermore, the onset
of apoptosis is dose-dependent: topoisomerase in-
Bcl-2 family
hibitors like adriamycin execute cell death directly in the
affected S-phase without proceeding to the following Overexpression of Bcl-2 in human tumour cells pro-
checkpoint; however, lower doses result in apoptosis duces resistance to a wide variety of toxic agents.71
in subsequent cell-cycle phases following checkpoint Unlike classic modes of drug resistance, Bcl-2 does not
controls.57 inhibit drug uptake or the ensuing cellular damage but
Thus, the present concept of drug-mediated cyto- instead blocks apoptosis.72 Experiments using antisense
toxicity, improved by new insights into programmed cell technology to interfere with Bcl-2 function have shown
death, dependency on checkpoint control, AUC, and the an inverse correlation between chemosensitivity and
kind of genomic injury, reflects the complexity of the Bcl-2 levels, indicating that endogenous Bcl-2 contrib-
cellular response to anticancer treatment. While this utes to the drug-resistant state.73 This also suggests that
concept provides an exciting new perspective on how agents designed to disrupt Bcl-2 function might be
anticancer drugs work, it also underlines that individual effective therapeutics.
effects are highly dependent on cell type, specific treat- Bcl-2 is the founding member of an expanding gene
ment, and environmental conditions. In addition, it family74 that includes other anti-apoptotic molecules
elucidates why modified treatment conditions may cause such as Bcl-xL as well as pro-apoptotic members such as
apoptotic cell death even in a formerly resistant setting. bax. Preclinical studies have shown that Bcl-xL over-
This aspect may be important for the understanding expression can confer resistance to anticancer drugs and
of high-dose strategies, which are able to override radiotherapy.75,76 By contrast, other members of the
resistance.58 Bcl-2 family like bax, bcl-xS, and bak appear to promote
apoptosis.77 While bax deficiency mediates drug resist-
ance,78 Bax overexpression enhances radiation sensitiv-
APOPTOTIC GENES AND ANTICANCER AGENT ity or sensitizes to anticancer drugs.79,80 The activity of
CYTOTOXICITY the Bcl-2 family members involves their ability to form
homodimeric or heterodimeric complexes. Several
Studies performed nearly 25 years ago noted that studies suggest that cell viability following an apoptotic
apoptosis accompanies tumour regression during cancer stimulus is associated with the Bcl-2/Bax ratio,81 but
therapy.59 However, the view that the integrity of apop- exactly how these complexes regulate apoptosis is
totic pathways can affect the outcome of cancer therapy unclear. Interestingly, antitubulin agents like paclitaxel
stems from basic and preclinical studies on the apoptotic or vincristine induce Bcl-2 hyperphosphorylation and
genes themselves. Typically, these studies use genetically inhibit its ability to heterodimerize with Bax.82
Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
130 C. A. SCHMITT AND S. W. LOWE

Fig. 1DNA damage and oncogenes initiate pathways involved in apoptosis, cell-cycle arrest,
or survival. p53 is activated by mitogenic oncogenes such as E1A and myc with p19ARF as an
intermediary, or by kinases such as the DNA-dependent protein kinase (DNA-PK) or the
product of the ataxia-telangiectasia gene (ATM) in response to DNA damage. The apoptotic
p53 pathway includes bax, cytochrome c release, activation of Apaf1, and the caspase family,
while the p53-target p21 mediates cell-cycle arrest. Oncogenes may also contribute to p53-
independent apoptosis by sensitization of the Fas/FasL death receptor pathway or by other
pro-apoptotic pathways. Induction of NF-B results in subsequent activation of survival genes

Death receptors and therapy the contribution of dysregulated caspases to anticancer


treatment resistance have not yet been published. How-
Some studies suggest that classic anticancer agents
ever, exogenous inhibition of caspases has conferred
require the FasL/Fas system to induce cell death,
resistance to different anticancer drugs.91 The potential
although this remains highly controversial. For example,
adriamycin-induced apoptosis of human T-cell leukae- impact of caspases and their activators on resistance is
mia cells can be blocked by anti-Fas antibodies or also supported by knock-out mouse models with germ-
up-regulation of FasL expression.83,84 In contrast, both line disruptions of the Apaf1, caspase-3 or caspase-9
Fas-resistant Jurkat cells and FADD-deficient cells are gene, which are resistant to various apoptotic stimuli in
defective in Fas-induced death but not chemotherapy- different tissues.4,25,26,92
induced apoptosis.15,85 In principle, cross-resistance
against Fas- and drug-induced apoptosis may be due to
escape mechanisms of shared downstream components Cell survival pathways
of the apoptotic machinery8587 (Fig. 1). Alternatively, Extracellular survival factors play an important role
the relationship may be more direct. In this regard, p53 in maintaining cell viability.93 Although these cell sur-
can transcriptionally activate DR5 in some contexts, vival pathways may not directly act downstream of
suggesting a mechanism whereby these pathways anticancer drugs, recent studies indicate that they can
could facilitate drug-induced death.88,89 Still, much synergize with anti-apoptotic mutations to reduce
work is needed to understand when and how these chemosensitivity.94 One critical factor in cell survival is
death receptor pathways contribute to radio- and NF-B, a transcription factor that is induced by a
chemosensitivity. variety of signals including oncogenes, cytokines, and
anticancer drugs.9599 Remarkably, inactivation of
NF-B dramatically enhances apoptosis following treat-
The terminal apoptotic cascade ment with TNF- and certain anticancer agents,100
At the final stage of apoptosis, the different death owing, at least partially, to NF-Bs ability to modulate
pathways converge to activate members of the caspase the expression of components of death receptor com-
family.90 Preclinical studies on tumour cells analysing plexes.17 Another survival signal cascade, the PI3K/Akt
Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
APOPTOSIS AND THERAPY 131

pathway, may link extracellular survival signals to the resistance in human tumours. However, not all studies
apoptotic machinery by phosphorylation of specific cell find correlations between sensitivity to apoptosis and
death-related molecules such as Bad.98 Recent evidence radio- and chemosensitivity. For example, some studies
suggests that the PI3K/Akt cell survival pathway can be fail to correlate p53 mutations with reduced anticancer
negatively regulated by the PTEN tumour suppressor, a agent toxicity and, indeed, others see loss of p53 enhanc-
lipid phosphatase that itself induces apoptosis.101,102 ing cell death.118,119 Moreover, in some settings, Bcl-2
Since PTEN is mutated in diverse tumour types, it will does not enhance cell viability following irradiation or
be interesting to determine whether loss of PTEN can drug treatment and in fact, some studies suggest that
influence the cytotoxicity of anticancer agents. Bcl-2 expression is indicative of a good prognosis and
outcome in breast cancer.120,121 These results confound
our understanding of the overall impact of apoptosis
APOPTOTIC MUTATIONS AND DRUG on cancer therapy. Although more research will be
RESISTANCE IN PATIENTS needed to resolve these inconsistencies, both methodo-
logical and technical problems contribute to this
Although laboratory studies like those described confusion.
above have established that mutations in apoptotic
programmes produce treatment-resistant tumours, the
Apoptosis and clonogenic survival
extent to which similar mechanisms operate in cancer
patients remains to be established. With varying degrees In some contexts, anti-apoptotic changes such as
of success, investigators have attempted to extend these Bcl-2 overexpression simply delay radiation or drug-
preclinical observations to patients. These studies can be induced apoptosis but do not enhance clonogenic
extremely difficult, as suitable clinical material is often survival, raising the possibility that non-apoptotic
hard to obtain and the genetics of human tumours are so mechanisms are ultimately responsible for cell
variable as to preclude simple comparisons. Neverthe- death.122124 While it is likely that delayed cell death can
less, emerging evidence suggests that mutations that contribute to the toxicity of anticancer agents, several
suppress apoptosis may indeed contribute to treatment caveats are worth mentioning. While the clonogenic
failure in human cancer. In fact, p53 mutations were survival assays are often considered the gold standard,
found to be associated with poor prognosis, de novo or recent evidence of the role of p21 in response to
acquired resistance, and relapse in a broad field of solid irradiation indicates that clonogenic survival may not
and haematological malignancies.67,103110 accurately reflect in vivo cell death and regrowth of
Recent data underline the link between extragenic long-term arrested cells.125 It is possible that autocrine
alterations of the apoptotic p53 pathway and impaired or paracrine survival factorsinfluenced by cell density
response to therapy. Mdm2 overexpression, resulting in or micro-environmentcan impact the mechanisms of
p53 degradation, correlates with early treatment failure drug toxicity. Indeed, a recent study has confirmed that
in childhood acute lymphoblastic leukaemia.111 Loss-of- Bcl-2 has dramatically different effects on the long-term
function mutations of the p53-target bax confer resist- survival of lymphoma cells depending on the culture
ance in haematopoietic malignancies.112 Expression conditions used in the clonogenic assay.94
levels of Bax are positively correlated with response to Despite the exceptions, a large number of studies
cytotoxic treatment and survival in breast cancer indicate that apoptotic genes can affect clonogenic sur-
patients.113,114 Increased Bcl-2/Bax ratios were observed vival and, as indicated above, can affect tumour regres-
in B-cell chronic lymphocytic leukaemia cells and were sion in animal models or patients. However, in addition
particularly pronounced in leukaemia cells from patients to acute apoptosis detectable in short-term survival
non-responsive to chemotherapy.115 Overexpression of assays, other forms of programmed cell death can
the anti-apoptotic long form of Bcl-x correlated strongly include delayed apoptosis and senescence-like tumour
with decreased response rates in multiple myeloma cell arrest.6,126 Genes that affect these latter processes
patient groups treated with different classes of anti- are poorly understood; therefore definitive experiments
cancer agents.116 Although fewer studies have focused are difficult to perform.
on apoptotic genes outside the p53 axis and Bcl-2 family,
overexpression of Bcr-Abl, a fusion protein arising from
Technical and conceptual considerations
the Philadelphia translocation in leukaemia, has been
shown to suppress apoptosis and reduce the therapeutic What other factors could explain a lack of correlation
outcome of relapsed childhood acute lymphoblastic between specific apoptotic genes and chemosensitivity
leukaemia.117 and/or therapeutic outcome in patients? A major issue
relates to the technical and conceptual problems that
accompany correlative studies examining the relation-
TO WHAT EXTENT DOES APOPTOSIS ship between single apoptotic genes, chemosensitivity,
CONTRIBUTE TO THE OUTCOME OF and/or therapeutic outcome. The problem can be illus-
CURRENT CANCER THERAPIES? trated by studying the issues associated with understand-
ing the role of one apoptotic gene, p53, in therapeutic
The above studies provide direct evidence that apop- outcome.
tosis can impact on therapy and correlative evidence In correlative studies using human cell lines or
that anti-apoptotic mutations can contribute to drug tumours, the ability to make statistically significant
Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
132 C. A. SCHMITT AND S. W. LOWE

comparisons requires that samples are accurately setting, leading to comparable and treatment-assessable
classified based on p53 status. Most clinical studies rely tumours. Since xenograft models have their own restric-
on p53 immunohistochemistry as a surrogate marker for tions rooted in the grafthost collision mice harbouring
p53 mutations, but evaluation is subjective and the germ-line mutations in apoptotic genes may be useful.
premise that detectable p53 reflects mutated p53 contrib- For example, we addressed the question of treatment-
utes to a potential misclassification of a subset of mediated apoptosis in the E-myc transgenic mouse.131
tumours.127 Direct sequencing of p53 is more accurate When crossed to mice heterozygous for p53, the F1
and can identify associations between p53 mutations and generation with wild-type p53 developed B-cell lym-
clinical parameters where immunohistochemistry can- phoma within the usual latency. However, lymphoma in
not,127 but is subject to normal cell contamination. In the p53 +/ mice occurred much faster. While the p53
studies with limited material, misclassification of even a wild-type tumours responded to radiation or adria-
few tumours can affect the conclusions. Even with mycin, two thirds of the p53 +/ derived tumours were de
accurate diagnosis of p53 status, tumour classification is novo-resistant and the responsive subgroup relapsed
confounded by the observation that not all p53 muta- after very short remission periods (C. Schmitt, R. R.
tions are functionally equivalent.128 Moreover, a large Wallace-Brodeur, M. E. McCurrach, and S. W. Lowe,
number of extragenic mutations can abolish p53 activity unpublished observations). This is exactly what one
in tumours that retain a wild-type p53 gene. Many such would predict with respect to p53s role in DNA
alterations are common in human tumours, including damage-mediated apoptosis and underlines the validity
HPV-E6 in cervical carcinomas, Mdm2 in a variety of of the model. In principle, this model or others may
tumours, and bax in HNPCC-type colon cancer.128130 become useful in genetically-oriented drug-discovery
In these settings, p53 status would be misclassified by programmes.
current technology. Given these caveats, it is impossible
to interpret negative results in clinical studies.
Finally, basic studies indicate that the role of p53 in NEW THERAPEUTIC APPROACHES
chemosensitivity is context-dependent. Specifically, the
prediction that p53 mutations should promote drug Irrespective of the relative extent to which apoptosis
resistance is largely based on the observations that contributes to the success of present-day cancer thera-
DNA-damaging agents can activate p53 to promote pies, our current understanding of apoptosis leaves no
apoptosis. However, p53 has additional biological doubt that these programmes can be manipulated to
activities, including the ability to promote cell-cycle produce massive changes in cell death. Hence, the genes
checkpoints and senescence, and little is known about and proteins that control apoptosis provide exciting new
how these activities modulate chemosensitivity. Perhaps targets for rational drug discovery. Of course, as indi-
positive correlations between p53 and chemosensitivity cated above, the clinic is filled with potent inducers of
will only occur where p53s apoptotic function predomi- tumour-cell apoptosismolecules which are also highly
nates. Whether p53 functions in an apoptotic mode will toxic due to their deleterious effects on normal cells.
depend on several criteria, including agent, dose, tissue Thus, strategies designed to identify novel compounds
type, and mutational background of the tumour cells that simply induce apoptosis are unlikely to provide
(see above). These are not understood in sufficient detail improvements over current therapies; rather, new strat-
to make accurate predictions as to p53s role in a given egies must exploit apoptosis in a selective manner. A
setting. sampling of current approaches to exploit apoptosis will
The question is: to what extent does apoptosis con- be discussed below.
tribute to the clinical outcome? Although p53 can be
used as an example to address the question of drug
sensitivity and resistance, the same caveats apply to Selective delivery
clinical studies investigating other single genes that One strategy to achieve selectivity is through tumour-
control apoptosis. Besides the difficulty in assessing specific delivery of apoptosis-inducing molecules. For
pathways instead of single genes, outcome not only example, viral vectors expressing apoptotic genes can be
depends on the therapeutic impact on cell death, but is directly introduced into the tumour, minimizing their
also the result of the tumour growth minus apoptosis uptake by normal cells (see, for example, refs 69, 132
equation. Progression under treatment may reflect resist- 134). This approach has proven particularly effective for
ance, but can occur in a fast-growing, sensitive tumour p53: intratumoural injection of p53-expressing adeno-
as well. On the one hand, poor prognosis can be solely viruses can induce apoptosis directly and synergizes with
due to an aggressive histological subtype, potentially cytotoxic anticancer agents to promote tumour regres-
due to genetic alterations of apoptotic genes. On the sion with minimal additional toxicity.69,135,136 Indeed,
other hand, it can be a result of an impaired treatment strategies using this approach are currently undergoing
response due to non-apoptotic mechanisms attenuating clinical trials.137 Alternatively, tumour-specific changes
the therapeutic efficacy. Between these extremes, a sub- in transcriptional regulation may allow selective expres-
stantial contribution of apoptotic dysregulation to treat- sion of apoptosis-inducing genes. One elegant approach
ment failure is likely, however, in patients technically takes advantage of the fact that E2F transcription
hard to prove. factors are often hyperactivated in tumour cells, owing
A promising approach to monitor drug-induced to defects in the Rb pathway. Toxic genes can be driven
apoptosis in vivo is animal models with a defined genetic from artificial promoters harbouring E2F binding sites,
Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
APOPTOSIS AND THERAPY 133

leading to preferential activation of the apoptosis gene E1A mutants unable to bind Rb (but retaining the
in tumours.138 Finally, since adenovirus inefficiently p300/CBP interaction) promote apoptosis in Rb-
infects normal haemopoietic cells, adenoviral vectors deficient cells but not in normal cells.148 In principle,
expressing apoptotic genes can be used to purge the these E1A mutants, or small molecules which mimic
bone marrow of metastatic tumour cells.139,140 their action, may provide tumour-specific anticancer
agents by exploiting the fact that the Rb pathway is
disrupted in the majority of human cancers.
Tumour-specific alterations in apoptotic programmes
Although mutations in death receptors seem to be
Since tumour evolution involves genetic changes that very rare events in human tumours and, so far, no
both positively and negatively affect apoptosis, the regu- cancer-related mutations of their ligands have been
lation of apoptosis in tumour cells may be fundamen- described,149151 changes accompany tumourigenesis
tally different from normal tissues. In many instances, have dramatic effects on the regulation of these path-
apoptosis has been disabled by dominant oncogenes, ways. Mitogenic oncogenes like myc and E1A can
and disrupting their function can produce remarkable increase sensitivity to Fas and TNF- in cultured
increases in cell death. In chronic myelogenous leukae- cells,152,153 and many human tumours have alterations
mia (CML), a chromosomal translocation leading to the in TRAIL-mediated death pathways. TRAIL is a TNF-
production of the Bcr-Abl fusion protein is critical for related protein that initiates a p53-independent cell
tumour evolution and may act by suppressing apopto- death programme by binding to its receptors DR4 or
sis.141 Since this translocation is unique to these tumour DR5. Normal cells are resistant to TRAIL-induced
cells, the gene or protein may represent a unique target. apoptosis, apparently because normal cells express
Indeed, inactivation of Bcr-Abl using hammerhead decoy receptors that compete with DR4 and DR5 for
ribozymes provides evidence that this approach can TRAIL but do not transmit a death-inducing signal.154
enhance apoptosis.142 Through an unknown mechanism, the expression of the
In many instances, tumour evolution leads to decoy receptor 1 seems to be widely lost on tumour cells,
up-regulation of one or more members of the Bcl-2 making them exquisitely susceptible to TRAIL-
family. Although targeting these proteins with small mediated cell death.154 Interestingly, DR5 is a p53-
molecules is the ultimate goal, substantial progress has responsive gene; hence, combination therapy with
been made using antisense inhibition;143 in fact, anti- TRAIL and classic cytotoxics may be particularly
sense bcl-2 oligonucleotides can produce substantial effective in treating tumours with functional p53.88
tumour regression when combined with chemotherapy Consequently, the use of TRAIL or other strategies
in xenograft models.144 Another gene-based approach to exploiting these well-characterized apoptotic pathways
target this family uses bcl-xS, a dominant-negative are promising avenues for drug development.
repressor of Bcl-2 and Bcl-xL. Adenovirus-mediated
gene transfer of bcl-xS can synergize with chemo-
Viral proteins
therapy145 and promote tumour regression in
xenografts,146 but produces minimal apoptosis when In addition to E1A, a number of other viral proteins
introduced into normal epithelial cells.147 Why would are in preclinical studies as selective inducers of apopto-
such therapeutic strategies be selective? The answer is sis. The adenovirus E4orf4 protein is responsible for
not known, but perhaps inactivation of these proteins p53-independent apoptosis observed during adenovirus
reveals the pro-apoptotic forces of certain oncogenic infection and can produce rapid cell death when
mutations (see earlier discussion). expressed in tumour cells,155 although the specificity of
Indeed, preclinical studies have demonstrated that it is this effect remains to be determined. Another intriguing
possible to harness directly the pro-apoptotic forces viral protein is apoptin, the VP3 product of chicken
produced by certain oncogenic mutations. As mentioned anaemia virus (CAV). This protein is responsible for the
earlier, oncogenes like c-myc and inactivation of tumour cytopathic effects of CAV156 and induces apoptosis in
suppressors such as Rb force proliferation but at the tumour cells but not normal cells.157 Although its mech-
same time promote apoptosis, presumably as a cellular anism of action remains unknown, apoptin-induced
safe-guard against tumourigenesis. These pro-apoptotic apoptosis is independent of p53 and is enhanced by
activities would be even more potent if it were not for Bcl-2.158 At the very least, the remarkable effects of
the survival genes simultaneously induced by the family apoptin underscore the point that selective induction of
of NF-B transcription factors, as reported for the apoptosis in tumour cells is achievable.
adenoviral oncoprotein E1A95 or for oncogenic Ras.98
Hence, inactivation of NF-B function with I-B can
enhance oncogene-induced apoptosis as well as cellular Chemoprotection
radio- and chemosensitivity.97,100 In a similar approach, As indicated above, there would be no truly drug-
studies on the adenovirus E1A oncogene provide proof resistant cell if cytotoxic agents could be applied in
of concept data that tumour-specific mutations can be sufficient dose. However, toxicity to normal tissues
exploited to selectively enhance apoptosis in tumour presents a major limitation in the use of current
cells. E1A promotes apoptosis and enhances chemo- apoptosis-inducing cytotoxic drugs. Most tissues are
sensitivity through a dual mechanism that involves relatively insensitive to apoptosis; however, normal
inactivation of the retinoblastoma protein and binding haematologic cells and certain stem cells of the small
the p300/CBP transcription co-activators. As a result, intestine are remarkably sensitive to apoptosis following
Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
134 C. A. SCHMITT AND S. W. LOWE

radiation or chemotherapy (see above). Since cell loss 6. Tang C, Willingham MC, Reed JC, et al. High levels of p26BCL-2
oncoprotein retard taxol-induced apoptosis in human pre-B leukemia cells.
from these tissues accounts for the most debilitating Leukemia 1994; 8: 19601969.
side-effects of cancer therapy, exogenous interventions 7. Adams JM, Cory S. The Bcl-2 protein family: arbiters of cell survival.
conferring resistance to normal cells might allow the Science 1998; 281: 13221326.
8. Ashkenazi A, Dixit VM. Death receptors: signalling and modulation.
administration of higher drug doses to combat the Science 1998; 281: 13051308.
tumour. In an ironic twist of thinking, several groups 9. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998; 281:
have attempted to introduce classical drug resistance 13091312.
10. Thornberry NA, Lazebnik Y. Caspases: enemies within. Science 1998; 281:
genes into normal cells as a chemoprotective measure. 13121316.
For example, the ex vivo transduction and subsequent 11. Golstein P. Cell death: TRAIL and its receptors. Curr Biol 1997; 7:
transplantation of haematopoietic progenitors with R750R753.
12. Nagata S. Apoptosis by death factor. Cell 1997; 88: 355365.
MDR1 or the aldehyde dehydrogenase gene suggest that 13. Cohen GM. Caspases: the executioners of apoptosis. Biochem J 1997; 326
such strategies might be effective.159,160 (Pt 1): 116.
Based on our understanding of apoptosis in these 14. Shu HB, Halpin DR, Goeddel DV. Casper is a FADD- and caspase-
related inducer of apoptosis. Immunity 1997; 6: 751763.
chemosensitive tissues, it should be possible to block this 15. Yeh WC, Pompa JL, McCurrach ME, et al. FADD: essential for embryo
cell death. Indeed, studies using mouse models have development and signalling from some, but not all, inducers of apoptosis.
clearly documented the importance of p53 for apoptosis Science 1998; 279: 19541958.
16. Takeuchi M, Rothe M, Goeddel DV. Anatomy of TRAF2. Distinct
in thymocytes, bone marrow, and intestinal stem domains for nuclear factor-kappaB activation and association with tumor
ells.60,161,162 Furthermore, inactivation of p53 allows necrosis factor signalling proteins. J Biol Chem 1996; 271: 19 93519 942.
recovery of intestinal crypts following therapy with high 17. Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS Jr.
NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1
dose 5-fluorouracil.163 Consequently, agents which sup- and c-IAP2 to suppress caspase-8 activation. Science 1998; 281: 1680
press p53 function would make effective chemoprotec- 1683.
tive agents. Although inactivation of p53 in tumours 18. Siliciano JD, Canman CE, Taya Y, Sakaguchi K, Appella E, Kastan MB.
DNA damage induces phosphorylation of the amino terminus of p53.
may be undesirable, such a strategy would likely be Genes Dev 1997; 11: 34713481.
suitable for advanced cancers, most of which have 19. Woo RA, McLure KG, Lees-Miller SP, Rancourt DE, Lee PW. DNA-
dependent protein kinase acts upstream of p53 in response to DNA
already lost p53 function. damage. Nature 1998; 394: 700704.
20. Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional
activator of the human bax gene. Cell 1995; 80: 293299.
21. Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A model for
CONCLUSION p53-induced apoptosis. Nature 1997; 389: 300305.
22. Skulachev VP. Cytochrome c in the apoptotic and antioxidant cascades.
Although the precise contribution of impaired apop- FEBS Lett 1998; 423: 275280.
23. Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human protein
totic capability to drug resistance in tumours remains to homologous to C. elegans CED-4, participates in cytochrome c-dependent
be determined, emerging clinical evidence correlates activation of caspase-3. Cell 1997; 90: 405413.
mutations of apoptotic genes and treatment failure. 24. Li P, Nijhawan D, Budihardjo I, et al. Cytochrome c and dATP-dependent
formation of Apaf-1/caspase-9 complex initiates an apoptotic protease
Understanding of the apoptotic network may help to cascade. Cell 1997; 91: 479489.
improve current treatment strategies. Certainly there is 25. Hakem R, Hakem A, Duncan GS, et al. Differential requirement for
caspase 9 in apoptotic pathways in vivo. Cell 1998; 94: 339352.
still a long way to go, but the fact that apoptosis is 26. Kuida K, Haydar TF, Kuan CY, et al. Reduced apoptosis and cytochrome
controlled by genes raises the exciting prospect that the c-mediated caspase activation in mice lacking caspase 9. Cell 1998; 94:
problem of drug sensitivity and resistance will be 325337.
27. Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a Bcl2 interacting
amenable to interventions based on molecular biology. protein, mediates cytochrome c release from mitochondria in response to
activation of cell surface death receptors. Cell 1998; 94: 481490.
28. Hu Y, Benedict MA, Wu D, Inohara N, Nunez G. Bcl-XL interacts with
ACKNOWLEDGEMENTS Apaf-1 and inhibits Apaf-1-dependent caspase-9 activation. Proc Natl
Acad Sci USA 1998; 95: 43864391.
29. Pan G, ORourke K, Dixit VM. Caspase-9 Bcl-XL, and Apaf-1 form a
ternary complex. J Biol Chem 1998; 273: 58415845.
We apologize to those investigators whose work, due 30. Reed JC, Jurgensmeier JM, Matsuyama S. Bcl-2 family proteins and
to space constraints, was not cited. We also thank M. E. mitochondria. Biochim Biophys Acta 1998; 1366: 127137.
McCurrach for editorial assistance, and acknowledge 31. Strasser A, Huang DC, Vaux DL. The role of the bcl-2/ced-9 gene family
in cancer and general implications of defects in cell death control for
support from a Dr Mildred Scheel Cancer Foundation tumourigenesis and resistance to chemotherapy. Biochim Biophys Acta
Postdoctoral Fellowship (C.A.S.) and from a Sidney 1997; 1333: F151F178.
Kimmel Scholar Award and Grant CA13106 from the 32. Lotem J, Sachs L. Cytokine suppression of protease activation in wild-type
p53-dependent and p53-independent apoptosis. Proc Natl Acad Sci USA
National Cancer Institute (S.W.L.). 1997; 94: 93499353.
33. Symonds H, Krall L, Remington L, et al. p53-dependent apoptosis
suppresses tumor growth and progression in vivo. Cell 1994; 78: 703711.
34. Bardeesy N, Beckwith JB, Pelletier J. Clonal expansion and attenuated
REFERENCES apoptosis in Wilms tumors are associated with p53 gene mutations.
1. Kerr JF, Winterford CM, Harmon BV. Apoptosis. Its significance in Cancer Res 1995; 55: 215219.
cancer and cancer therapy. Cancer 1994; 73: 20132026. 35. Evan GI, Wylie AH, Gilbert CS, et al. Induction of apoptosis in fibroblasts
2. Evan G, Littlewood T. A matter of life and cell death. Science 1998; 281: by c-myc protein. Cell 1992; 69: 119128.
13171322. 36. Harrington EA, Bennett MR, Fanidi A, Evan GI. c-Myc-induced apop-
3. Thompson CB. Apoptosis in the pathogenesis and treatment of disease. tosis in fibroblasts is inhibited by specific cytokines. EMBO J 1994; 13:
Science 1995; 267: 14561462. 32863295.
4. Woo M, Hakem R, Soengas MS, et al. Essential contribution of caspase 37. Graeber TG, Osmanian C, Jacks T, et al. Hypoxia-mediated selection of
3/CPP32 to apoptosis and its associated nuclear changes. Genes Dev 1998; cells with diminished apoptotic potential in solid tumours. Nature 1996;
12: 806819. 379: 8891.
5. Chao DT, Korsmeyer SJ. BCL-2 family: regulators of cell death. Annu Rev 38. Hermeking H, Eick D. Mediation of c-Myc-induced apoptosis by p53.
Immunol 1998; 16: 395419. Science 1994; 265: 20912093.

Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
APOPTOSIS AND THERAPY 135

39. Lowe SW, Jacks T, Housman DE, Ruley HE. Abrogation of oncogene- 69. Fujiwara T, Grimm EA, Mukhopadhyay T, Zhang WW, Owen-Schaub
associated apoptosis allows transformation of p53-deficient cells. Proc LB, Roth JA. Induction of chemosensitivity in human lung cancer cells in
Natl Acad Sci USA 1994; 91: 20262030. vivo by adenovirus-mediated transfer of the wild-type p53 gene. Cancer Res
40. Morgenbesser SD, Williams BO, Jacks T, DePinho RA. p53-dependent 1994; 54: 22872291.
apoptosis produced by Rb-deficiency in the developing mouse lens. Nature 70. Blagosklonny MV, El-Deiry WS. Acute overexpression of wt p53 facili-
1994; 371: 7274. tates anticancer drug-induced death of cancer and normal cells. Int J
41. Wu X, Levine AJ. p53 and E2F-1 cooperate to mediate apoptosis. Proc Cancer 1998; 75: 933940.
Natl Acad Sci USA 1994; 91: 36023606. 71. Reed JC. Bcl-2 family proteins: regulators of apoptosis and chemoresist-
42. Serrano M, Lee H, Chin L, Cordon-Cardo C, Beach D, DePinho RA. ance in hematologic malignancies. Semin Hematol 1997; 34 (Suppl 5):
Role of the INK4a locus in tumor suppression and cell mortality. Cell 919.
1996; 85: 2737. 72. Fisher TC, Milner AE, Gregory CD, et al. bcl-2 modulation of apoptosis
43. de Stanchina E, McCurrach ME, Zindy F, et al. E1A signalling to p53 induced by anticancer drugs: resistance to thymidylate stress is indepen-
involves the p19(ARF) tumor suppressor. Genes Dev 1998; 12: 24342442. dent of classical resistance pathways. Cancer Res 1993; 53: 33213326.
44. Zindy F, Eischen CM, Randle DH, et al. Myc signalling via the ARF 73. Kamesaki S, Kamesaki H, Jorgensen TJ, Tanizawa A, Pommier Y,
tumor suppressor regulates p53-dependent apoptosis and immortalization. Cossman J. bcl-2 protein inhibits etoposide-induced apoptosis through its
Genes Dev 1998; 12: 24242433. effects on events subsequent to topoisomerase II-induced DNA strand
45. Stott FJ, Bates S, James MC, et al. The alternative product from the breaks and their repair. Cancer Res 1993; 53: 42514256.
human CDKN2A locus, p14(ARF), participates in a regulatory feedback 74. Cory S. Regulation of lymphocyte survival by the bcl-2 gene family. Annu
loop with p53 and MDM2. EMBO J 1998; 17: 50015014. Rev Immunol 1995; 13: 513543.
46. Bates S, Phillips AC, Clark PA, et al. p14ARF links the tumour suppressors 75. Datta R, Manome Y, Taneja N, et al. Overexpression of Bcl-XL by
RB and p53. Nature 1998; 395: 124125. cytotoxic drug exposure confers resistance to ionizing radiation-induced
47. Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell internucleosomal DNA fragmentation. Cell Growth Differ 1995; 6: 363
survival and cooperates with c-myc to immortalize pre-B cells. Nature 370.
1988; 335: 440442. 76. Kojima H, Endo K, Moriyama H, et al. Abrogation of mitochondrial
48. Christofori G, Naik P, Hanahan D. A second signal supplied by insulin- cytochrome c release and caspase-3 activation in acquired multidrug
like growth factor II in oncogene-induced tumorigenesis. Nature 1994; 369: resistance. J Biol Chem 1998; 273: 16 64716 650.
414418. 77. Reed JC. Mechanisms of Bcl-2 family protein function and dysfunction in
49. Elson A, Deng C, Campos-Torres J, Donehower LA, Leder P. The health and disease. Behring Inst Mitt 1996; 97: 72100.
MMTV/c-myc transgene and p53 null alleles collaborate to induce T-cell 78. McCurrach ME, Connor TM, Knudson CM, Korsmeyer SJ, Lowe SW.
lymphomas, but not mammary carcinomas in transgenic mice. Oncogene bax-deficiency promotes drug resistance and oncogenic transformation by
1995; 11: 181190. attenuating p53-dependent apoptosis. Proc Natl Acad Sci USA 1997; 94:
50. Dive C, Hickman JA. Drugtarget interactions: only the first step in the 23452349.
commitment to a programmed cell death? Br J Cancer 1991; 64: 192196. 79. Sakakura C, Sweeney EA, Shirahama T, et al. Overexpression of bax
51. Lowe SW, Ruley HE, Jacks T, Housman DE. p53-dependent sensitizes human breast cancer MCF-7 cells to radiation-induced apopto-
apoptosis modulates the cytotoxicity of anticancer agents. Cell 1993; 74: sis. Int J Cancer 1996; 67: 101105.
957967. 80. Kobayashi T, Ruan S, Clodi K, et al. Overexpression of Bax gene
52. Lotem J, Sachs L. Regulation by bcl-2, c-myc, and p53 of susceptibility to sensitizes K562 erythroleukemia cells to apoptosis induced by selective
induction of apoptosis by heat shock and cancer chemotherapy com- chemotherapeutic agents. Oncogene 1998; 16: 15871591.
pounds in differentiation-competent and -defective myeloid leukemic cells. 81. Oltvai ZN, Korsmeyer SJ. Checkpoints of dueling dimers foil death
Cell Growth Differ 1993; 4: 4147. wishes. Cell 1994; 79: 189192.
53. Rupnow BA, Murtha AD, Alarcon RM, Giaccia AJ, Knox SJ. Direct 82. Srivastava RK, Srivastava AR, Korsmeyer SJ, Nesterova M, Cho-Chung
evidence that apoptosis enhances tumor responses to fractionated radio- YS, Longo DL. Involvement of microtubules in the regulation of Bcl2
therapy. Cancer Res 1998; 58: 17791784. phosphorylation and apoptosis through cyclic AMP-dependent protein
54. Fisher DE. Apoptosis in cancer therapy: crossing the threshold. Cell 1994; kinase. Mol Cell Biol 1998; 18: 35093517.
78: 539542. 83. Friesen C, Herr I, Krammer PH, Debatin KM. Involvement of the CD95
55. Del Bino G, Lassota P, Darzynkiewicz Z. The S-phase cytotoxicity of (APO-1/FAS) receptor/ligand system in drug-induced apoptosis in leuke-
camptothecin. Exp Cell Res 1991; 193: 2735. mia cells. Nature Med 1996; 2: 574577.
56. Erba E, Sen S, Lorico A, DIncalci M. Potentiation of etoposide cyto-
84. Gamen S, Anel A, Lasierra P, et al. Doxorubicin-induced apoptosis in
toxicity against a human ovarian cancer cell line by pretreatment with
human T-cell leukemia is mediated by caspase-3 activation in a Fas-
non-toxic concentrations of methotrexate or aphidicolin. Eur J Cancer
independent way. FEBS Lett 1997; 417: 360364.
1992; 28: 6671.
85. Eischen CM, Kottke TJ, Martins LM, et al. Comparison of apoptosis in
57. Darzynkiewicz Z. Apoptosis in antitumor strategies: modulation of cell
wild-type and Fas-resistant cells: chemotherapy-induced apoptosis is not
cycle or differentiation. J Cell Biochem 1995; 58: 151159.
dependent on Fas/Fas ligand interactions. Blood 1997; 90: 935943.
58. Tsuboi K, Tsuchida Y, Nose T, Ando K. Cytotoxic effect of accelerated
carbon beams on glioblastoma cell lines with p53 mutation: clonogenic 86. Landowski TH, Gleason-Guzman MC, Dalton WS. Selection for drug
survival and cell-cycle analysis. Int J Radiat Biol 1998; 74: 7179. resistance results in resistance to Fas-mediated apoptosis. Blood 1997; 89:
59. Searle J, Lawson TA, Abbott PJ, Harmon B, Kerr JF. An electron- 18541861.
microscope study of the mode of cell death induced by cancer- 87. Los M, Herr I, Friesen C, Fulda S, Schulze-Osthoff K, Debatin KM.
chemotherapeutic agents in populations of proliferating normal and Cross-resistance of CD95- and drug-induced apoptosis as a consequence of
neoplastic cells. J Pathol 1975; 116: 129138. deficient activation of caspases (ICE/Ced-3 proteases). Blood 1997; 90:
60. Lowe SW, Schmitt EM, Smith SW, Osborne BA, Jacks T. p53 is required 31183129.
for radiation-induced apoptosis in mouse thymocytes. Nature 1993; 362: 88. Wu GS, Burns TF, McDonald ER, et al. KILLER/DR5 is a DNA
847849. damage-inducible p53-regulated death receptor gene. Nature Genet 1997;
61. Clarke AR, Gledhill S, Hooper ML, Bird CC, Wyllie AH. p53 dependence 17: 141143.
of early apoptotic and proliferative responses within the mouse intestinal 89. Sheikh MS, Burns TF, Huang Y, et al. p53-dependent and -independent
epithelium following gamma-irradiation. Oncogene 1994; 9: 17671773. regulation of the death receptor KILLER/DR5 gene expression in
62. Griffiths SD, Clarke AR, Healy LE, et al. Absence of p53 permits response to genotoxic stress and tumor necrosis factor alpha. Cancer Res
propagation of mutant cells following genotoxic damage. Oncogene 1997; 1998; 58: 15931598.
14: 523531. 90. Green DR. Apoptotic pathways: the roads to ruin. Cell 1998; 94: 695698.
63. Lotem J, Sachs L. Hematopoietic cells from mice deficient in wild-type p53 91. Antoku K, Liu Z, Johnson DE. Inhibition of caspase proteases by CrmA
are more resistant to induction of apoptosis by some agents. Blood 1993; enhances the resistance of human leukemic cells to multiple chemothera-
82: 10921096. peutic agents. Leukemia 1997; 11: 16651672.
64. Merritt AJ, Potten CS, Kemp CJ, et al. The role of p53 in spontaneous and 92. Yoshida H, Kong Y-Y, Yoshida R, et al. Apaf1 is required for mitochon-
radiation-induced apoptosis in the gastrointestinal tract of normal and drial pathways of apoptosis and brain development. Cell 1998; 94:
p53-deficient mice. Cancer Res 1994; 54: 614617. 739750.
65. Pritchard DM, Potten CS, Hickman JA. The relationships between 93. Eastman A. Survival factors, intracellular signal transduction, and the
p53-dependent apoptosis, inhibition of proliferation and 5-fluorouracil- activation of endonucleases in apoptosis. Semin Cancer Biol 1995; 6:
induced histopathology in murine intestinal epithelia. Cancer Res (in 4552.
press). 94. Walker A, Taylor ST, Hickman JA, Dive C. Germinal center-derived
66. Lowe SW, Bodis S, McClatchey A, et al. p53 status and the efficacy of signals act with Bcl-2 to decrease apoptosis and increase clonogenicity of
cancer therapy in vivo. Science 1994; 266: 807810. drug-treated human B lymphoma cells. Cancer Res 1997; 57: 19391945.
67. Lowe SW. Cancer therapy and p53. Curr Opin Oncol 1995; 7: 547553. 95. Schmitz ML, Indorf A, Limbourg FP, Stadtler H, Traenckner EB,
68. Weinstein JN, Myers TG, OConnor PM, et al. An information-intensive Baeuerle PA. The dual effect of adenovirus type 5 E1A 13S protein on
approach to the molecular pharmacology of cancer. Science 1997; 275: NF-kappaB activation is antagonized by E1B 19K. Mol Cell Biol 1996; 16:
343349. 40524063.

Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
136 C. A. SCHMITT AND S. W. LOWE

96. Ware CF, VanArsdale S, VanArsdale TL. Apoptosis mediated by the 124. Han JW, Dionne CA, Kedersha NL, Goldmacher VS. p53 status affects
TNF-related cytokine and receptor families. J Cell Biochem 1996; 60: the rate of the onset but not the overall extent of doxorubicin-induced cell
4755. death in rat-1 fibroblasts constitutively expressing c-Myc. Cancer Res 1997;
97. Mayo MW, Wang CY, Cogswell PC, et al. Requirement of NF-kappaB 57: 176182.
activation to suppress p53-independent apoptosis induced by oncogenic 125. Waldman T, Zhang Y, Dillehay L, et al. Cellcycle arrest versus cell death
Ras. Science 1997; 278: 18121815. in cancer therapy. Nature Med 1997; 3: 10341036.
98. Downward J. Ras signalling and apoptosis. Curr Opin Genet Dev 1998; 8: 126. Di Leonardo A, Linke SP, Clarkin K, Wahl GM. DNA damage triggers a
4954. prolonged p53-dependent G1 arrest and long-term induction of Cip1 in
99. Jeremias I, Kupatt C, Baumann B, Herr I, Wirth T, Debatin KM. normal human fibroblasts. Genes Dev 1994; 8: 25402551.
Inhibition of nuclear factor kappaB activation attenuates apoptosis resist- 127. Sjogren S, Inganas M, Norberg T, et al. The p53 gene in breast cancer:
ance in lymphoid cells. Blood 1998; 91: 46244631. prognostic value of complementary DNA sequencing versus immunohis-
100. Wang CY, Mayo MW, Baldwin AS Jr. TNF- and cancer therapy-induced tochemistry. J Natl Cancer Inst 1996; 88: 173182.
apoptosis: potentiation by inhibition of NF-kappaB. Science 1996; 274: 128. Ko LJ, Prives C. p53: puzzle and paradigm. Genes Dev 1996; 10:
784787. 10541072.
101. Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephos- 129. Paquette RL, Lee YY, Wilczynski SP, et al. Mutations of p53 and human
phorylates the lipid second messenger, phosphatidylinositol 3,4,5- papillomavirus infection in cervical carcinoma. Cancer 1993; 72: 1272
trisphosphate. J Biol Chem 1998; 273: 13 37513 378. 1280.
102. Myers MP, Pass I, Batty IH, et al. The lipid phosphatase activity of PTEN 130. Rampino N, Yamamoto H, Ionov Y, et al. Somatic frameshift mutations
is critical for its tumor suppressor function. Proc Natl Acad Sci USA (in in the BAX gene in colon cancers of the microsatellite mutator phenotype.
press). Science 1997; 275: 967969.
103. Aas T, Borresen AL, Geisler S, et al. Specific P53 mutations are associated 131. Adams JM, Harris AW, Pinkert CA, et al. The c-myc oncogene driven by
with de novo resistance to doxorubicin in breast cancer patients. Nature immunoglobulin enhancers induces lymphoid malignancy in transgenic
Med 1996; 2: 811814. mice. Nature 1985; 318: 533538.
104. Benhattar J, Cerottini JP, Saraga E, Metthez G, Givel JC. p53 mutations 132. Sandig V, Brand K, Herwig S, Lukas J, Bartek J, Strauss M. Adenovirally
as a possible predictor of response to chemotherapy in metastatic colorec- transferred p16INK4/CDKN2 and p53 genes cooperate to induce apop-
tal carcinomas. Int J Cancer 1996; 69: 190192. totic tumor cell death. Nature Med 1997; 3: 313319.
105. Iwadate Y, Fujimoto S, Tagawa M, et al. Association of p53 gene 133. Fueyo J, Gomez-Manzano C, Yung WK, et al. Overexpression of E2F-1 in
mutation with decreased chemosensitivity in human malignant gliomas. Int glioma triggers apoptosis and suppresses tumor growth in vitro and in vivo.
J Cancer 1996; 69: 236240. Nature Med 1998; 4: 685690.
106. Righetti SC, Della Torre G, Pilotti S, et al. A comparative study of p53 134. Nielsen LL, Maneval DC. P53 tumor suppressor gene therapy for cancer.
gene mutations, protein accumulation, and response to cisplatin-based Cancer Gene Ther 1998; 5: 5263.
chemotherapy in advanced ovarian carcinoma. Cancer Res 1996; 56: 135. Spitz FR, Nguyen D, Skibber JM, Cusack J, Roth JA, Cristiano RJ. In
689693. vivo adenovirus-mediated p53 tumor suppressor gene therapy for colorec-
107. Taubert H, Meye A, Wurl P. Prognosis is correlated with p53 mutation tal cancer. Anticancer Res 1996; 16: 34153422.
type for soft tissue sarcoma patients. Cancer Res 1996; 56: 41344136. 136. Badie B, Kramar MH, Lau R, Boothman DA, Economou JS, Black KL.
108. Ichikawa A, Kinoshita T, Watanabe T, et al. Mutations of the p53 gene as Adenovirus-mediated p53 gene delivery potentiates the radiation-induced
a prognostic factor in aggressive B-cell lymphoma. N Engl J Med 1997; growth inhibition of experimental brain tumors. J Neurooncol 1998; 37:
337: 529534. 217222.
109. Cordone I, Masi S, Mauro FR, et al. p53 expression in B-cell chronic 137. Roth JA, Swisher SG, Merritt JA, et al. Gene therapy for non-small cell
lymphocytic leukemia: a marker of disease progression and poor progno- lung cancer: a preliminary report of a phase I trial of adenoviral p53 gene
sis. Blood 1998; 91: 43424349. replacement. Semin Oncol 1998; 25 (Suppl 8): 3337.
110. Drach J, Ackermann J, Fritz E, et al. Presence of a p53 gene deletion in 138. Parr MJ, Manome Y, Tanaka T, et al. Tumor-selective transgene expres-
patients with multiple myeloma predicts for short survival after sion in vivo mediated by an E2F-responsive adenoviral vector. Nature Med
conventional-dose chemotherapy. Blood 1998; 92: 802809. 1997; 3: 11451149.
111. Marks DI, Kurz BW, Link MP, et al. Altered expression of p53 and 139. Seth P, Brinkmann U, Schwartz GN, et al. Adenovirus-mediated gene
mdm-2 proteins at diagnosis is associated with early treatment failure in transfer to human breast tumor cells: an approach for cancer gene therapy
childhood acute lymphoblastic leukemia. J Clin Oncol 1997; 15: 1158 and bone marrow purging. Cancer Res 1996; 56: 13461351.
1162. 140. Wroblewski JM, Lay LT, Meeker TC. Adenoviral vectors in hematology:
112. Meijerink JP, Mensink EJ, Wang K, et al. Hematopoietic malignancies purging, stem cell gene transfer, or both. Blood 1997; 89: 46644665.
demonstrate loss-of-function mutations of BAX. Blood 1998; 91: 2991 141. Bedi A, Zehnbauer BA, Barber JP, Sharkis SJ, Jones RJ. Inhibition of
2997. apoptosis by BCR-ABL in chronic myeloid leukemia. Blood 1994; 83:
113. Krajewski S, Blomqvist C, Franssila K, et al. Reduced expression of 20382044.
proapoptotic gene BAX is associated with poor response rates to combi- 142. Wright LA, Milliken S, Biggs JC, Kearney P. Ex vivo effects associated
nation chemotherapy and shorter survival in women with metastatic breast with the expression of a bcr-abl-specific ribozyme in a CML cell line.
adenocarcinoma. Cancer Res 1995; 55: 44714478. Antisense Nucleic Acid Drug Dev 1998; 8: 1523.
114. Sjostrom J, Krajewski S, Franssila K, et al. A multivariate analysis of 143. Reed JC. Promise and problems of Bcl-2 antisense therapy. J Natl Cancer
tumour biological factors predicting response to cytotoxic treatment in Inst 1997; 89: 988990.
advanced breast cancer. Br J Cancer 1998; 78: 812815. 144. Jansen B, Schlagbauer-Wadl H, Brown BD, et al. bcl-2 antisense therapy
115. Pepper C, Hoy T, Bentley DP. Bcl-2/Bax ratios in chronic lymphocytic chemosensitizes human melanoma in SCID mice. Nature Med 1998; 4:
leukaemia and their correlation with in vitro apoptosis and clinical 232234.
resistance. Br J Cancer 1997; 76: 935938. 145. Han JS, Nunez G, Wicha MS, Clarke MF. Targeting cancer cell death
116. Tu Y, Renner S, Xu F, et al. BCL-X expression in multiple myeloma: with a bcl-XS adenovirus. Springer Semin Immunopathol 1998; 19: 279
possible indicator of chemoresistance. Cancer Res 1998; 58: 256262. 288.
117. Beyermann B, Adams HP, Henze G. Philadelphia chromosome in relapsed 146. Ealovega MW, McGinnis PK, Sumantran VN, Clarke MF, Wicha MS.
childhood acute lymphoblastic leukemia: a matched-pair analysis. Berlin bcl-xs gene therapy induces apoptosis of human mammary tumors in nude
FrankfurtMunster Study Group. J Clin Oncol 1997; 15: 22312237. mice. Cancer Res 1996; 56: 19651969.
118. Hawkins DS, Demers GW, Galloway DA. Inactivation of p53 enhances 147. Clarke MF, Apel IJ, Benedict MA, et al. A recombinant bcl-x s adenovirus
sensitivity to multiple chemotherapeutic agents. Cancer Res 1996; 56: selectively induces apoptosis in cancer cells but not in normal bone
892898. marrow cells. Proc Natl Acad Sci USA 1995; 92: 11 02411 028.
119. Fan S, Smith ML, Rivet DJN, et al. Disruption of p53 function sensitizes 148. Samuelson AV, Lowe SW. Selective induction of p53 and chemosensitivity
breast cancer MCF-7 cells to cisplatin and pentoxifylline. Cancer Res 1995; in RB-deficient cells by E1A mutants unable to bind the RB-related
55: 16491654. proteins. Proc Natl Acad Sci USA 1997; 94: 12 09412 099.
120. Lipponen P, Pietilainen T, Kosma VM, Aaltomaa S, Eskelinen M, 149. Beltinger C, Kurz E, Bohler T, Schrappe M, Ludwig WD, Debatin KM.
Syrjanen K. Apoptosis suppressing protein bcl-2 is expressed in well- CD95 (APO-1/Fas) mutations in childhood T-lineage acute lymphoblastic
differentiated breast carcinomas with favourable prognosis. J Pathol 1995; leukemia. Blood 1998; 91: 39433951.
177: 4955. 150. Pai SI, Wu GS, Ozoren N, et al. Rare loss-of-function mutation of a death
121. Berardo MD, Elledge RM, de Moor C, Clark GM, Osborne CK, Allred receptor gene in head and neck cancer. Cancer Res 1998; 58: 35133518.
DC. bcl-2 and apoptosis in lymph node positive breast carcinoma. Cancer 151. Tamiya S, Etoh K, Suzushima H, Takatsuki K, Matsuoka M. Mutation of
1998; 82: 12961302. CD95 (Fas/Apo-1) gene in adult T-cell leukemia cells. Blood 1998; 91:
122. Yin DX, Schimke RT. BCL-2 expression delays drug-induced apoptosis 39353942.
but does not increase clonogenic survival after drug treatment in HeLa 152. Wold WS. Adenovirus genes that modulate the sensitivity of virus-infected
cells. Cancer Res 1995; 55: 49224928. cells to lysis by TNF. J Cell Biochem 1993; 53: 329335.
123. Lock RB, Stribinskiene L. Dual modes of death induced by etoposide in 153. Dong J, Naito M, Tsuruo T. c-Myc plays a role in cellular susceptibility to
human epithelial tumor cells allow Bcl-2 to inhibit apoptosis without death receptor-mediated and chemotherapy-induced apoptosis in human
affecting clonogenic survival. Cancer Res 1996; 56: 40064012. monocytic leukemia U937 cells. Oncogene 1997; 15: 639647.

Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)
APOPTOSIS AND THERAPY 137

154. Pan G, Ni J, Wei YF, Yu G, Gentz R, Dixit VM. An antagonist decoy 159. Koc ON, Allay JA, Lee K, Davis BM, Reese JS, Gerson SL. Transfers of
receptor and a death domain-containing receptor for TRAIL. Science drug resistance genes into hematopoietic progenitors to improve chemo-
1997; 277: 815818. therapy tolerance. Semin Oncol 1996; 23: 4665.
155. Marcellus RC, Lavoie JN, Boivin D, Shore GC, Ketner G, Branton PE. 160. Magni M, Shammah S, Schiro R, Mellado W, Dalla-Favera R, Gianni
The early region 4 orf4 protein of human adenovirus type 5 induces AM. Induction of cyclophosphamide-resistance by aldehyde-
p53-independent cell death by apoptosis. J Virol 1998; 72: 71447153. dehydrogenase gene transfer. Blood 1996; 87: 10971103.
156. Noteborn MH, Todd D, Verschueren CA, et al. A single chicken anemia 161. Wilson JW, Pritchard DM, Hickman JA, Potten CS. Radiation-induced
virus protein induces apoptosis. J Virol 1994; 68: 346351. p53 and p21WAF-1/CIP1 expression in the murine intestinal epithelium:
157. Danen-Van Oorschot AA, Fischer DF, Grimbergen JM, et al. Apoptin apoptosis and cell cycle arrest. Am J Pathol 1998; 153: 899909.
induces apoptosis in human transformed and malignant cells but not in 162. Wlodarski P, Wasik M, Ratajczak MZ, et al. Role of p53 in hematopoietic
normal cells. Proc Natl Acad Sci USA 1997; 94: 58435847. recovery after cytotoxic treatment. Blood 1998; 91: 29983006.
158. Noteborn MHM, Zhang Y, van der Eb AJ. Apoptin(R) specifically causes 163. Pritchard DM, Watson AJ, Potten CS, Jackman AL, Hickman JA.
apoptosis in tumor cells and after UV-treatment in untransformed Inhibition by uridine but not thymidine of p53-dependent intestinal
cells from cancer-prone individuals: a review. Mutat Res 1998; 400: apoptosis initiated by 5-fluorouracil: evidence for the involvement of RNA
447455. perturbation. Proc Natl Acad Sci USA 1997; 94: 17951799.

Copyright  1999 John Wiley & Sons, Ltd. J. Pathol. 187: 127137 (1999)

You might also like