Abstract
Apoptosis, or programmed cell death, is central to the development and homeostasis of the hematopoietic system. Dysregulation of apoptosis plays an important role in the development of a variety of human pathologies, including cancer, autoimmune diseases and neurodegenerative disorders. Studies carried out in the last years have shown that leukemia cells invariably have abnormalities in one or more apoptotic pathways, determining a survival advantage of these cells over their normal counterpart. Furthermore, abnormalities in the apoptotic response also play a role in the development of drug resistance by leukemic cells. The identification of the different components of the apoptotic pathways has enabled the detection of various biochemical defects present in leukemic cells compared to their normal counterparts. These defects contribute to the survival advantage of the leukemic clone over the normal hematopoietic cells and are also frequently associated with a low rate of response to standard chemotherapy treatment and with poor survival. Furthermore, these findings have lead to the identification of many potential apoptotic targets for the development of new drugs targeting anti-apoptotic molecules abnormally expressed or regulated in leukemic cells. Many of these drugs restore the sensitivity of leukemic cells to apoptotic stimuli and some of them are under investigation at a clinical level.A cute myelogenous leukemia (AML) is a heterogeneous group of malignant disorders characterized by uncontrolled proliferation of clonal leukemic hematopoietic precursor cells blocked at various stages of normal myeloid development, associated with severely impaired normal hematopoiesis, thus leading to neutropenia, anemia and thrombocytopenia. The development of AML is associated with accumulation of acquired genetic alterations and epigenetic changes in hematopoietic progenitor cells which alter normal mechanisms of cell growth, proliferation, death and differentiation. Approximately 50–75% of adult patients with AML achieve complete remission when treated with cytabarine and daunorubicin or idarubicin, or with the topoisomerase II inhibitor, mitoxantrone.1,2 Apoptotic processes play a key role in the control of tissue homeostasis, particularly of rapidly renewing tissues, such as hematopoietic tissue. Studies carried out during the last twenty years have dramatically improved our understanding of the role of apoptotic mechanisms in the control of differentiation, proliferation and survival of normal and leukemic cells.
Cell death pathways
The most common and well defined form of programmed cell death is apoptosis, a physiological process of cellular suicide required for the maintenance of cell homeostasis, embryonic development and for the differentiation and function of hematopoietc and lymphoid cells. A common feature of the apoptotic process is the constant involvement of caspases, a family of intracellular cysteine proteases (cysteine aspartyl-specific proteases) (Figure 1). These enzymes are present as inactive zymogens in all animal cells, but can be triggered to assume an active state, usually through proteolytic processing at conserved aspartic acid residues.
Based on their level of action caspases are distinguished in initiatior caspases (which include caspase-8, -2, -9 and -10) and effector caspases (caspase-3, -6 and -7): the former act at the origin of the apoptotic process, while the latter act during the late steps of the apoptotic process (Figure 1). Once activated, the effector caspases are responsible for the proteolytic cleavage of a broad spectrum of cellular proteins, which ultimately culminates with cell death. The initiator caspases are structurally characterized by an extended N-terminal region, comprising one or two adaptor domains that are essential for their function; in contrast, the effector caspases contain 20–30 amino acid residues in their prodomain region. There are two pathways by which caspase activation is induced, the extrinsic and the intrinsic apoptotic pathways (Figures 2 and 3). Both the pathways converge on activation of effector caspases but require different initiation caspases to start the process. The extrinsic pathway is activated by engagement of death receptors on the cell membrane. Binding of ligands, such as FasL, tumor necrosis factor (TNF) and TNF-related apoptosis-inducing ligand (TRAIL) to their respective membrane receptors Fas, TNF-R and TRAIL-R induces the formation of the oligomeric death-induced signaling complex (DISC) (Figure 2). DISC in turn promotes caspase-8 recruitment and a cascade of activation of caspase enzymes that culminates with cell death. The activation of caspase-8 is antagonized by cellular FLICE inhibitory protein (c-FLIP), an enzymatically inactive relative of caspase-8 and -10 that binds to DISC. The knockdown of c-FLIP augments DISC recruitment, activation and processing of caspase-8, thereby enhancing effector-caspase stimulation and apoptosis. The intrinsic pathway is triggered by various extracellular and intracellular stresses, including growth factor deprivation, DNA damage, oncogene induction, hypoxia and cytotoxic drugs (Figure 3). Cellular signals originated by various mechanisms by these different stresses converge on a cellular target represented by mitochondria. Mitochondrial membrane permeability is controlled by pro-apoptotic (Bax, Bak, Bad, Bid, Bim, Bmf, NOXA, PUMA) and anti-apoptotic (Bcl-2, Bcl-XL, Mcl-1) members of the Bcl-2 family, inducing or preventing heterodimerization of pro-apoptotic members. A series of biochemical events is induced that lead to damage of the outer mitochondrial membrane, with the consequent release of cytochrome c and other pro-apoptotic molecules, such as Smac/DIABLO, from the inner membrane into the cytosol enabling the formation of the apoptosome, a large molecular complex formed by cytochrome c, apoptotic protease activating factor 1 (APAF-1) and caspase-9, and massive activation of caspases (Figure 3).
Members of the Bcl-2 family either promote or prevent apoptosis. The pro-survival Bcl-2 family members include Bcl-2, Bcl-XL, Bcl-w, A1 and Mcl-1, all characterized by the presence of four Bcl-2 homology domains (BH1-BH4), with the exception of Mcl-1 that contains only three BH domains. These proteins all play crucial roles for cell survival and the loss of any of these proteins causes major deregulation of survival of some cell types. The pro-apoptotic members of the Bcl-2 family can be subdivided into two groups: (i) Bax-like proteins (Bax, Bak, Bok, Bcl-G, and Bfk) contain three BH regions; (ii) BH3-only proteins (Bad, Bik, Bid, Bim, Noxa, PUMA, Bmf and Hrk) possess only the BH3 domain. BH3-only proteins are essential for cell death initiation through the intrinsic pathway, whereas the Bax/Bak-like proteins play a key role in the progression of the apoptotic signaling stimuli at the level of permeabilization of the outer mitochondrial membrane. The molecular mode of action of BH3-only protein consists in binding to a surface groove, formed by the BH1, BH2 and BH3 domains, on the surface of Bcl-2-like molecules and, through this mechanism, antagonizing their pro-survival function.
Deregulation of apoptosis in acute myeloid leukemia: general mechanisms
Deregulation of apoptosis disrupts the complex and delicate balance between cell proliferation, cell survival and cell death and plays a major role in the development of diseases such as cancer, and particularly leukemia. The evasion of programmed cell death has been regarded as one of the six essential alterations in cellular physiology that dictate the growth of cancer cells and is a hallmark of virtually all cancers.3 Moreover, tumors that have alterations in proteins involved in cell death signaling are very frequently resistant to chemotherapy and are difficult to treat with chemotherapeutic agents that primarily act by inducing apoptosis.4
Leukemia development is regarded as a multistep process characterized by progressive genetic alterations that drive the transformation of normal human hematopoietic stem/progenitor cells into leukemic derivatives. Leukemia originates from a single cell that has undergone malignant transformation driven by frequent genetic mutations. These events are followed by a clonal selection of mutated cells that display increasingly aggressive behavior. There is clear evidence indicating that the cellular and molecular events leading to leukemia are orchestrated by leukemic stem cells.5 Using in vivo models in which human AML cells were transplanted into immunodeficient mice, a leukemia initiating cell was identified that showed properties of an immature progenitor cell, expressing a CD34/CD38-phenotype.6,7 By contrast, despite the fact that they showed a leukemic blast phenotype, CD34/CD38 leukemic cells were unable to initiate leukemia in immunodeficient mice.6,7 Although the origin of acute leukemias from leukemic stem cells was clearly established, it should be noted that only about 50% of AML are able to initiate leukemia in NOD/SCID mice.8 The analysis of the membrane phenotype of leukemic stem cells provided evidence of a great similarity with normal hematopoietic stem cells, consistent with the hypothesis that AML arises from a hematopoietic stem cell.9
About 50–55% of AML are characterized by the presence of specific balanced chromosome abnormalities involved in the formation of specific fusion genes that encode for fusion proteins, playing a key role in the development of leukemia through their effect on cell proliferation, survival and apoptosis.2 Simultaneous alterations in the normal regulation of cell proliferation and apoptosis induce leukemic cells to accumulate, thus promoting further genetic alterations and leukemogenesis. The key role of apoptosis in the pathogenesis of AML has been elucidated over the last years. Some fusion proteins interact with mediators of apoptosis, sending anti-apoptotic signals that favor the preferential survival of leukemic cells: PML/RAR-α or CBF/SMMHC through the p53 pathway or AML1/ETO through the Bcl2-related pathway.10–12 Furthermore, it has been suggested that MLL fusion proteins modify PPP1R15A function and inhibit apoptosis.13 Finally, the PML/RARα fusion protein was shown to exert an anti-apoptotic activity by downmodulating the expression of some death-inducing genes, such as TNF-R114 and TRAIL-R1/-R2.15 Interestingly, a high frequency of mutations of nucleophosmin was observed in AML that do not display chromosomal translocations.16 In fact, mutations affecting the carboxyterminal domain of nucleophosmin occur in a high percentage of adult patients with AML not associated with chromosomal translocations and these alterations create an additional nuclear export signal that relocalizes much of the protein from its normal nucleolar stores to the cytoplasm.16 Furthermore, nucleophosmin is also frequently found in chromosomal translocations associated with myelodysplasia/AML.17 Nucleophosmin acts as a cellular p53 negative regulator to protect hematopoietic cells from stress-induced apoptosis.18 Furthermore, nucleophosmin has been implicated in the control of chromosomal ploidy and DNA repair.19 The mutated nucleophosmin could perturb p53-dependent and independent activities of the ARF tumor suppressor protein, and through this mechanism could protect AML cells from apoptosis.20
These mutations are not sufficient to cause the development of AML and additional mutations, occurring at the level of signal transduction molecules (the receptor tyrosine kinases Flt3 or c-kit, NRAS and KRAS), are required for the generation of disease. Flt3 is overexpressed in the majority of AML and, importantly, is mutated in about 35% of AML [in about 25% of patients an internal tandem duplication (ITD) within the juxtamembrane domain of Flt3 is observed, while in about 10% of patients mutations, small deletions or insertion within the activation loop of the second kinase domain of Flt3 are observed].21 These mutations determine a constitutive activation and signaling of Flt3, resulting in the triggering of anti-apoptotic pathways. Flt3 ITD mutants constitutively activate MAPK, AKT and Stat5, leading to Pim-1 activation and Bcl-XL hyperexpression.22,23
Another membrane receptor tyrosine kinase frequently mutated in AML is c-kit. In fact, c-kit mutations at the level of exon 8 (encoding for a part of the extracellular domain of the receptor) or mutations at the level of exon 17, codon 816 in the activation loop of the catalytic domain occur in about 20% and 30% of AML patients with t(8;21) or inv(16)t(16;26).24 More recently, a third type of c-kit mutation was described in pediatric AML patients and was found to be associated with constitutive receptor signaling.25 Extracellular c-kit mutations resulted in c-kit receptor hyperactivation in response to kit ligand, with subsequent strong activation of MAPK and PI3K, while codon 816 c-kit mutations induced constitutive Stat-3 activation and upregulation of Bcl-XL and c-myc. The KITD816 mutations occur in t(8;21)/AML-1/ETO-positive AML and represent a reliable molecular marker identifying patients with a poor prognosis in an otherwise prognostically favorable AML group.26 Similarly, both types of c-kit mutations have an adverse impact on relapse risk and overall survival in AML with inv(16).27
KRAS and NRAS make part of a family of guanine nucleotide-binding proteins, activated by a large spectrum of cytokine receptors. KRAS and NRAS mutations are present in approximately 15% and 25% of AML patients: these mutations are usually point mutations and determine constitutive GTPase activity.28 NRAS mutations lead to increased activity of the RAS pathway, resulting in increased proliferation and decreased apoptosis.29 Constitutive RAS activation in AML without RAS mutations is also frequently observed, probably due to deregulation of signaling proteins acting upstream of RAS. Recent studies have shown that oncogenic NRAS mutants rapidly induce AML-like disease in mice, thus indicating that mutated NRAS can function as an initiating oncogene in the induction of myeloid malignancies.30 Animal models indicate that abnormalities in the apoptosis pathway may lead to the development of AML. Thus, Fas-deficient mice constitutively expressing Bcl-2 in myeloid cells develop a disease closely resembling the M2 FAB subtype of AML.31 These observations indicate that molecules involved in the control of apoptosis may act as tumor suppressors to control leukemogenic transformation in myeloid progenitor cells.
In addition to traditional cytogenetic analysis or the detection of mutations in individual genes, global gene expression profiling through microarray technology has provided an additional tool to probe and to try to classify in a rational way the marked biological heterogeneity of AML.32–35 This approach has enabled the identification of several clusters of AML, exhibiting different molecular signatures, prognoses and responses to treatment. Cluster A is characterized by AML exhibiting a high frequency of NPM1 mutations, a so-called stem-cell molecular signature and overexpression of genes that promote apoptosis, such as LTBP1 and caspase-3. Cluster B, represented by a group of AML patients with a very poor response to induction therapy and a very bad prognosis, is characterized by overexpression of genes inducing chemoresistance, such as ABCG2, almost complete absence of NPM1 mutations and often a normal cytogenetic analysis. AML patients in clustter C exhibit a high proliferative activity gene signature, a high incidence of karyotypic abnormalities, a low prevalence of NPM1 mutations and overexpression of IRF4 and IL10R. A high rate of response to induction therapy, but also a high rate of relapse have been observed in this cluster of patients. Cluster F is characterized by AML with monocytic features, with the highest white cell counts and a poor response to therapy. Finally, acute prolymphocytic leukemia (APL) form a separate cluster with a specific molecular signature and with a favorable prognosis related to the identification of specific first line and second line therapies.
Some signaling pathways, including phosphoinositide 3-kinase (PI3K)/AKT, JAK/STAT, RAS/Raf, /MEK/ERK and protein kinase C α, are crucial to many aspects of cell growth, survival and apoptosis, and their constitutive activation has been implicated in both the pathogenesis and progression of AML. These pathways have been extensively studied in AML. AKT was found to be constitutively phosphorylated in the majority of AML patients with high blast cell counts and was an adverse prognostic factor. 36 Phosphorylated AKT, in turn, phosphorylates caspases, Bad, nTOR, Waf1, and NF-κB, resulting in protection from apoptosis and increased proliferation.37 Activated AKT may promote leukemogenesis and inhibition of AKT activity induces apoptosis of AML blasts.38 The RAS/Raf/MEK/ERK pathway is a key signaling transduction pathway that induces increased cell proliferation and survival: constitutive activation of this pathway is observed in the majority of AML patients39 and its blockade with specific pharmacologic agents induces blast cell death.40 Finally, recent studies41 have provided clear evidence that a significant proportion of AML exhibit the simultaneous activation of AKT, ERK and PKCα and exhibit a worse prognosis, associated with short survival.
Death ligands and death receptors
There is growing interest in the development of therapeutic strategies that kill leukemic cells via activation of the extrinsic apoptotic pathway. Some members of the TNF-family directly trigger apoptosis and their activation could be used to induce the killing of tumor cells. Three ligands (TNF-α, FasL and TRAIL) of the TNF-family and their respective four receptors (TNF-R1, Fas, TRAIL-R1 and TRAIL-R2) have been considered for their potential use as anti-cancer therapeutics. The first of these molecules to be studied for its potential use as an anti-cancer drug was TNF-α. The demonstration that TNF-α selectively kills tumor cells but not normal cells, raised hopes about its potential use for cancer treatment.42 Unfortunately, these hopes were not fulfilled since the marked pro-inflammatory effects of TNF-α precluded its systemic administration. Subsequent studies were focused on the two other molecules, FasL and TRAIL. Unlike TNF-α, these two cytokines do not have proinflammatory effects and are suitable as potential anticancer drugs. Unfortunately, agonistic antibodies triggering Fas activation are highly hepatotoxic causing death in mouse models.43 In contrast, TRAIL and agonistic anti-TRAIL-R1/TRAIL-R2 antibodies appear to be well tolerated in vivo. In this context, initial studies have shown that TRAIL/Apo-2L exhibits potent anti-tumor activity and induces little cytotoxic effects in immunodeficient mice xenograft models implanted with several human tumor cell lines.44 However, the in vivo half-life of the TRAIL-ligand is very short (<4 minutes), suggesting that agonistic anti-TRAIL-R1 or -TRAIL-R2 could have a better pharmacologic impact.45–46 Another advantage of agonistic TRAILR1 and TRAIL-R2 antibodies is that they, at variance with the TRAIL ligand, do not bind to TRAIL decoy receptors, TRAIL-R3 and TRAIL-R4 are often present on the membrane of tumor cells.
Two anti-TRAIL-R have been developed for clinical use. One of these two antibodies is called HGS-ETR1 and is a fully human agonistic antibody with high affinity and specificity for TRAIL-R1.47 This antibody induces cell killing of tumor cell lines through activation of both extrinsic and intrinsic apoptotic pathways. Importantly, HGS-ETR1 was shown to have a long half-life in vivo (7–9 days in mouse) and suppressed the growth of several tumors in xenograft models in athymic mice.45 Finally, EGS-ETR1 potentiated the anti-tumor efficacy of several chemotherapeutic drugs.47 These observations clearly indicated that HGS-ETR1 has significant potential as a cancer therapeutic agent. The HGS-ETR1 antibody (Human Genome Sciences) was evaluated in phase I/II clinical trial in patients with advanced solid or hematologic tumors, revealing little toxicity.48
Expression of death receptors and sensitivity to death ligands in AML
Several studies have explored the sensitivity of AML to the pro-apoptotic effects of TRAIL showing that virtually all cases are resistant to this death ligand.49,50 The analysis of the pattern of TRAIL-R expression showed that TRAILR3 and TRAIL-R4 are frequently expressed in AML,50,51 while TRAIL-R1 expression is usually associated with AML exhibiting monocytic features.50 The presence of either TRAIL-R1/TRAIL-R2 expression is associated with TRAIL decoy receptor expression and does not confer sensitivity to TRAIL.51 The analysis of HL60 TRAIL-resistant subclones provided evidence that TRAIL resistance is related either to TRAIL-R1/TRAIL-R2 down-modulation or to caspase-8 mutations52 Anti-leukemic drugs49 or synthetic triterpenoids53 are able to render AML blasts sensitive to TRAIL-mediated apoptosis. Finally, other studies have shown that histone deacetylase inhibitors induce TRAIL-mediated apoptosis of AML cells.53
Recently, natural dihydroflavonol compounds purified from plants were shown to increase TRAIL sensitivity of leukemic cells by enhancing TRAIL-R2 expression.54 Similarly, arsenic trioxide (ATO), a compound used in the treatment of APL, sensitized AML cells to TRAIL via TRAIL-R2 upmodulation and c-FLIP downmodulation.55 These effects elicited by ATO seem to be related to an inhibitory effect on PI3K: in fact, PI3K inhibitors enhanced TRAIL-R2 and down-modulated c-FLIP expression and sensitized AML cells to TRAIL.55
Particular emphasis was given to the study of APL. In this context, a first study provided evidence that retinoic acid induced TRAIL expression in APL cells and, through this mechanism, induced apoptosis of leukemic cells.56 These findings, however, were not confirmed by a subsequent study since APL blasts, both before and after ATRA treatment, were resistant to TRAIL-mediated apoptosis; furthermore, undifferentiated APL blasts were shown to express TRAIL, whose levels greatly decreased after retinoic acid treatment.50 Finally, studies performed on monocytic cell lines engineered to express the fusion protein PML/RAR-α clearly showed that its induction was associated with reduced TRAIL-R1 expression and reduced TRAIL sensitivity.50 It is important to note that other studies have documented a stimulatory effect of this death ligand on cell proliferation57,58 in a fraction of primary AML leukemic cells resistant to the pro-apoptotic effects of TRAIL.
Fas associated protein with death domain (FADD)
FADD is a key adaptor molecule transmitting the death signal mediated by death receptors.59 FADD is the main signal-transducing intermediate adaptor molecule of several death receptors including Fas, TNF-RI, death receptor 3, TRAIL-R1 and TRAIL-R2. The cytoplasmic tails of all these receptors possess a death domain able, once the receptors are activated, to recruit the death domains of FADD (Figure 4). The function of FADD in death receptor signaling is inhibited by c-FLIP, which, through their death domains, are able to bind to the death domains of FADD, thus preventing its recruitment by activated death receptors (Figure 4). However, FADD is also critical for cell cycle progression and proliferation, activities that are regulated by phosphorylation of its C-terminal Ser 194, which has been implicated in the sensitivity of cancer cells to chemotherapeutic agents and in regulating intracellular localization.60
Studies in animal models of tumor development have shown that FADD may act as a tumor suppressor.59 This interpretation is supported by recent studies showing that about two thirds of leukemic blasts of AML show low levels or no FADD protein. The absence of FADD protein in AML is associated with a poor prognosis.61 Absence or low levels of FADD protein in AML may represent one of the factors responsible for the resistance of AML blasts to Fas and TRAIL-mediated apoptosis. In fact, the large majority of AML patients are resistant to Fas-mediated cell death despite expressing Fas62 and Fas ligand.63 The absence of FADD expression may confer AML blasts a resistance to immune-mediated attack and resistance to some chemo therapeutic drugs, such as anthracyclines and etoposide, which are known to enhance TRAIL-R2 expression and sensitivity to TRAIL and Fas-mediated apoptosis.64
FLIC-inhibitory protein (FLIP)
The death effector domains (DED) present in caspase-8 and FADD are responsible for their reciprocal interaction with consequent formation of death receptor signaling complexes. However, other DED-containing proteins such as FLIP (also known as flame, casper, usurpin) exert a negative role in the control of death receptor signaling. FLIP exhibits extensive homology with pro-caspase-8 and -10,65 containing two N-terminal DED, followed by a pseudo-caspase domain that lacks critical residues required for protease activity, including the catalytic cysteine (Figure 4).66,67 FLIP associates with pro-caspase-8 and competes with pro-caspase-8 and -10 for binding to FADD, thus inhibiting the assembly of the machinery required for death receptor signaling. On the mRNA level c-FLIP exists in multiple splice variants, whereas on the protein level three forms, c-FLIPL (55kDa), c-FLIPS (26kDa) and c-FLIPR (24kDa) have been detected so far (Figure 4).68 All three FLIP contain two NH2-terminus DED domains, essential for their biological activity. The C-terminus of c-FLIPL consists of two inactive caspase-like domains (p20 and p12) (Figure 4). c-FLIPS and c-FLIPR block caspase activation at the level of DISC. c-FLIPL promotes the activation of full length caspase-8, and its initial processing to p43 caspase-8, but inhibits further cleavage to fully active caspase-8. Selective knockdown of c-FLIPL provided definitive evidence that the long variant of c-FLIP functions primarily as an inhibitor of death receptor-mediated apoptosis. 69 c-FLIP is abundantly and constitutively expressed in a wide array of normal cell types, including CD34 hematopoietic progenitors. The transcription of the c-FLIP gene is controlled by NF-κB.70 Overexpression of c-FLIP occurs frequently in a variety of B-cell malignancies, some AML and some chronic lymphocytic leukemias. The presence of elevated c-FLIP levels was strongly related to a poor prognosis in Burkitt’s lymphoma.71 In HTLV-1 Tacute lymphoblastic leukemia (T-ALL) Tax protein induces strong expression of c-FLIP in leukemic blasts, responsible for their resistance to TRAIL and FasL-mediated apoptosis.72
No compound directly able to inhibit c-FLIP has been described. However, several experimental agents have been reported to decrease c-FLIP expression and seemingly to predispose through this mechanism to apoptosis. One of these agents is the synthetic triterpenoid CDDO (2-cyano-3, 12-dioxooleana-1, 9-dien-28-oic acid), another is its derivative CDDO-Im. Both CDDO and CDDO-Im have been shown to inhibit cellular proliferation and induce apoptosis of leukemic cells.50,73 The mechanisms of the pro-apoptotic effects induced by CDDO and CDDOIm are certainly complex and only in part defined. One of the main effects induced by CDDO is ubiquitination and proteasome-dependent destruction of FLIPs in cultured cancer cells72 (Figure 5). CDDO and CDDO-Im down-regulate the c-FLIP and upregulate TRAIL-R1 and TRAIL-R2 in many tumor cell types, rendering them sensitive to TRAIL. CDDO and CDDO-Im are well tolerated in mice, administered alone or in combination with TRAIL.75 CDDO is now under investigation in phase I clinical studies. Recently, the screening of a 50,000-compound library led to the identification of small molecules that sensitize various types of tumor cells to TNF death receptors through c-FLIP down-modulation.76
Initiator caspases in AML
The initiator caspases are able to activate effector caspases through two different molecular mechanisms: either by direct activation or by an indirect mechanism involving a secondary messenger.77 Potential abnormalities of the various initiator caspases in AML have been explored. Pro-caspase-8 has two DED in its N-terminal region, required for interaction with FADD and two catalytic regions termed p18 and p10 (Figure 4). In addition to this standard pro-caspase-8, there are several caspase-8 isoforms: one of them, caspase-8L has several important biological properties. In fact, caspase-8L is shorter than standard caspase-8, lacks the catalytic site of caspase-8, but retains the two N-terminal repeats of the DED.78 Caspase-8L acts as an inhibitor of caspase-8 by interfering with the binding of caspases-8 to FADD.78 The expression of caspase-8L was examined both in hematopoietic progenitor cells and in leukemic blasts derived from AML.79 Interestingly, CD34 cells selectively express the anti-apoptotic splice variant of caspase-8, caspase-8L; in line with this observation, AML with an immature phenotype (i.e., M0 and M1 AML) predominantly express caspase-8L.79 The analysis of caspase-8 expression in AML showed a great heterogeneity in the levels of this caspase, with <10% of cases exhibiting no or low caspase-8 levels.80 The levels of caspase-8 in AML do not seem to be a determinant of drug sensitivity of AML blasts.
There have also been studies exploring possible abnormalities in the expression of other effector caspases in AML. Caspase-2 was found to be involved in several apoptotic settings, including death receptor ligation, endoplasmic reticulum stress and genotoxic stress. Recent studies clearly support a major role of caspase-2 as a proximal mediator of heat shock-induced apoptosis81 and in apoptosis in response to DNA damage.82,83 As observed for caspase-8, caspase-2 levels varied greatly among AML patients and only a minority of them (ranging from 10–20% in the various studies) displayed low pro-caspase-2 levels.84,85 The significance of caspase-2 levels in AML remains unclear since some studies have suggested that high caspase-2 levels, associated with high caspase-3 levels, represent a negative prognostic factor,84 while other studies carried out on pediatric ALL and AML patients have shown that low pro-caspase-2 levels are associated with cellular drug resistance.85 Finally, yet other studies have failed to find any correlation between caspase-2 levels and response to therapy in AML.86 Interestingly, a recent report provided evidence that caspase-2 is localized at the level of the promyelocytic leukemia protein nuclear bodies, nuclear macromolecular complexes involved in the control of apoptosis and disrupted in APL.86 In spite of this localization, caspase-2 activation does not seem to be involved in the mechanism of cell death induced by arsenic trioxide in APL cells.87,88
Effector caspases in AML
Levels of caspases-3 have been explored in AML, providing some interesting findings. Particularly, in a study carried out on newly diagnosed AML increased levels of procaspase-3 in leukemic cells were correlated with a poor prognosis.84 However, patients exhibiting spontaneously active caspases-3 at the time of diagnosis showed improved survival.84 Likewise, in ALL patients increased levels of active caspase-3 correlated with an improved rate of complete remission.89 The poor prognosis among patients with high levels of procaspase-3 may be related to defects in caspase activation with subsequent accumulation of uncleaved caspase-3. The better prognosis observed among patients with increased levels of cleaved caspase-3 may be related to the activation of apoptotic pathways in these leukemic cells.
Bcl-2 in AML
Overexpression of the anti-apoptotic protein Bcl-2, a key member of a family of proteins operative in the mitochondrial pathway of apoptosis, results in resistance to a wide variety of pro-apoptotic stimuli, including radiation, chemotherapeutic agents and corticosteroids.90,91 Bcl-2 was found to be overexpressed in AML patients showing a low rate of complete remission92 and downregulated in patients with the favorable t(8;21) karyotype.93 Furthermore a high ratio of Bcl-2 to Bax, a pro-apoptotic member of the Bcl-2 family of proteins, was shown to be predictive of worse outcome.94 Bcl-2 is over-expressed preferentially among CD34 AML.95 The combined expression of Bcl-2 and MDR-1 identifies M4/M5 AML associated with a very poor prognosis.91 CD34 fractions are more resistant to apoptosis than are corresponding CD34 AML fractions, and this is paralleled by higher Bcl-2, Bcl-XL, Mcl-1 and lower Bax expression.96 Recently a sub-analysis of Bcl-2 in AML patients stratified into lowrisk, intermediate-risk and high-risk groups showed that in low-risk AML patients there is a perfect correlation between Bcl-2 levels and C/EBPα levels: according to these observations it was suggested that the transcription factor C/EBPα, which is mutated in about 10% of AML, could inhibit apoptosis via induction of Bcl-2 expression.97 It seemed conceivable that Bcl-2 downregulation might lower the apoptotic threshold of leukemic cells and, through this mechanism, favor response to chemotherapy98 (Figure 5). A phase I study using an antisense to Bcl-2 in elderly AML patients showed promising results:94 in this study AML patients received induction with oblimersen, cytabarine and daunorubicin and, on achievement of complete remission, consolidation with oblimersen (antisense Bcl-2) and high-dose cytabarine. Of the 29 treated patients, 14 achieved a complete remission; during 12.6 months of follow-up, seven of these patients relapsed.98 A phase II multicenter study of oblimersen plus gentuzamab, a cytotoxic antibody directed against CD33 cells, in relapsed AML patients has been reported: 12 out of 39 treated patients achieved a complete remission.99 Ten of the 12 responders survived more than 6 months.100 Finally, pharmacodynamic studies showed that therapy with oblimersen produced intracellular concentrations of the drug sufficient to down-regulate Bcl-2.100 The findings from these studies have been extended into an ongoing phase III study of about 400 patients with untreated AML who are receiving daunorubicin plus cytosine arabinoside with or without oblimersen.
The MCL-1 gene encodes a 36kDa protein with a carboxy terminal domain similar to Bcl-2. Hoewever, in contrast to Bcl-2, which is a long-lived protein, Mcl-1 has a short half-life. The analysis of Mcl-1 protein expression in AML showed great heterogeneity, but the levels of the protein do not seem to correlate with response to standard chemotherapy.101 Interestingly, Mcl-1 levels increase at the time of a leukemic relapse.101
AML blasts frequently express elevated levels of the anti-apoptotic Bcl-2 family member, Bcl-XL. Elevated levels of Bc-XL have been observed in CD34 AML cells and contribute to chemoresistance of these cells.102 This finding is not surprising since Bc-XL is a target gene of Stat-5, whose constitutive activation is frequently observed in AML.103 Recent studies have shown that AML with Flt3 mutations are usually associated with elevated Bc-XL levels, seemingly induced via constitutive Stat-5 activation. 104,105 Furthermore Flt3 internal-tandem mutants induce BAD phosphorylation.106
The X-linked inhibitor of apoptosis protein (XIAP) in AML
XIAP was found to be expressed in the large majority of adult cases of AML.107,108 In a significant proportion of patients high levels of XIAP were detected. An initial study carried out on 78 adult AML patients suggested a potential relationship between the level of XIAP expression and response to therapy, patients with high XIAP levels having a shorter survival than that observed in patients with low XIAP levels108 However, in a second study, carried out on 172 adult patients with primary AML, the same authors failed to observe a significant correlation between the level of XIAP expression in AML blasts and complete remission rate or overall survival or other clinical parameters.109 In a subsequent study the same authors explored XIAP expression in a group of 92 adult patients with AML and observed that the level of XIAP expression correlated with the monocytic differentiation of leukemic blasts; in this group of patients, those with low levels of XIAP enjoyed a better overall survival in response to standard treatment.110
Studies on the prognostic value of XIAP and survivin levels have also been carried out in pediatric AML. In a group of 45 children with de novo AML an inverse correlation was observed between XIAP levels (high levels of XIAP being observed mostly in M/M1 and M4/M5 AML) and overall survival.111 Survivin expression was detected in the majority of AML, but there was no evidence that its levels in leukemic blasts correlated with the response to treatment or with survival.108, 112
Recently, an in vitro assay was used to explore the downstream part of the mitochondrial pathway of caspase activation in patients with de novo and with relapsing AML. Interestingly, impaired caspase activation was frequent in samples from patients with rapidly relapsing disease, but was rare in samples from patients with newly diagnosed AML.113 Defects in caspase activation did not correlate with IAP or caspase-3 levels.113 Similarly, the simultaneous detection of cytochrome c release, a key initial event in the activation of the intrinsic apoptotic pathway, and caspase-3 activation in leukemic cells seems to represent an important approach to assess the functional integrity of apoptosis signaling in leukemic cells.114 Using this approach it was shown that patients with B-acute lymphoblastic leukemia (B-ALL) with deficient caspase-3 activation have a poorer prognosis than those with normal caspase-3 activation mediated by cytochrome-c release.115
Two approaches are currently being used to develop XIAP inhibitors: (i) antisense oligonucleotides that are in clinical trials and (ii) small molecule inhibitors that are in preclinical development.116,117 Interestingly, polyphenylurea-based small-molecule XIAP antagonists induce apoptosis of AML cells at low micromolar concentrations through a mechanism involving activation of downstream caspase-3 and -7, independently of Bcl-2 levels.118 Triptolide, a diterpenoid isolated from the Chinese herb Tripterygium Wilfordii, decreases XIAP and potently induces caspase-dependent apoptosis of primary AML blasts mediated through the mitochondrial pathway, even at low concentrations.119
Abnormalities in apoptosome formation in AML cells
Following cytochrome c release, the next step in the apoptotic process is apoptosome formation. Correct apoptosome formation requires APAF-1. The primary structure of APAF-1, a key member of the apoptosome, consists of an amino-terminal domain with 12 or 13 WD-40 repeats, a central ATPase domain with homology to CED-4, and a carboxy-terminal caspase recruitment domain (CARD). Four major isoforms of the protein have been identified: only isoforms containing an addition WD-40 repeat between repeats 5 and 6 are able to activate caspase-9 in response to cytochrome c and ATP. Activated APAF-1 interacts with procaspase-9 via a CARD-CARD interaction.
Defects in apoptosome proteins, such as APAF-1, have been involved in the development of some human malignancies, including AML. Studies carried out in leukemic cell lines, as well as in few cases of fresh primary leukemic cells, have shown that leukemic cells express low levels of APAF-1 in a significant proportion of cases and that their pattern of APAF-1 expression directly correlates with ultraviolet light-induced apoptosis.120 Subsequent studies showed that 25% of human leukemic cell lines and 42% of primary AML cells express low levels of APAF-1 mRNA, due to methylation silencing of the APAF-1 gene.121 Treatment of leukemic cells with 5-aza-2’-deoxycytidine, a specific inhibitor of DNA methylation, restored APAF-1 expression in leukemic cells.121 The deregulation of APAF-1 expression in leukemic cells is very complex: in fact, other studies found that APAF-1 mRNA levels did not correlate with APAF-1 protein levels in AML blasts,122 thus suggesting the existence of post-transcriptional defects, in addition to the transcriptional defects.
Conclusions
AML is an aggressive hematologic malignancy. The development and progression of the leukemic disease invariably involve deregulation in the apoptotic response. Despite major efforts during the past 40 years, limited improvements in disease survival have been made. The current primary treatment for AML is chemotherapy, which induces cell death by apoptosis mainly mediated through the intrinsic mitochondrial pathway but also through the extrinsic death receptor pathway, both of which lead to caspase activation and cell death.
The progress made in our understanding of the central role of apoptosis deregulation in the pathogenesis of leukemia and in the development of chemoresistance has launched the academic community, as well as the pharmaceutical and biotechnology industries into a search to identify new compounds for the treatment of leukemia. Targeted therapies that are designed to induce apoptosis in leukemic cells are currently the most promising antileukemia strategies. These strategies are aimed at targeting and killing the leukemia cells specifically with no or limited collateral damage to normal hematopoietic progenitor cells. The elucidation of the molecular apoptotic machinery and of its defects in AML lays the basis for developing new drugs able to trigger apoptosis of leukemia cells (Figure 5).
Figures and Tables
Footnotes
- Author Contributions Both authors contributed equally to this work.
- Conflict of Interest The authors reported no potential conflicts of interest.
References
- Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukaemia. Blood. 2005; 106:1154-63. PubMedhttps://doi.org/10.1182/blood-2005-01-0178Google Scholar
- Frohling S, Scholl C, Gilliland DG, Levine RL. Genetics of myeloid malignancies: pathogenetic and clinical implications. J Clin Oncol. 2005; 23:6285-95. PubMedhttps://doi.org/10.1200/JCO.2005.05.010Google Scholar
- Hanahn P, Weinberg RA. The hallmarks of cancer. Cell. 2000; 100:57-70. PubMedhttps://doi.org/10.1016/S0092-8674(00)81683-9Google Scholar
- Reya T, Morrison SJ, Calrke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001; 414:105-11. PubMedhttps://doi.org/10.1038/35102167Google Scholar
- Lapidot T. A cell initiating human acute myeloid leukemia after transplantation into SCID mice. Nature. 1994; 367:645-8. PubMedhttps://doi.org/10.1038/367645a0Google Scholar
- Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature Med. 1997; 3:730-7. PubMedhttps://doi.org/10.1038/nm0797-730Google Scholar
- Alcalay M, Orleth A, Sebastiani C, Meani N, Chiaradonna F, Casciani C. Common themes in the pathogenesis of acute myeloid leukemia. Oncogene. 2001; 20:5680-94. PubMedhttps://doi.org/10.1038/sj.onc.1204642Google Scholar
- Pearce DJ, Taussing D, Zibara K, Smith LL, Lister TA, Bonnet D. AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML. Blood. 2006; 107:1166-73. PubMedhttps://doi.org/10.1182/blood-2005-06-2325Google Scholar
- Taussing DC, Pearce DJ, Simpson C, Rohatiner AZ, Lister TA, Kelly G. Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia. 2005; 106:4086-92. Google Scholar
- Klampfer L, Zhang J, Zelenetz AO, Uchida H, Nimer SD. The AML1/ETO fusion protein activates transcription of BCL-2. Proc Natl Acad Sci USA. 1996; 95:11863-8. https://doi.org/10.1073/pnas.95.20.11863Google Scholar
- Britos-Bray M, Ramirez M, Cao W, Wang X, Liu PP, Civin CI, Friedman AD. CBF-SMMHC, expressed in M4eo acute myeloid leukaemia, reduces p53 induction and slows apoptosis in hematopoietic cells exposed to DNA-damaging agents. Blood. 1998; 92:4344-52. PubMedGoogle Scholar
- Pandolfi PP. Oncogenes and tumor suppressors in the molecular pathogenesis of acute promyelocytic leukaemia. Hum Mol Genet. 2001; 10:769-75. PubMedhttps://doi.org/10.1093/hmg/10.7.769Google Scholar
- Adler HT, Chinery R, Wu DY, Sussick SJ, Payne JM, Fornace AJ. Leukemic HRX fusion proteins inhibit GADD34-induced apoptosis and associate with the GADD34 and hSNF5/INH proteins. Mol Cell Biol. 1999; 19:7050-60. PubMedGoogle Scholar
- Testa U, Grignani F, Samoggia P, Zanetti C, Riccioni R, Lo Coco F. The PML/RAR_ fusion protein inhibits tumor necrosis factor-α-induced apoptosis in U937 cells and acute promyelocytic leukemia blasts. J Clin Invest. 1998; 101:2278-89. PubMedGoogle Scholar
- Riccioni R, Pasquini L, Mariani G, Saulle E, Rossini A, Diverio D. TRAIL decoy receptors mediate resistance of acute myeloid leukemia cells to TRAIL. Haematologica. 2005; 90:612-24. PubMedGoogle Scholar
- Falini B, Mecucci C, Tiacci F, Alacalay M, Lo-Coco F, Pelicci PG. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005; 352:254-66. PubMedhttps://doi.org/10.1056/NEJMoa041974Google Scholar
- Verhaak RG, Goudswaard CS, Lowenberg B, Valk PJ. Mutations in nucleophosmin (NPM1) in acute myeloid leukaemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favourable prognostic significance. Blood. 2005; 106:3747-54. PubMedhttps://doi.org/10.1182/blood-2005-05-2168Google Scholar
- Lambert B, Buckle M. Characterization of the interface between nucleophosmin (NPM) and p53: potential role in p53 stabilization. FEBS Lett. 2006; 6580:345-50. Google Scholar
- Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nature Rev Cancer. 2006; 6:493-505. PubMedhttps://doi.org/10.1038/nrc1885Google Scholar
- Colombo E, Bonetti P, Falini B, Pelicci PG. Nucleophosmin is required for DNA integrity and p19Arf protein stability. Mol Cell Biol. 2005; 25:8874-86. PubMedhttps://doi.org/10.1128/MCB.25.20.8874-8886.2005Google Scholar
- Stirewalt DL, Radich JP. The role of FLT3 in hemopoietic malignancies. Nat Rev Cancer. 2003; 3:650-65. PubMedhttps://doi.org/10.1038/nrc1169Google Scholar
- Minami Y, Yamamoto K, Kiyoi H, Ueda R, Saito H, Naoe T. Different antiapoptotic pathways between wild-type and mutated FLT3: insights into therapeutic targets in leukemia. Blood. 2003; 102:2969-75. PubMedhttps://doi.org/10.1182/blood-2002-12-3813Google Scholar
- Kim KT, Baird K, Ahn JY, Meltzer P, Lilly M, Levis M. Pim-1 is upregulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival. Blood. 2005; 105:1759-67. PubMedhttps://doi.org/10.1182/blood-2004-05-2006Google Scholar
- Kohl TM, Schnittger S, Ellwart JW. Kit exon 8 mutations associated with core-binding factor (CBF)-acute myeloid leukemia (AML) cause hyperactivation of the receptor in response to stem cell factor. Blood. 2005; 105:3319-21. PubMedhttps://doi.org/10.1182/blood-2004-06-2068Google Scholar
- Corbacioglu S, Kilic M, Westhoff MA, Reinhardt D, Fulda S, Debatin KM. Newly identified c-kit receptor tyrosine kinase ITD in childhood AML induces ligand independent growth and is responsive to a synergistic effect of imatinib and rapamycin. Blood. 2006; 108:3504-13. PubMedhttps://doi.org/10.1182/blood-2006-05-021691Google Scholar
- Schnittger S, Kohl TM, Haferlach T, Kern W, Hiddemann W, Spiekemann K. KIT-816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006; 107:1790-9. Google Scholar
- Paschka P, Marcucci G, Ruppert AS, Mrozek K, Chen H, Kolitz JE. Adverse prognostic significance of KIT mutations in adult acute myeloid leukaemia with inv(16) and t(8;21): a cancer and leukaemia group B study. J Clin Oncol. 2006; 24:3904-11. PubMedhttps://doi.org/10.1200/JCO.2006.06.9500Google Scholar
- Reuter CW, Morgan MA, Bergmann L. Targeting the Ras signaling pathway. A rational mechanism-based treatment for hematologic malignancies?. Blood. 2000; 96:1665-9. Google Scholar
- Bacher U, Haferlach T, Schoch C, Kern W, Schnittger S. Implications of NRAS mutations in AML: a study of 2502 patients. Blood. 2006; 107:3847-53. PubMedhttps://doi.org/10.1182/blood-2005-08-3522Google Scholar
- Parikh C, Subrahmanyam R, Ren R. Oncogenic NRAS rapidly and efficiently induces CMML- and AML like diseases in mice. Blood. 2006. Google Scholar
- Traver D, Akashi K, Weissman IL, Lagasse E. Mice defective in two apoptosis pathways in the myeloid lineage develop acute myeloblastric leukaemia. Immunity. 1998; 9:47-57. PubMedhttps://doi.org/10.1016/S1074-7613(00)80587-7Google Scholar
- Bullinger L, Valk PJM. Gene expression profiling in acute myeloid leukaemia. J Clin Oncol. 2005; 23:6296-305. PubMedhttps://doi.org/10.1200/JCO.2005.05.020Google Scholar
- Valk PJM, Verhaak RGW, Lowenberg B, Ruud D. Prognostically useful gene-expression profiles in acute myeloid leukaemia. N Engl J Med. 2004; 350:1617-28. PubMedhttps://doi.org/10.1056/NEJMoa040465Google Scholar
- Bullinger L, Dohner K, Bair E, Frohling S, Schlenk RF, Tibshirani R. Use of gene-expression profiling to identify prognostic subclasses in adult myeloid leukaemia. N Engl J Med. 2004; 350:1605-16. PubMedhttps://doi.org/10.1056/NEJMoa031046Google Scholar
- Wilson CS, Davidson GS, Martin SB, Andries E, Potter J, Harvey R. Gene expression profiling of adult acute myeloid leukaemia identifies novel biologic clusters for risk classification and outcome prediction. Blood. 2006; 108:685-96. PubMedhttps://doi.org/10.1182/blood-2004-12-4633Google Scholar
- Min YH, Cheong JW, Kim JY. Cytoplasmic mislocalization of p27 Kip 1 protein is associated with constitutive phosphorylation of Akt or protein kinase B and poor prognosis in acute myelogenous leukaemia. Cancer Res. 2004; 64:5225-31. PubMedhttps://doi.org/10.1158/0008-5472.CAN-04-0174Google Scholar
- Martelli AM, Nyakjern M, Bortul R, Tarzani PL, Evalengelisti C, Cocco L. Phospha phoinositide 3-kinase/Aky signaling pathway and its therapeutical impklications for human acute myeloid leucemia. Leukemia. 2006; 20:911-28. PubMedhttps://doi.org/10.1038/sj.leu.2404245Google Scholar
- Xu Q, Simpson SE, Scialla TJ, Bagg A, Carroll M. Survival of acute myeloid leucemia cell requires PI3 kinase activation. Blood. 2003; 102:972-80. PubMedhttps://doi.org/10.1182/blood-2002-11-3429Google Scholar
- Kim SC, Hahn JS, Min YH. Constitutive activation of extracellular signal-regulated kinase in human acute leukemias: combined role of activation of MEK, hyperexpression of extracellular signal-regulated kinase, and downregulation of a phosphatase, PACI. Blood. 1999; 93:3893-9. PubMedGoogle Scholar
- Milella M, Kornblau SM, Estrov Z. Therapeutic targeting of the MEK/MAPK signal transduction module in acute myeloid leukemia. J Clin Invest. 2001; 108:851-9. PubMedhttps://doi.org/10.1172/JCI200112807Google Scholar
- Kornblau SM, Womble M, Qiu YH, Jackson E, Chen W, Konopleva M. Simultaneous activation of multiple signal transduction pathways confers poor prognosis in acute myelogenous leukemia. Blood. 2006; 108:2358-65. PubMedhttps://doi.org/10.1182/blood-2006-02-003475Google Scholar
- Bazzoni F, Butler B. The tumor necrosis factor ligand and receptor families. N Engl J Med. 1996; 334:1717-25. PubMedhttps://doi.org/10.1056/NEJM199606273342607Google Scholar
- Ogasarawa J, Watanabe-Fukunaga R, Adachi M, Matsuzawa A, Kasugai T, Kita mura Y. Lethal effect of the anti-Fas antibody in the mice. Nature. 1993; 364:806-9. PubMedhttps://doi.org/10.1038/364806a0Google Scholar
- Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA, Marsters SA. Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest. 1999; 104:155-62. PubMedhttps://doi.org/10.1172/JCI6926Google Scholar
- Kelley S, Harris L, Xie D, DeForge L, Totpal K, Bussiere J. Preclinical studies to predict the disposition of Apo2L/tumor necrosis factor-related apoptosis-inducing ligand in humans: characterization on in vivo efficacy, pharmacokinetics, and safety. J Pharmacol Exp Ther. 2001; 299:31-8. PubMedGoogle Scholar
- Yagita H, Takeda K, Hayakawa Y, Smyth M, Okumura K. TRAIL and its receptors as targets for cancer therapy. Cancer Sci. 2004; 95:777-83. PubMedhttps://doi.org/10.1111/j.1349-7006.2004.tb02181.xGoogle Scholar
- Pulac L, Kanakara P, Humphreys R, Alder son R, Dobson C, Salcedo T. HGS-ETR1, a fully human TRAIL-receptor 1 monoclonal antibody, induces cell death in multiple tumor types in vitro and in vivo. Br J Cancer. 2005; 92:1430-41. PubMedhttps://doi.org/10.1038/sj.bjc.6602487Google Scholar
- Le L.Paper presented at: ; Google Scholar
- Wuchter C, Krappmann D, Cai Z, Ruppert V, Scheidereit C, Dorken B. In vitro susceptibility to TRAIL induced apoptosis of acute leukemia cells in the context of TRAIL receptor gene expression and constitutive NF-κB activity. Leukemia. 2001; 15:921-8. PubMedhttps://doi.org/10.1038/sj.leu.2402131Google Scholar
- Riccioni R, Pasquini L, Mariani G, Saulle E, Rossini A, Diverio D. TRAIL decoy receptors mediate resistance of acute myeloid leukemia cells to TRAIL. Haematologica. 2005; 90:612-24. PubMedGoogle Scholar
- Cappellini A, Mantovani I, Tazzari PL, Grafone T, Martinelli G, Cocco L, Martelli AM. Application of flow cytometry to molecular medicine: detection of tumor necrosis factorrelated apoptosis-inducing ligand receptors in acute myeloid leukemia blasts. Int J Mol Med. 2005; 16:1041-8. PubMedGoogle Scholar
- Suh WS, Kim YS, Schimmer AD, Kitada S, Minden M, Andreef M. Synthetic triterpenoids activate a pathway for apoptosis in AML cells involving downregulation of FLIP and sensitization to TRAIL. Leukemia. 2003; 17:2122-9. PubMedhttps://doi.org/10.1038/sj.leu.2403112Google Scholar
- Nebbioso A, Clarke N, Voltz E, Germain E, Ambrosino C, Bontempo P. Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leucemia cells. Nat Med. 2005; 11:77-84. PubMedhttps://doi.org/10.1038/nm1161Google Scholar
- Hasegawa H, Yamada Y, Masuda M, Takasu N, Kamihira S. Dihydroflavonol BB-1, an extract of natural plant Blumea balsamifera, abrogates TRAIL resistance in leukemia cells. Blood. 2006; 107:679-88. PubMedhttps://doi.org/10.1182/blood-2005-05-1982Google Scholar
- Szegezdi E, Cahill S, Meyer M, O’Dwyer M, Samali A. TRAIL sensitization by arsenic trioxide is caspases-8 dependent and involves modulation of death receptor component and Akt. Br J Cancer. 2006; 94:398-406. PubMedhttps://doi.org/10.1038/sj.bjc.6602954Google Scholar
- Altucci L, Rossin A, Raffelsberger W, Reitmair A, Chomienne C, Gronemeyer H. Retinoic acid-induced apoptosis in leukemia cells is mediated by paracrine action of tumorselective death ligand TRAIL. Nat Med. 2001; 7:680-6. PubMedhttps://doi.org/10.1038/89050Google Scholar
- Ehrhardt H, Fulda S, Schmid I, Hiscott J, Debatin KM, Jeremias I. TRAIL induced survival and proliferation in cancer cells resistant towards TRAIL-induced apoptosis mediated by NF- B. Oncogene. 2003; 22:3842-52. PubMedhttps://doi.org/10.1038/sj.onc.1206520Google Scholar
- Baader E, Toloczko A, Fuchs U, Schmid I, Beltinger C, Ehrhardt H. Tumor necrosis factor-related apoptosis-inducing ligand-mediated proliferation of tumor cells with receptor-proximal apoptosis defects. Cancer Res. 2005; 65:7888-95. PubMedGoogle Scholar
- Tourneur L, Buzyn A, Chiocchia G. FADD adaptor in cancer. Med Immunol. 2005; 4:1-15. PubMedhttps://doi.org/10.1186/1476-9433-4-1Google Scholar
- Alappat EC, Feig C, Boyerinas B, Volkland J, Samuels M, Murmann AE. Phosphorylation of FADD at serine 194 by CKI regulates its nonapoptotic activities. Mol Cell. 2005; 19:321-32. PubMedhttps://doi.org/10.1016/j.molcel.2005.06.024Google Scholar
- Tourneur L, Delluc S, Levy V, Valensi F, Radford-Weiss I, Legrand O. Absence or low expression of Fasassociated protein with death domain in acute myeloid leukaemia cells predicts resistance to chemotherapy and poor outcome. Cancer Res. 2004; 64:8101-8. PubMedhttps://doi.org/10.1158/0008-5472.CAN-04-2361Google Scholar
- Iijima N, Mioyamura K, Itou T, Tanimoto M, Sobue R, Saito H. Functional expression of Fas (CD95) in acute myeloid leukaemia cells in the context of CD34 and CD38 expression: possible correlation with sensitivity to chemotherapy. Blood. 1997; 90:4901-9. PubMedGoogle Scholar
- Lewis NR, Pallis M, Russell NH. Fas receptor-Fas ligand system is independent of both CD34 status and chemosensitivity in acute myeloid leukaemia. Exp Hematol. 2000; 28:535-42. PubMedhttps://doi.org/10.1016/S0301-472X(00)00132-6Google Scholar
- Lacour S, Hammann A, Wotawa A, Corcos L, Solary E, Dimanche-Boitrel MT. Anti cancer agents sesitize tumor cells to tumor necrosis factor-related apoptosis-inducing ligand-mediated caspases-8 activation and apoptosis. Cancer Res. 2001; 61:1645-51. PubMedGoogle Scholar
- Budd RC, Yeh WC, Tschopp J. cFLIP regulation of lymphocyte activation and development. Nat Rev Immunol. 2006; 6:159-204. PubMedhttps://doi.org/10.1038/nri1776Google Scholar
- Wallach D. Placing death under control. Nature. 1997; 388:123-6. PubMedhttps://doi.org/10.1038/40516Google Scholar
- Tschopp J, Irmler M, Thome M. Inhibition of Fas death signals by FLIPs. Curr Opin Immunol. 1998; 10:552-8. PubMedhttps://doi.org/10.1016/S0952-7915(98)80223-9Google Scholar
- Gloks A, Brenner D, Fritsch C, Krammer PH, Lavrik IN. c-FLIPR, a new regulator of death receptormediated caspase-8 activation and apoptosis. J Biol Chem. 2005; 280:14507-13. PubMedhttps://doi.org/10.1074/jbc.M414425200Google Scholar
- Sharp DA, Lawrence DA, Ashkenazi A. Selective knockdown of the long variant of cellular FLICE inhibitory protein augments death receptormediated caspases-8 activation and apoptosis. J Biol Chem. 2005; 280:19401-9. PubMedhttps://doi.org/10.1074/jbc.M413962200Google Scholar
- Micheau O, Lens S, Gaide O, Alevidopoulos K, Tschopp J. NF-κB signals induce the expression of c-FLIP. Mol Cell Biol. 2001; 21:5299-305. PubMedhttps://doi.org/10.1128/MCB.21.16.5299-5305.2001Google Scholar
- Valnet-Rabier MB, Chalier B, Thiebault S, Angonin R, Margueritte G, Moyugin C. c-FLIP protein expression in Burkitt’s lymphomas is associated with a poor clinical outcome. Br J Haematol. 2005; 128:767-73. PubMedhttps://doi.org/10.1111/j.1365-2141.2005.05378.xGoogle Scholar
- Krueger A, Fas SC, Giaisi M, Bleumink M, Merling A, Stumpf C. HTLV-1 tax protects against CD95-mediated apoptosis by induction of the cellular FLIC-inhibitory protein (c-FLIP). Blood. 2006; 107:3933-9. PubMedhttps://doi.org/10.1182/blood-2005-06-2567Google Scholar
- Konopleva M, Tsao T, Estrov Z, Lee RM, Wang RY, Belmont J. The synthetic CDDO induces caspasedependent and independent apoptosis in acute myelogenous leukaemia. Cancer Res. 2004; 64:7927-33. PubMedhttps://doi.org/10.1158/0008-5472.CAN-03-2402Google Scholar
- Kim Y, Suh N, Sporn MB, Reed JC. An inducible pathway for degradation of FLIP protein sensitizes tumor cells to TRAIL-induced apoptosis. J Biol Chem. 2003; 277:22320-9. https://doi.org/10.1074/jbc.M202458200Google Scholar
- Hyer ML, Croxton R, Krajewska M, Sporn MB, Cryns VL, Zapata JM. Synthetic triterpenoids cooperate with tumor necrosis factor-related apoptosis-inducing ligand to induce apoptosis of breast cancer cells. Cancer Res. 2005; 65:4799-808. PubMedhttps://doi.org/10.1158/0008-5472.CAN-04-3319Google Scholar
- Schimmer AD, Thomas MP, Hurren R, Gronda M, Pellecchia M, Pond GR. Identification of small molecules that sensitize resistant tumor cells to tumor factor-family death receptors. Cancer Res. 2006; 66:2367-75. PubMedhttps://doi.org/10.1158/0008-5472.CAN-05-1061Google Scholar
- Ho Pk, Hawkins CJ. Mammalian initiator apoptotic caspases. FEBS J. 2005; 272:5436-53. PubMedhttps://doi.org/10.1111/j.1742-4658.2005.04966.xGoogle Scholar
- Himeji D, Horiuchi T, Tsukamoto H, Hayashi K, Watanabe T, Harada M. Characterization of caspase-8L: a novel isoform of caspase-8 that behaves as an inhibitor of the caspase cascade. Blood. 2002; 99:4070-8. PubMedhttps://doi.org/10.1182/blood.V99.11.4070Google Scholar
- Mohr A, Zwacka RM, Jarmy G, Buneker C, Schrezenmeier H, Dohner K. Caspase-8L expression protects CD34+ hematopoietic progenitor cells and leukemic cells from CD95-mediated apoptosis. Oncogene. 2005; 24:2421-9. PubMedhttps://doi.org/10.1038/sj.onc.1208432Google Scholar
- Svingen PA, Karp JE, Kraiewski S, Mesner PW, Gore SD, Burke PJ. Evaluation of Apaf-1 and procaspases- 2, -3, -7, and -9 as potential prognostic markers in acute leukaemia. Blood. 2000; 96:3922-31. PubMedGoogle Scholar
- Tu S, McStay GP, Boucher LM, Mak T, Beere HM, Green DR. In situ trapping of activated initiator caspases reveals a role for caspase-2 in heat shock-induced apoptosis. Nat Cell Biol. 2006; 8:72-7. PubMedhttps://doi.org/10.1038/ncb1340Google Scholar
- 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. PubMedhttps://doi.org/10.1126/science.1095432Google Scholar
- Janssens S, Tinel A, Lippens S, Tschopp J. PIDD mediates NF- B activation in response to DNA damage. Cell. 2005; 123:1079-92. PubMedhttps://doi.org/10.1016/j.cell.2005.09.036Google Scholar
- Estrov Z, Thall PF, Talpaz M, Estey EH, Kantarjian HM, Harris D. Caspase 2 and caspase 3 protein levels as predictors of survival in acute myelogenous leukaemia. Blood. 1998; 92:3090-7. PubMedGoogle Scholar
- Holleman A, Den Boer M, Kazemeier KM, Beverloo HB, Von Bergh A, Janka-Schaub GE. Decreased PARP and procaspase-2 protein levels are associated with cellular drug resistance in childhood lymphoblastic leukemia. Blood. 2005; 106:1817-23. PubMedhttps://doi.org/10.1182/blood-2004-11-4296Google Scholar
- Tang J, Xie W, Yang X. Association of caspase-2 with the promyelocytic leukemia protein nuclear bodies. Cancer Biol Ther. 2006; 4:645-9. Google Scholar
- Scholz C, Wieder T, Starck L, Essmann F, Schulze-Osthoff K, Dorken B. Arsenic trioxide triggers a regulated form of caspaseindependent necrotic cell death via the mitochondrial death pathway. Oncogene. 2005; 24:1904-13. PubMedhttps://doi.org/10.1038/sj.onc.1208233Google Scholar
- Scholz C, Richter A, Lehmann M, Schulze-Osthoff K, Dorken B, Daniel PT. Arsenic trioxide induces regulated, death receptor-independent cell death through a Bcl-2-controlled pathway. Oncogene. 2005; 24:7031-42. PubMedhttps://doi.org/10.1038/sj.onc.1208868Google Scholar
- Fardel S, Thall PF, Kantarjian HM. Caspase 2 and caspases 3 as predictors of complete remission and survival in adults with acute lymphoblastic leukaemia. Clin Cancer Res. 2005; 5:4041-7. Google Scholar
- Campos L, Rouault JP, Sabido O. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood. 1993; 81:3091-6. PubMedGoogle Scholar
- Andreef M, Konopleva M. Mechanisms of drug resistance in AML. Cancer Treat Res. 2002; 112:237-62. PubMedGoogle Scholar
- Banker DE, Radich J, Becker E. The t(8;21) translocation is not consistently associated with high Bcl-2 expression in de novo acute myeloid leukemias of adults. Clin Cancer Res. 1998; 4:3051-62. PubMedGoogle Scholar
- Del Poeta G, Venditti A, Del Principe MI. Amount of spontaneous apoptosis detected by Bax/Bcl-2 ratio predicts outcome in acute myeloid leukemia (AML). Blood. 2003; 101:2125-31. PubMedhttps://doi.org/10.1182/blood-2002-06-1714Google Scholar
- Venditti A, Del Poeta G, Maurillo L, Buccisano F, Del Principe MI, Mazzone C. Combined analysis of bcl-2 and MDR1 proteins in 256 cases of acute myeloid leukemia. Haematologica. 2004; 89:934-9. PubMedGoogle Scholar
- Konopleva M, Tari AM, Estrov Z, Harris D, Xie Z, Zhao S. Liposomal Bcl-2 antisense oligonucleotides enhance proliferation, sensitize acute myeloid leukemia to cytosine-arabinoside, and induce apoptosis independent of other antiapoptotic proteins. Blood. 2000; 95:3929-38. PubMedGoogle Scholar
- Van Stijn A, Van Der Pol MA, Kok A, Bontje PM, Roemen G, Beelen R. Differences between the CD34+ and CD34-blast compartments in apoptosis resistance in acute myeloid leukaemia. Haematologica. 2003; 88:497-508. PubMedGoogle Scholar
- Paz-Priel I, Cai DH, Wang D, Kowalski J, Gombart AF, Koeffler HP. CCAAT/Enhancer binding protein _ (C/EBPα) and C/EBPα myeloid oncoproteins induce Bcl-2 via interaction of their basic regions with nuclear factor-κB p50. Mol Cancer Res. 2005; 3:585-96. PubMedhttps://doi.org/10.1158/1541-7786.MCR-05-0111Google Scholar
- Marcucci G, Stock W, Dai G. Phase I study of oblimersed sodium, an antisense to Bcl-2, in untreated older patients with acute myeloid leukemia: pharmacokinetics, pharmacodynamics, and clinical activity. J Clin Oncol. 2005; 23:3404-11. PubMedhttps://doi.org/10.1200/JCO.2005.09.118Google Scholar
- Moore LO, Seiter K, Kolitz JE, Stock W, Yu R, Frankel SR. Phase 2 study of oblimersen sodium (G31\39; Bcl-2 antisense; Gena sense) plus gentuzumab ozogamicin (Myelotarg) in elderly patients with relapsed acute myeloid leukemia (AML). Blood. 2004; 104:865. PubMedhttps://doi.org/10.1182/blood-2003-11-3750Google Scholar
- Dai G, Chan KK, Liu S. Cellular uptake and intracellular levels of the Bcl-2 antisense G3139 in cultured cells and treated patients with acute myeloid leukemia. Clin Cancer Res. 2005; 11:2998-3008. PubMedhttps://doi.org/10.1158/1078-0432.CCR-04-1505Google Scholar
- Kaufmann SH, Karp JE, Svingen PA, Kraiewski S, Burke PJ, Gore SD, Reed JC. Elevated expression of the apoptotic regulator Mcl-1 at the time of leukemic relapse. Blood. 1998; 91:991-1000. PubMedGoogle Scholar
- Konopleva M, Zhao S, Hu W, Jiang S, Snell V, Weidner D. The antiapoptotic genes Bcl-X(L) and Bcl-2 are overexpressed and contribute to chemoresistance of non-proliferating leukaemic CD34+ cells. Br J Haematol. 2002; 118:521-34. PubMedhttps://doi.org/10.1046/j.1365-2141.2002.03637.xGoogle Scholar
- Birkenkamp KU, Geugien M, Lemmink HH, Kruj W, Vellenga E. Regulation of constitutive STAT5 phosphorylation in acute myeloid leukaemia blasts. Leukemia. 2001; 15:1923-31. PubMedhttps://doi.org/10.1038/sj.leu.2402317Google Scholar
- Minami Y, Yamamaoto K, Kiyoi H, Ueada R, Saito H, Naoe T. Different antiapoptotic pathways between wild-type and mutated FLT3: insights into therapeutic targets in leukemia. Blood. 2003; 102:2969-75. PubMedhttps://doi.org/10.1182/blood-2002-12-3813Google Scholar
- Bragintseva K, Geisenhof S, Kern R, Eichenlaub S, Reindl C, Elwart JW. FLT3-ITD-TKD dual mutants associated with AML confer resistance to FLT3 PTK inhibitors and cytotoxic agents by overexpression of Bcl-X(L). Blood. 2005; 105:3679-85. PubMedhttps://doi.org/10.1182/blood-2004-06-2459Google Scholar
- Yang X, Liu L, Sternberg D, Tang L, Galinsky I, DeAngelo D. The FLT3 internal tandem duplication mutation prevents apoptosis in interleukin-3-deprived BaF3 cells due to protein kinase A and ribosomal S6 kinase 1-mediated BAD phosphorylation a serine 112. Cancer Res. 2005; 65:7338-47. PubMedhttps://doi.org/10.1158/0008-5472.CAN-04-2263Google Scholar
- Tamm I, Kornblau SM, Segal H, Krajewski S, Welsh K, Kitada S. Expression and prognostic significance of IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res. 2000; 6:1796-803. PubMedGoogle Scholar
- Carter BZ, Kornblau SM, Tsao T, Wang RY, Schober WD, Milella M. Caspase-independent cell death in AML: caspase inhibition in vitro with pan-caspase inhibitors or in vivo by XIAP or Survivin does not affect cell survival or prognosis. Blood. 2003; 102:4179-86. PubMedhttps://doi.org/10.1182/blood-2003-03-0960Google Scholar
- Tamm I, Richter S, Oltersdorf D, Creutzig U, Harbott J, Scolz F. High expression levels of X-linked inhibitor of apoptosis protein and surviving correlate with poor overall survival in childhood de novo acute myeloid leukaemia. Clin Cancer Res. 2004; 10:3737-44. PubMedhttps://doi.org/10.1158/1078-0432.CCR-03-0642Google Scholar
- Tamm I, Richter S, Scholtz F, Schmelz K, Oltersdorf D, Karawajew L. XIAP expression correlates with monocytic differentiation in adult de novo AML: impact on prognosis. Hematol J. 2004; 5:489-95. PubMedhttps://doi.org/10.1038/sj.thj.6200549Google Scholar
- Tamm I, Richter S, Oltersdiorf D, Creutzig U, Harbott J, Scholz F. High expression levels of X-linked inhibitor of apoptosis protein and surviving correlate with poor overall survival in childhood de novo acute myeloid leukaemia. Clin Cancer Res. 2004; 10:3737-44. PubMedhttps://doi.org/10.1158/1078-0432.CCR-03-0642Google Scholar
- Adida C, Recher C, Raffoux E. Expression and prognostic significance of IAP family genes in de novo acute myeloid leukemia. Br J Haematol. 2000; 111:196-203. PubMedhttps://doi.org/10.1046/j.1365-2141.2000.02328.xGoogle Scholar
- Gronda M, Brandwein J, Minden MD, Pond GR, Schub AC, Wells RA. Assessment of the downstream portion of the mitochondrial pathway of caspase activation in patients with acute myeloid leukemia. Apoptosis. 2005; 10:1285-94. PubMedhttps://doi.org/10.1007/s10495-005-2764-4Google Scholar
- Mohr A, Zwacka RM, Debatin KM, Stahnke K. A novel method fort he combined flow cytometric analysis of cell cycle and cytochrome c release. Cell Death Differ. 2004; 11:1153-4. PubMedhttps://doi.org/10.1038/sj.cdd.4401480Google Scholar
- Meyer LH, Karawajew L, Schrappe M, Ludwig WD, Debatin KM, Stahnke K. Cytochrome c related caspase-3 activation determines treatment response and relapse in childhood precursor B-cell ALL. Blood. 2006. Google Scholar
- Schimmer AD, Dalili S, Batey RA, Riedl SJ. Targeting XIAP for the treatment of malignancy. Cell Death Differ. 2006; 13:179-88. PubMedhttps://doi.org/10.1038/sj.cdd.4401826Google Scholar
- Schimmer AD, Dalili S. Targeting the IAP family of caspase inhibitors as an emerging therapeutic strategy. American Society Hematology. 2005;215-9. Google Scholar
- Carter BZ, Gronda M, Wang Z, Chun K, Reed JC, Schimmer AD. Small-molecule XIAP inhibitors derepress downstream effector caspases and induce apoptosis of acute myeloid leukaemia cells. Blood. 2005; 105:4043-50. PubMedhttps://doi.org/10.1182/blood-2004-08-3168Google Scholar
- Carter BZ, Mak DH, Schober WD, McQueen T, Harris D, Estrov Z. Triptolide induces caspasedependent cell death mediated via the mitochondrial pathway in leukemic cells. Blood. 2006. Google Scholar
- Jia L, Srinivasula S, Liu FT, Newland AC, Fernandes-Alnemri T, Alnemri ES. Apaf-1 protein deficiency confers resistance to cytochrome cdependent apoptosis in human leukemic cells. Blood. 2001; 98:414-21. PubMedhttps://doi.org/10.1182/blood.V98.2.414Google Scholar
- Furukawa Y, Sutheesophon K, Wada T, Nishimura M, Saito Y, Hishii H. Methylation silencing of the Apaf-1 gene in acute leukaemia. Mol Cancer Res. 2005; 3:325-34. PubMedhttps://doi.org/10.1158/1541-7786.MCR-04-0105Google Scholar
- Fu WN, Bertoni F, Kelsey SM, McElwaine SM, Cotter FE, Newland AC. Role of DNA methylation in the suppression of Apaf-1 protein in human leukaemia. Oncogene. 2003; 22:451-5. PubMedhttps://doi.org/10.1038/sj.onc.1206147Google Scholar