U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].

Show details

Caspase Activation in Cancer Therapy

and .

Different anticancer therapies including cytotoxic drugs, γ-irradiation, suicide gene therapy, or immunotherapy appear to induce tumor cell death by activating key elements of apoptosis, the cell's intrinsic death program. Activation of the cascade of proteolytic enzymes known as caspases is a critical component of the execution phase of cell death in most forms of apoptosis. Two main caspase cascades, one triggered by death receptor stimulation and the other one initiated at the mitochondria, have been identified in response to various inducers of cellular stress such as DNA damage. Activation of caspases and apoptosis is tightly regulated at several levels, e.g., by Bcl-2 family members, by inhibitor of apoptosis proteins (IAPs) and upstream inhibitors such as FLIP. Failure to activate apoptotic pathways in response to drug treatment may lead to resistance of tumors cells to anticancer therapies. Therefore, factors affecting caspase activation might be important determinants of drug sensitivity. In addition to caspase-dependent apoptosis, caspase-independent forms of cell death may also play a role for treatment response. Insights into the mechanisms regulating caspase activation as well as other forms of cell death pathways provide a molecular basis for novel strategies targeting resistance of tumor cells.

Introduction

Killing of tumor cells by diverse cytotoxic approaches such as anticancer drugs, γ-irradiation, suicide genes, or immunotherapy has been shown to be mediated through induction of apoptosis in target cells.1–7 Apoptosis or programmed cell death occurs upon the activation of distinct, intrinsic cell death programs under certain physiological and pathological situations.8 The underlying mechanism for initiation of an apoptosis response upon cytotoxic therapy may be different for various stimuli and is only partially understood. However, damage to DNA or to other critical molecules and/or subcellular structures appears to be a common early hit by some inducers which is then propagated by the cellular stress response.9 Multiple stress-inducible molecules, e.g., JNK, MAPK/ERK, NF-κB or ceramide may have a profound impact on apoptosis pathways.10–12 On the other hand, cytotoxic T cells or NK cells may release compounds such as granzyme B which directly activates downstream apoptosis effector mechanisms inside the cell.8 Apoptosis is characterized by typical morphological and biochemical hallmarks including cell shrinkage, nuclear DNA fragmentation and membrane blebbing.8 Proteolytic enzymes such as caspases play an important role as effector molecules in apoptosis including cytotoxic therapy-induced cell death.13–19 Because of the potential detrimental effects on cell survival in case of inappropriate caspase activity, activation of caspases has to be tightly controlled. The antiapoptotic mechanisms regulating activation of caspases have also been postulated to be involved in drug resistance of tumor cells.20–22 However, the concept that anticancer therapies primarily act by triggering apoptosis has also been challenged, since a consistent link between the ability of tumor cells to undergo apoptosis in vitro and their susceptibility to anticancer therapy in vivo has not always been observed.23 Therefore, nonapoptotic modes of cell death, e.g., necrosis or some forms of cell death that cannot be easily classified, may mediate the cell death response to cytotoxic therapy.24,25 Also, non-caspase-dependent apoptosis has been found to be induced by anticancer drugs in some cells.26–28 Thus, a better understanding of these diverse modes of tumor cell death following cytotoxic therapies will provide a molecular basis for new strategies targeting caspase-dependent and independent death pathways in apoptosis-resistant forms of cancer.

Caspases as Central Death Effector Molecules

Most signaling pathways activated by anticancer drugs ultimately result in activation of caspases, a family of cysteine proteases that act as common death effector molecules in various forms of cell death (Fig. 1).8,13–18 12 human caspases with different substrate specificity have so far been identified that cleave next to aspartate residues.13–18 Caspases are involved in apoptosis signaling and also in cytokine processing.13–18 Caspases are synthesized as inactive zymogens and they are activated by proteolytic cleavage.13–18 Upon activation, each caspase forms a tetramer of the two large and the two small subunits.13–18 The hierarchy and partial substrate redundance allows to a form proteolytic, signalling cascade with positive feedback properties.13–18

Figure 1. Activation of apoptosis pathways by anticancer therapy.

Figure 1

Activation of apoptosis pathways by anticancer therapy. Anticancer therapy-induced apoptotic pathways can be initiated through different entry sites, e.g., at the plasma membrane by death receptor-mediated signaling (receptor pathway) or at the mitochondria (more...)

Caspases involved in apoptosis signaling are currently categorized into initiator and effector caspases, respectively.13–18 Initiator caspases transduce various signals into protease activity and are directly linked to death inducing signaling complexes (DISCs): caspase-8 or caspase-10 via their death effector domain (DED) interact with adaptor proteins (FADD) recruited and bound to activated death receptors while caspase-9 is recruited to the apoptosome via its CARD domain.13 Effector caspases cleave various cytoplasmatic or nuclear substrates prompting the occurrence of morphologic features of apoptosis.13–18 For example, polynucleosomal DNA fragmentation is initiated by cleavage of ICAD (inhibitor of caspase-activated DNase), the inhibitor of the endonuclease CAD (caspase-activated DNase) that cleaves DNA into the characteristic oligomeric fragments.8 DNA condensation is caused by AIF, a mitochondrial protein that translocates to the nucleus upon death triggering, and by Acinus, which stands for “apoptotic chromation condensation inducer in the nucleus”.29,30 AIF may also mediate caspase-independent cleavage of DNA into larger fragments.29,30 Likewise, loss of overal cell shape is due to proteolysis of cytoskeletal proteins including fodrin, gelsolin, actin, plectrin, cytokeratin, while nuclear shrinking and budding occurs after degradation of lamin.8

Pathways of Caspase Activation

Activation of caspases can principially be triggered by two different mechanisms: according to the induced proximity model initiator caspases such as caspase-8 or 9 are activated in a multimeric complex, e.g., caspase-8 in the death inducing signaling complex (DISC) and caspase-9 at the apoptosome.8,13,31–33 Alternatively, caspases are activated by catalytic processing of the zymogens at specific cleavage sites.13 Caspase activation can be initiated through different entry sites, e.g., at the plasma membrane by death receptor-mediated signaling (receptor pathway) or at the mitochondria (mitochondrial pathway).8,13 Stimulation of death receptors of the tumor necrosis factor (TNF) receptor superfamily such as CD95 (APO-1/Fas) or TRAIL receptors results in receptor aggregation and recruitment of the adaptor molecule Fas-associated death domain (FADD) and caspase-8.31–33 Upon recruitment caspase-8 becomes activated and initiates apoptosis by direct cleavage of downstream effector caspases.31–33 The mitochondrial pathway is initiated by the release of apoptogenic factors such as cytochrome c, apoptosis inducing factor (AIF), Smac/DIABLO, Omi/HtrA2, endonuclease G, caspase-2 or caspase-9 from the mitochondrial intermembrane space.34–39 The release of cytochrome c into the cytosol triggers caspase-3 activation through formation of the cytochrome c/Apaf-1/caspase-9-containing apoptosome complex.40,41 Smac/DIABLO and Omi/HtrA2 promote caspase activation through neutralizing the inhibitory effects to IAPs, while AIF and endonuclease G cause DNA condensation.30,38,39,42,43

The receptor and the mitochondrial pathway can be interconnected at different levels.44 Following death receptor stimulation activation of caspase-8 may result in cleavage of Bid, a BH3 domain containing protein of the Bcl-2 family which assumes cytochrome c-releasing activity upon cleavage thereby initiating a mitochondrial amplification loop.43,44 In addition, mitochondria-triggered caspase-6 cleavage may feed back to the receptor pathway by cleaving caspase-8.16

Signaling Pathways in Cancer Therapy

The relative contribution of the receptor and the mitochondrial pathway to drug-induced apoptosis has been a subject of controversial discussion.1,2,4 A number of studies suggested that cancer therapy-triggered apoptosis involves the CD95 system by upregulating the expression of CD95L which then binds to its receptor and stimulates the receptor pathway in an autocrine or paracrine manner.45–64 In support of this model, upregulation of CD95 mRNA and protein was found in a variety of tumor cell lines derived from T cell leukemia cells, neuroblastoma, malignant brain tumors, hepatoma, colon, breast or small lung cell carcinoma upon treatment with cytotoxic drugs such as doxorubicin, etoposide, cisplatin, 5-FU or bleomycin.45–59 The increase in CD95L transcription and mRNA levels was found to be related to drug-induced activation of the transcription factors AP-1 and NF-κB.55,60,61 Also, CD95 expression increased upon drug treatment, in particular in cells harboring wild-type p53, as the CD95 promotor contains p53 binding sites.51,57,58 In addition, soluble antagonistic CD95 receptors, antagonistic CD95L antibodies or DN-FADD reduced drug-induced apoptosis under certain circumstances.45,49,57,59 Moreover, it was recently demonstrated in vivo that 5-fluorouracil induced apoptosis in mouse thymocytes via activation of the CD95 system, since apoptosis was blocked by neutralizing CD95L antibodies or in lpr mice lacking a functional CD95 receptor.62 Also, CD95L-independent activation of the receptor pathway through CD95 receptor oligomerization has been reported, e.g., by UV irradiation, cytotoxic drugs or suicide gene therapy using the herpes simplex thymidine kinase (HSV/TK) system.65–67

Other reports however challenged the concept that death receptor signaling is involved in drug-mediated cell death.68–74 Antagonistic antibodies against CD95L or CD95 did not protect from cancer chemotherapeutically induced death in several cell line models.72–74 Although splenocytes from lpr mice exhibit decreased sensitivity to γ-irradiation, thymocytes of these mice did not show increased proliferation upon γ-irradiation or cytotoxic drugs.68 Moreover, overexpression of FLIP, DN-FADD or the serpin CrmA that inhibits caspase-8 did not confer protection.69,72–74 In addition, targeted disruption of genes involved in death receptor signaling suggested a dispensable role of the CD95 system in drug-induced apoptosis, at least in nontransformed cells. FADD−/− and caspase-8−/− fibroblasts are resistant to death receptor stimulation, but equally sensitive to cytotoxic drugs.75,76 In contrast, caspase-9−/− embryonic stem cells and Apaf1−/− thymocytes remain sensitive to death receptor triggering, however, and are resistant to cytotoxic drugs.77,78 The discrepancies in data may be explained by the relative contribution of the death receptor versus the mitochondrial pathway depending on the cytotoxic drug, dose and kinetics or on differences between certain cell types. For CD95 signaling, 2 different cell types have been identified:79 type I cells undergo CD95-triggered apoptosis independent of mitochondria, since caspase-8 is already efficiently activated at the DISC upstream of mitochondria. In contrast, type II cells depend on the mitochondrial pathway, since only little caspase-8 is recruited and activated at the DISC.79 A similar cell type dependent signaling has also been identified in response to drug treatment.54 Although the CD95 system is involved in anticancer drug-induced apoptosis under certain circumstances, the majority of cytotoxic drugs initiate cell death by triggering the cytochrome c/Apaf-1/caspase-9 dependent pathway through the mitochondria. Collectively, these data point to a crucial role of the mitochondrial pathway in drug-induced apoptosis, while the CD95 system may amplify and accelerate drug-induced apoptosis under certain conditions. Importantly, this amplification of the chemoresponse may be clinically meaningful, since it may critically affect the time required for execution of the death program.80

Regulation of Caspase Activation

Given the important role of caspases as effector molecules in various forms of cell death including drug-induced apoptosis, the ability of anticancer agents to trigger caspase activation appears to be a critical determinant of sensitivity or resistance to cytotoxic therapies. As a consequence, inhibition of caspase activation may be an important factor in chemoresistance. Given the central role of caspases for cell death execution one might expect a high frequency of caspase mutations in tumors. Interestingly however, screening for mutations in initiator or executioner caspases in a variety of human tumors has not reveiled a high frequency of genomic aberrations in caspase genes.81,82 Instead, caspase expression and function appears to be epigenetically downregulated in tumors by mechanisms described below, suggesting that restoration of a functional caspase system may be important to overcome resistance in tumors.82,83

Caspase Expression

First, expression levels of individual caspases may have an impact on their overall activity, since activation of caspases may simply be impaired by deficient expression levels of caspases.84–86 For example, MCF-7 breast carcinoma cells completely lack caspase-3 expression due to a frameshift mutation within exon 3 of the caspase-3 gene.85 These cells can be sensitized by transfection of pro-caspase-3 towards treatment with cytotoxic drugs.86 Next, caspase expression may be impaired by epigenetic alterations such as promotor hypermethylation. To this end, caspase-8 expression was found to be frequently inactivated by hypermethylation of regulatory sequences of the caspase-8 gene in a number of different tumor cells derived from neuroblastoma, malignant brain tumors, Ewing tumor and small lung cell carcinoma both in vitro and also in vivo in primary tumor samples.82,83 Importantly, restoration of caspase-8 expression by gene transfer or by demethylation treatment sensitized resistant tumor cells for death-receptor or drug-induced apoptosis.82,83 Alternative splicing has been identified as another level of transcriptional regulation of caspase expression. The genes encoding procaspase-2 or procaspase-9 can generate short isoforms that prevent apoptosis in a dominant-negative fashion.87,88 Conversely, enhanced transcription of caspase genes in response to cytotoxic treatment may increase expression levels. Thus, treatment with IFN-γ resulted in enhanced expression of caspase proteins mediated by direct activation of STAT1, a downstream transcription factor involved in IFN-γ signaling.89 Moreover, transcriptional upregulation of caspase-3 or 8 was reported upon drug treatment independent of STAT1.90,91 In addition, subcellular compartmentalization of caspases may regulate their activation. Interestingly, in addition to their cytoplasmic localization, several caspases including caspase-2, -3 and 9 are found inside mitochondria.92,93 Death signals targeting mitochondria trigger the translocation of caspases from the mitochondrial intermembrane space into the cytosol and also promote their nuclear translocation.92–94

Bcl-2 Proteins

Bcl-2 family proteins play a pivotal role in the regulation of the mitochondrial death pathway.100–104 The family comprises antiapoptotic members, e.g., Bcl-2, Bcl-XL, Mcl-1, proapoptotic molecules such as Bax, Bak, Bad, as well as BH3 domain only molecules which link the death receptor pathway to the mitochondrial pathway (Bid, Bim, PUMA, Noxa).100,101 Upon apoptosis induction proapoptotic Bcl-2 proteins with multidomains such as Bax or Bak translocate from the cytoplasm to the outer mitochondrial membrane, where they oligomerize to form a pore-like structure thereby promoting cytochrome c release.102 The translocation to mitochondria can be triggered by so called “BH3-only” Bcl-2 family proteins.100 BH3-only proteins include Bid, which is activated by caspase-8-mediated cleavage, Bim, a microtubule-associated protein, or Noxa and PUMA, two p53-induced proteins.100,101 Bcl-2 or Bcl-XL exert their antiapoptotic function, at least in part, by sequestering BH3-only proteins in stable mitochondrial complexes thereby preventing activation and translocation of Bax or Bak to mitochondria.102 In addition, Bcl-2 and Bcl-XL block apoptosis by preventing cytochrome c release through a direct effect on mitochondrial channels such as the voltage-dependent anion channel (VDAC) or the permeability transition pore complex (PTPC).35,36 Several clinical correlative studies indicate that high expression of antiapoptotic Bcl-2 proteins confers a clinically relevant chemoresistant phenotype in various malignancies, including AML, ALL, CLL, multiple myeloma, prostate carcinoma, malignant brain tumors and neuroblastoma.22,105–107 Likewise, reduced Bax level have been associated with poor responses to chemotherapy and shorter overall survival in breast or colorectal carcinoma.108 Conversely, enhanced Bax expression correlated in several cell types with sensitivity to chemotherapy in vivo.109

Inhibitors of Apoptosis Proteins (IAPs)

The family of endogenous caspase inhibitors “inhibitor of apoptosis proteins” (IAPs) are highly conserved throughout evolution and comprise the human analogues XIAP, cIAP1, cIAP2, survivin, livin and MLIAP.110–112 Their common structural features consist of 1–3 baculovirus inhibitor repeat (BIR) domains, that mediate binding to caspases, and a RING domain, that acts as ubiquitin ligase thereby promoting ubiquitination and proteasomal degradation of the bound caspases.110 IAPs have been reported to directly inhibit active caspase-3 and 7 and to block caspase-9 activation.110 In addition to regulation of apoptosis, IAP members such as survivin have been found to be involved in the regulation of mitosis.112,113 The activity of IAPs are controlled at various levels, e.g., by the transcription factor NF-κB that has been reported to stimulate expression of cIAP1, cIAP and XIAP.110 IAPs are negatively regulated by caspase-mediated cleavage.110 In addition, Smac/DIABLO and Omi, two proteins released from mitochondria upon apoptosis induction, neutralize IAPs through their binding, thereby displacing them from caspases.111 Likewise, XAF1 has been found to displace IAPs from bound caspases in the nucleus.111 Inhibition of apoptosis by IAPs in response to cytotoxic therapy has been suggested by several experimental studies.114–119 XIAP, cIAP1 or cIAP2 suppressed apoptosis in vitro following treatment with cisplatin, cytarabine, TRAIL, staurosporine or after γ-irradiation.118,119 Also, increased IAPs expression correlated with poor treatment response in myeloid leukemia cells and elevated survivin expression predicted adverse prognosis in several tumors, e.g., neuroblastoma, AML, colon, lung and esophagus carcinoma.114–117

Death Receptors

Activation of caspases may also be controlled by upstream regulation at the level of death receptors. First, death receptor expression may vary between different cell types and can be downregulated in tumor cells, thus contributing to the escape from negative growth control.31–33 Signaling by death receptors can be negatively regulated by proteins that associate with their cytoplasmatic domains, e.g., SODD, or by proteins such as FLIP that prevent the interaction between the adaptor molecule FADD and pro-caspase-8.95–97 High FLIP expression which has been found in many tumor cells has been correlated with resistance to CD95- and TRAIL-induced apoptosis.96 In addition, FLIP expression was associated with tumor escape from T-cell immunosurveillance and enhanced tumor progression in experimental studies in vivo, pointing to a role of FLIP as a tumorprogression factor.97 The impact of FLIP on apoptosis sensitivity towards cytotoxic drugs may vary between cell types, since overexpression of FLIP did not confer protection against cytotoxic drugs in T cell leukemia cells, while FLIP antisense oligonucleotides sensitized osteosarcoma cells for cisplatin.69,98 Elevated FLIP expression has been found in clinical samples from Burkitt lymphoma, pancreatic carcinoma, melanoma or neuroblastoma and in tumor cells that developed resistance upon chemotherapy suggesting that FLIP may play a role in chemoresistance of tumors.97–99

Caspase-Independent Cell Death

Although a large body of data point to an essential role of caspase-mediated tumor cell death upon cytotoxic therapy, this concept has also been challenged.1–7,23 Thus, a clear, consistent link between the cells' ability to undergo apoptosis and their susceptibility to anticancer therapy could not be observed.23 In addition, the p53 status did not always correlate with the ability of a tumor cell to respond to treatment.120 Cells harboring wild-type p53 may fail to respond and those lacking functional p53 may even respond better.120 Moreover, nonapoptotic modes of cell death, e.g., necrosis or some forms of cell death that cannot be clearly classified, have also been taken into consideration as response to cytotoxic therapy.23–28 Also, delayed regression of tumors upon e.g., irradiation has been taken as evidence against a predominant apoptotic mode of cell death, since apoptosis appears to be induced fairly rapidly in vitro and in vivo upon appropriate stimulation.23 Although signaling pathways and molecules involved in these alternative forms of cell death have not yet exactly been defined, non-caspase proteases such as calpains or cathepsins, Bax or Bax-like molecules and AIF or endonuclease G may be involved.8,23–28 The relative contribution of these different modes of cell death for chemoresponses in vitro and in vivo remains to be defined.

Role of Caspases for Treatment Response in Vivo

What is the clinical impact of caspase expression and/or activity on individual patient's response to anticancer therapy in vivo? Unfortunately, this question is far from being answered yet. Aside from Bcl-2 family proteins, most molecules involved in the regulation of apoptosis including caspases have not extensively been studied in clinical specimens.22,43 Although the potential relation between expression levels of procaspases in clinical samples and patients' response to chemotherapy has been addressed in several studies, the conclusive answer is still missing.121–125 Some investigators postulated a correlation between procaspase-3 expression and clinical response, e.g., in leukemia, Hodgkin's disease or NSCLC.121–124 However, this conclusion was not always based on a direct correlation between procaspase-3 expression levels and individual treatment responses and further studies did not confirm these findings. In bone marrow samples with predominance of leukemic blasts, a wide variation of caspase-2, -3, -7, -8, and -9 was found among different specimens, however, their level did not correlate with prognostic factors or response to induction chemotherapy.125 Loss of spontaneous caspase-3 cleavage was reported in ALL samples at relapse compared to those at initial diagnosis.107 As discussed before, while mutations in caspase genes have only infrequently been found in tumors, inactivation of caspase expression by epigenetic alterations such as promotor hypermethylation appears to be a primary mechanism of disabling of the caspase cascades in tumors.81–83 How inactivation of caspase expression by DNA hypermethylation will correlate with clinical outcome remains a subject of future studies. The prospective study of clinical samples is further complexed by the necessity of multiparameter analysis. The expression level and activity of caspases is affected in vivo by positive and negative apoptosis regulators, such as Bcl-2 family proteins or IAPs.100,110 Thus, an assessment of the impact of caspase expression and/or function on chemoresponse in vivo will require multiparamter analysis, e.g., by expression profiling.

Conclusion

Numerous studies over the last years have indicated that anticancer therapies primarily act by activating the apoptosis response pathway in tumor cells.1–7 However, several points still remain to be addressed: First, most of the apoptosis signaling components have not been studied in clinical samples.2,4,6,7,22 Second, many experimental studies indicate that alterations in components of the apoptotic machinery have an impact on sensitivity of tumor cells towards cytotoxic therapy, this premise remains to be tested in clinical settings.1–5,22 Moreover, the biology that determines the individual responses of different tumors to cytotoxic therapies warrants further investigations to provide the basis for more specific therapeutic interventions. Finally, the concept that apoptosis represents the major mechanism by which tumor cells are eliminated by cytotoxic therapies may not universally apply and caspase-independent modes of cell death have also also to be considered.23–28

Nonetheless, studies on the regulation of apoptosis signaling pathways triggered by anticancer therapies have provided substantial insights into the molecular mechanisms regulating the response of tumor cells towards current therapies. Future studies on the role of apoptosis signaling molecules in individual tumors both in vitro and in vivo in tumor cells of patients under chemotherapy, e.g., by DNA microarrays or proteomic studies, may provide the basis for “tailored” tumor therapy and may identify new targets for therapeutic interventions.

References

1.
Debatin K M. The role of the CD95 system in chemotherapy In: Broxterman HJA, ed.Drug Resistance Updates Edinburgh: Churchill Livingstone, 199985–90. [PubMed: 11504475]
2.
Herr I, Debatin K M. Cellular stress response and apoptosis in cancer therapy. Blood. 2001;98:2603–2614. [PubMed: 11675328]
3.
Debatin K M. Anticancer drugs, programmed cell death and the immune system: defining new roles in an old play. J Natl Cancer Inst. 1997;89:750–753. [PubMed: 9182966]
4.
Kaufmann S H, Earnshaw W C. Induction of apoptosis by cancer chemotherapy. Exp Cell Res. 2000;256:42–49. [PubMed: 10739650]
5.
Solary E, Droin N, Bettaieb A. et al. Positive and negative regulation of apoptotic pathways by cytotoxic agents in hematological malignancies. Leukemia. 2000;14:1833–1849. [PubMed: 11021759]
6.
Lowe S W, Lin A W. Apoptosis in cancer. Carcinogenesis. 2000;21:485–495. [PubMed: 10688869]
7.
Kaufmann S H, Gores G J. Apoptosis in cancer: Cause and cure. Bioessays. 2000;22:1007–1017. [PubMed: 11056477]
8.
Hengartner M O. The biochemistry of apoptosis. Nature. 2000;407:770–777. [PubMed: 11048727]
9.
Rich T, Allen R L, Wyllie A H. Defying death after DNA damage. Nature. 2000;407:777–783. [PubMed: 11048728]
10.
Leppa S, Bohmann D. Diverse functions of JNK signaling and c-Jun in stress response and apoptosis. Oncogene. 1999;18:6158–6162. [PubMed: 10557107]
11.
Davis R J. Signal transduction by the JNK group of MAP kinases. Cell. 2000;103:239–252. [PubMed: 11057897]
12.
Mayo M W, Baldwin A S. The transcription factor NF-κB: Control of oncogenesis and cancer therapy resistance. Biochim Biophys Acta. 2000;1470:M55–62. [PubMed: 10722927]
13.
Thornberry N, Lazebnik Y. Caspases: Enemies within. Science. 1998;281:1312–1316. [PubMed: 9721091]
14.
Los M, Wesselborg S, Schulze-Osthoff K. The role of caspases in development, immunity, and apoptotic signal transduction: Lessons from knockout mice. Immunity. 1999;10:629–639. [PubMed: 10403638]
15.
Degen W G J, Pruijn G J M, Raats J M H. et al. Caspase-dependent cleavage of nucleic acids. Cell Death Differ. 2000;7:616–627. [PubMed: 10889506]
16.
Slee E A, Adrain C, Martin S J. Serial killers: Ordering caspase activation events in apoptosis. Cell Death Differ. 1999;6:1067–1074. [PubMed: 10578175]
17.
Utz P J, Anderson P. Life and death decisions: Regulation of apoptosis by proteolysis of signaling molecules. Cell Death Differ. 2000;7:589–602. [PubMed: 10889504]
18.
Earnshaw W C, Martins L M, Kaufmann S H. Mammalian caspases: Structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem. 1999;68:383–424. [PubMed: 10872455]
19.
Kaufmann S H. Induction of endonucleolytic DNA cleavage in human acute myelogenous leukemia cells by etoposide, camptothecin, and other cytotoxic anticancer drugs: A cautionary note. Cancer Res. 1989;49:5870–5878. [PubMed: 2790800]
20.
Los M, Herr I, Friesen C. et al. Cross-resistance of CD95- and drug-induced apoptosis as a consequence of deficient activation of caspases (ICE/Ced-3 proteases). Blood. 1997;90:3118–3129. [PubMed: 9376593]
21.
Goyal L. Cell death inhibition: Keeping caspases in check. Cell. 2001;104:805–808. [PubMed: 11290317]
22.
Reed J C. Dysregulation of apoptosis in cancer. J Clin Oncol. 1999;17:2941–2953. [PubMed: 10561374]
23.
Finkel E. Does cancer therapy trigger cell suicide? Science. 1999;286:2256–2258. [PubMed: 10636781]
24.
Sperandio S, de Belle I, Bredesen D E. An alternative, nonapoptotic form of programmed cell death. Proc Natl Acad Sci USA. 2000;97:14376–14381. [PMC free article: PMC18926] [PubMed: 11121041]
25.
Wyllie A H, Golstein P. More than one way to go. Proc Natl Acad Sci USA. 2000;98:11–13. [PMC free article: PMC33349] [PubMed: 11136242]
26.
Borner C, Monney L. Apoptosis without caspases: An inefficient molecular guillotine? Cell Death Differ. 1999;6:497–507. [PubMed: 10381652]
27.
Johnson D E. Noncaspase proteases in apoptosis. Leukemia. 2000;14:1695–1703. [PubMed: 10995018]
28.
Leist M, Jaattela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol. 2001;2:589–598. [PubMed: 11483992]
29.
Ferri K F, Kroemer G. Control of apoptotic DNA degradation. Nat Cell Biol. 2000;2:E63–64. [PubMed: 10783253]
30.
Daugas E, Nochy D, Ravagnan L. et al. Apoptosis-inducing factor (AIF): A ubiquitous mitochondrial oxidoreductase involved in apoptosis. FEBS Lett. 2000;476:118–123. [PubMed: 10913597]
31.
Schulze-Osthoff K, Ferrari D, Los M. et al. Apoptosis signaling by death receptors. Eur J Biochem. 1998;254:439–459. [PubMed: 9688254]
32.
Krammer P H. CD95's deadly mission in the immune system. Nature. 2000;407:789–795. [PubMed: 11048730]
33.
Walczak H, Krammer P H. The CD95 (APO-1/Fas) and the TRAIL (APO-2L) apoptosis systems. Exp Cell Res. 2000;256:58–66. [PubMed: 10739652]
34.
Costantini P, Jacotot E, Decaudin D. et al. Mitochondrion as a novel target of anticancer chemotherapy. J Natl Cancer Inst. 2000;92:1042–1053. [PubMed: 10880547]
35.
Kroemer G, Reed J C. Mitochondrial control of cell death. Nat Med. 2000;6:513–519. [PubMed: 10802706]
36.
Martinou J C, Green D R. Breaking the mitochondrial barrier. Nat Rev Mol Cell Biol. 2001;2:63–67. [PubMed: 11413467]
37.
Suzuki Y, Imai Y, Nakayama H. et al. A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell. 2001;8:613–621. [PubMed: 11583623]
38.
Du C, Fang M, Li Y. et al. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell. 2000;102:33–42. [PubMed: 10929711]
39.
Verhagen A M, Ekert P G, Pakusch M. et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell. 2000;102:43–53. [PubMed: 10929712]
40.
Bratton S B, MacFarlane M, Cain K. et al. Protein complexes activate distinct caspase cascades in death receptor and stress-induced apoptosis. Exp Cell Res. 2000;256:27–33. [PubMed: 10739648]
41.
Adrain C, Martin S J. The mitochondrial apoptosome: A killer unleashed by the cytochrome seas. Trends Biochem Sci. 2001;26:390–397. [PubMed: 11406413]
42.
Li L Y, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature. 2001;412:95–99. [PubMed: 11452314]
43.
Schimmer A D, Hedley D W, Penn L Z. et al. Receptor- and mitochondrial-mediated apoptosis in acute leukemia: a translational view. Blood. 2001;98:3541–3553. [PubMed: 11739155]
44.
Roy S, Nicholson D W. Cross-talk in cell death signaling. J Exp Med. 2000;192:21–26. [PubMed: 11034597]
45.
Friesen C, Herr I, Krammer P H. et al. Involvement of the CD95 (APO-1/FAS) receptor/ligand system in drug-induced apoptosis in leukemia cells. Nat Med. 1996;2:574–577. [PubMed: 8616718]
46.
Friesen C, Fulda S, Debatin K M. Drugs and the CD95 pathway. Leukemia. 1999;13:1854–1858. [PubMed: 10557062]
47.
Friesen C, Fulda S, Debatin K M. Deficient activation of the CD95 (APO-1/Fas) system in drug-resistant cells. Leukemia. 1997;11:1833–1841. [PubMed: 9369415]
48.
Friesen C, Fulda S, Debatin K M. Activation of the CD95 system by doxorubicin is modulated by the redox state in chemosensitive- and drug-resistant tumor cells. Cell Death Differ. 1999;6:471–480. [PubMed: 10381639]
49.
Fulda S, Sieverts H, Friesen C. et al. The CD95 (APO-1/Fas) system mediates drug-induced apoptosis in neuroblastoma cells. Cancer Res. 1997;57:3823–3829. [PubMed: 9288794]
50.
Fulda S, Los M, Friesen C. et al. Chemosensitivity of solid tumor cells is associated with activation of the CD95 system. Int J Cancer. 1998;76:105–114. [PubMed: 9533769]
51.
Fulda S, Scaffidi C, Pietsch T. et al. Activation of the CD95 (APO-1/Fas) pathway in drug- and g-irradiation-induced apoptosis of brain tumor cells. Cell Death Differ. 1998;5:884–893. [PubMed: 10203687]
52.
Fulda S, Susin S A, Kroemer G. et al. Molecular ordering of apoptosis induced by anticancer drugs in neuroblastoma cells. Cancer Res. 1998;58:4453–4460. [PubMed: 9766678]
53.
Fulda S, Strauss G, Meyer E. et al. Functional CD95 ligand and CD95 DISC in activation-induced cell death and doxorubicin-induced apoptosis in leukemic T cells. Blood. 2000;95:301–308. [PubMed: 10607716]
54.
Fulda S, Meyer E, Susin S A. et al. Cell type specific activation of death receptor and mitochondrial pathways in drug-induced apoptosis. Oncogene. 2001;20:1063–1075. [PubMed: 11314043]
55.
Herr I, Wilhelm D, Bohler T. et al. Activation of CD95 (APO-1/Fas) signaling by ceramide mediates cancer therapy-induced apoptosis. EMBO J. 1997;16:6200–6208. [PMC free article: PMC1326304] [PubMed: 9321399]
56.
Houghton J A, Harwood F G, Tillman D M. Thymineless death in colon carcinoma cells is mediated via fas signaling. Proc Natl Acad Sci USA. 1997;94:8144–8149. [PMC free article: PMC21571] [PubMed: 9223329]
57.
Muller M, Wilder S, Bannasch D. et al. p53 activates the CD95 (APO-1/Fas) gene in response to DNA damage by anticancer drugs. J Exp Med. 1998;188:2033–2045. [PMC free article: PMC2212386] [PubMed: 9841917]
58.
Muller M, Strand S, Hug H. et al. Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO-1/Fas) receptor/ligand system and involves activation of wild-type p53. J Clin Invest. 1997;99:403–413. [PMC free article: PMC507813] [PubMed: 9022073]
59.
Reap E A, Roof K, Maynor K. et al. Radiation and stress-induced apoptosis: A role for Fas/Fas ligand interactions. Proc Natl Acad Sci USA. 1997;94:5750–5755. [PMC free article: PMC20851] [PubMed: 9159145]
60.
Kasibhatla S, Brunner T, Genestier L. et al. DNA-damaging agents induce expression of Fas ligand and subsequent apoptosis in T lymphocytes via the activation of NF-kappa B and AP-1. Mol Cell. 1998;1:543–551. [PubMed: 9660938]
61.
Eichhorst S T, Muller M, Li-Weber M. et al. A novel AP-1 element in the CD95 ligand promoter is required for induction of apoptosis in hepatocellular carcinoma cells upon treatment with anticancer drugs. Mol Cell Biol. 2000;20:7826–7837. [PMC free article: PMC86378] [PubMed: 11003676]
62.
Eichhorst S T, Muerkoster S, Weigand M A. et al. The chemotherapeutic drug 5-fluorouracil induces apoptosis in mouse thymocytes in vivo via activation of the CD95(APO-1/Fas) system. Cancer Res. 2001;61:243–248. [PubMed: 11196169]
63.
Leverkus M, Yaar M, Gilchrest B A. Fas/Fas ligand interaction contributes to UV-induced apoptosis in human keratinocytes. Exp Cell Res. 1997;232:255–262. [PubMed: 9168800]
64.
Mo Y Y, Beck W T. DNA damage signals induction of fas ligand in tumor cells. Mol Pharmacol. 1999;55:216–222. [PubMed: 9927611]
65.
Beltinger C, Fulda S, Kammertoens T. et al. Herpes simplex virus thymidine kinase/ganciclovir-induced apoptosis involves ligand-independent death receptor aggregation and activation of caspases. Proc Natl Acad Sci USA. 1999;96:8699–8704. [PMC free article: PMC17579] [PubMed: 10411938]
66.
Rehemtulla A, Hamilton C A, Chinnaiyan A M. et al. Ultraviolet radiation-induced apoptosis is mediated by activation of CD-95 (Fas/APO-1). J Biol Chem. 1997;272:25783–25786. [PubMed: 9325306]
67.
Micheau O, Solary E, Hammann A. et al. Fas ligand-independent, FADD-mediated activation of the Fas death pathway by anticancer drugs. J Biol Chem. 1999;274:7987–7992. [PubMed: 10075697]
68.
Reap E A, Roof K, Maynor K. et al. Markedly diminished radiation-induced lymphocyte apoptosis in lpr mice suggests a role for Fas in eliminating damaged cells. Ann NY Acad Sci. 1997;815:116–118. [PubMed: 9186646]
69.
Kataoka T, Schroter M, Hahne M. et al. FLIP prevents apoptosis induced by death receptors but not by perforin/granzyme B, chemotherapeutic drugs, and gamma irradiation. J Immunol. 1998;161:3936–3942. [PubMed: 9780161]
70.
Landowski T H, Shain K H, Oshiro M M. et al. Myeloma cells selected for resistance to CD95-mediated apoptosis are not cross-resistant to cytotoxic drugs: evidence for independent mechanisms of caspase activation. Blood. 1999;94:265–274. [PubMed: 10381522]
71.
Eischen C M, Kottke T J, Martins L M. et al. Comparison of apoptosis in wild-type and Fas-resistant cells: chemotherapy-induced apoptosis is not dependent on Fas/Fas ligand interactions. Blood. 1997;90:935–943. [PubMed: 9242521]
72.
Villunger A, Egle A, Kos M. et al. Drug-induced apoptosis is associated with enhanced Fas (Apo-1/CD95) ligand expression but occurs independently of Fas (Apo-1/CD95) signaling in human Tacute lymphatic leukemia cells. Cancer Res. 1997;57:3331–3334. [PubMed: 9269989]
73.
Wesselborg S, Engels I H, Rossmann E. et al. Anticancer drugs induce caspase-8/FLICE activation and apoptosis in the absence of CD95 receptor/ligand interaction. Blood. 1999;93:3053–3063. [PubMed: 10216102]
74.
Glaser T, Wagenknecht B, Groscurth P. et al. Death ligand/receptor-independent caspase activation mediates drug-induced cytotoxic cell death in human malignant glioma cells. Oncogene. 1999;18:5044–5053. [PubMed: 10490841]
75.
Yeh W C, Pompa J L, McCurrach M E. et al. FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis. Science. 1998;279:1954–1958. [PubMed: 9506948]
76.
Varfolomeev E E, Schuchmann M, Luria V. et al. Targeted disruption of the mouse caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally. Immunity. 1998;9:267–276. [PubMed: 9729047]
77.
Hakem R, Hakem A, Duncan G S. et al. Differential requirement for caspase 9 in apoptotic pathways in vivo. Cell. 1998;94:339–352. [PubMed: 9708736]
78.
Yoshida H, Kong Y Y, Yoshida R. et al. Apaf1 is required for mitochondrial pathways of apoptosis and brain development. Cell. 1998;94:739–750. [PubMed: 9753321]
79.
Scaffidi C, Fulda S, Srinivasan A. et al. Two CD95 (APO-1/Fas) signaling pathways. EMBO J. 1998;17:1675–1687. [PMC free article: PMC1170515] [PubMed: 9501089]
80.
Tang D, Lahti J M, Kidd V J. Caspase-8 activation and bid cleavage contribute to MCF7 cellular execution in a caspase-3-dependent manner during staurosporine-mediated apoptosis. J Biol Chem. 2000;275:9303–9307. [PubMed: 10734071]
81.
Mandruzzato S, Brasseur F, Andry G. et al. A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. J Exp Med. 1997;186:785–793. [PMC free article: PMC2199018] [PubMed: 9271594]
82.
Teitz T, Wei T, Valentine M B. et al. Caspase 8 is deleted or silenced preferentially in childhood neuroblastomas with amplification of MYCN. Nat Med. 2000;6:529–535. [PubMed: 10802708]
83.
Fulda S, Kufer M U, Meyer E. et al. Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer. Oncogene. 2001;20:5865–5877. [PubMed: 11593392]
84.
Joseph B, Ekedahl J, Sirzen F. et al. Differences in expression of pro-caspases in small cell and non-small cell lung carcinoma. Biochem Biophys Res Commun. 1999;262:381–387. [PubMed: 10462484]
85.
Janicke R U, Sprengart M L, Wati M R. et al. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. J Biol Chem. 1998;273:9357–9360. [PubMed: 9545256]
86.
Yang X H, Sladek T L, Liu X. et al. Reconstitution of caspase 3 sensitizes MCF-7 breast cancer cells to doxorubicin- and etoposide-induced apoptosis. Cancer Res. 2001;61:348–354. [PubMed: 11196185]
87.
Wang L, Miura M, Bergeron L. et al. Ich-1, an Ice/ced-3-related gene, encodes both positive and negative regulators of programmed cell death. Cell. 1994;78:739–750. [PubMed: 8087842]
88.
Srinivasula S M, Ahmad M, Guo Y. et al. Identification of an endogenous dominant-negative short isoform of caspase-9 that can regulate apoptosis. Cancer Res. 1999;59:999–1002. [PubMed: 10070954]
89.
Fulda S, Debatin K M. IFN-γ sensitizes for apoptosis by upregulating caspase-8 expression through the Stat1 pathway. Oncogene. 2002;21:2295–308. [PubMed: 11948413]
90.
Droin N, Dubrez L, Eymin B. et al. Upregulation of CASP genes in human tumor cells undergoing etoposide-induced apoptosis. Oncogene. 1998;16:2885–2894. [PubMed: 9671409]
91.
Micheau O, Hammann A, Solary E. et al. STAT-1-independent upregulation of FADD and procaspase-3 and -8 in cancer cells treated with cytotoxic drugs. Biochem Biophys Res Commun. 1999;256:603–607. [PubMed: 10080945]
92.
Samali A, Cai J, Zhivotovsky B. et al. Presence of a pre-apoptotic complex of pro-caspase-3, Hsp60 and Hsp10 in the mitochondrial fraction of Jurkat cells. EMBO J. 1999;18:2040–2048. [PMC free article: PMC1171288] [PubMed: 10205158]
93.
Susin S A, Lorenzo H K, Zamzami N. et al. Mitochondrial release of caspase-2 and -9 during the apoptotic process. J Exp Med. 1999;189:381–394. [PMC free article: PMC2192979] [PubMed: 9892620]
94.
Budihardjo I, Oliver H, Lutter M. et al. Biochemical pathways of caspase activation during apoptosis. Annu Rev Cell Dev Biol. 1999;15:269–290. [PubMed: 10611963]
95.
Jiang Y, Woronicz J D, Liu W. et al. Prevention of constitutive TNF receptor 1 signaling by silencer of death domains. Science. 1999;283:543–546. [PubMed: 9915703]
96.
Tschopp J, Irmler M, Thome M. Inhibition of fas death signals by FLIPs. Curr Opin Immunol. 1998;10:552–558. [PubMed: 9794838]
97.
French L E, Tschopp J. Inhibition of death receptor signaling by FLICE-inhibitory protein as a mechanism for immune escape of tumors. J Exp Med. 1999;190:891–894. [PMC free article: PMC2195651] [PubMed: 10510078]
98.
Kinoshita H, Yoshikawa H, Shiiki K. et al. Cisplatin (CDDP) sensitizes human osteosarcoma cell to Fas/CD95-mediated apoptosis by down-regulating FLIP-L expression. Int J Cancer. 2000;88:986–991. [PubMed: 11093825]
99.
Elnemr A, Ohta T, Yachie A. et al. Human pancreatic cancer cells disable function of Fas receptors at several levels in Fas signal transduction pathway. Int J Oncol. 2001;18:311–316. [PubMed: 11172597]
100.
Antonsson B, Martinou J C. The Bcl-2 protein family. Exp Cell Res. 2000;256:50–57. [PubMed: 10739651]
101.
Oda E, Ohki R, Murasawa H. et al. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science. 2000;288:1053–1058. [PubMed: 10807576]
102.
Cheng E H, Wei M C, Weiler S. et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol Cell. 2001;8:705–711. [PubMed: 11583631]
103.
Zhang L, Yu J, Park B H. et al. Role of BAX in the apoptotic response to anticancer agents. Science. 2000;290:989–992. [PubMed: 11062132]
104.
Minn A J, Rudin C M, Boise L H. et al. Expression of bcl-xL can confer a multidrug resistance phenotype. Blood. 1995;86:1903–1910. [PubMed: 7655019]
105.
Campos L, Rouault J P, Sabido O. et al. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood. 1993;81:3091–3096. [PubMed: 7684624]
106.
Bargou R C, Daniel P T, Mapara M Y. et al. Expression of the bcl-2 gene family in normal and malignant breast tissue: low bax-alpha expression in tumor cells correlates with resistance towards apoptosis. Int J Cancer. 1995;60:854–859. [PubMed: 7896458]
107.
Prokop A, Wieder T, Sturm I. et al. Relapse in childhood acute lymphoblastic leukemia is associated with a decrease of the Bax/Bcl-2 ratio and loss of spontaneous caspase-3 processing in vivo. Leukemia. 2000;14:1606–1613. [PubMed: 10995007]
108.
Sturm I, Kohne C H, Wolff G. et al. Analysis of the p53/BAX pathway in colorectal cancer: low BAX is a negative prognostic factor in patients with resected liver metastases. J Clin Oncol. 1999;17:1364–1374. [PubMed: 10334520]
109.
Sturm I, Petrowsky H, Volz R. et al. Analysis of p53/BAX/p16(ink4a/CDKN2) in esophageal squamous cell carcinoma: high BAX and p16(ink4a/CDKN2) identifies patients with good prognosis. J Clin Oncol. 2001;19:2272–2281. [PubMed: 11304781]
110.
Deveraux Q L, Reed J C. IAP family proteins-suppressors of apoptosis. Genes Dev. 1999;13:239–252. [PubMed: 9990849]
111.
Holcik M, Korneluk R G. XIAP, the guardian angel. Nat Rev Mol Cell Biol. 2001;2:550–556. [PubMed: 11433370]
112.
Reed J C, Bischoff J R. BIRinging chromosomes through cell division--And survivin' the experience. Cell. 2000;102:545–548. [PubMed: 11007472]
113.
Velculescu V E, Madden S L, Zhang L. et al. Analysis of human transcriptomes. Nat Genet. 1999;23:387–388. [PubMed: 10581018]
114.
Tamm I, Kornblau S M, Segall H. et al. Expression and prognostic significance of IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res. 2000;6:1796–1803. [PubMed: 10815900]
115.
Adida C, Recher C, Raffoux E. et al. Expression and prognostic significance of survivin in de novo acute myeloid leukaemia. Br J Haematol. 2000;111:196–203. [PubMed: 11091201]
116.
Adida C, Berrebi D, Peuchmaur M. et al. Anti-apoptosis gene, survivin, and prognosis of neuroblastoma. Lancet. 1998;351:882–883. [PubMed: 9525374]
117.
Li J, Feng Q, Kim J M. et al. Human ovarian cancer and cisplatin resistance: possible role of inhibitor of apoptosis proteins. Endocrinology. 2001;142:370–380. [PubMed: 11145600]
118.
Datta R, Oki E, Endo K. et al. XIAP regulates DNA damage-induced apoptosis downstream of caspase-9 cleavage. J Biol Chem. 2000;275:31733–31738. [PubMed: 10930419]
119.
Suliman A, Lam A, Datta R. et al. Intracellular mechanisms of TRAIL: Apoptosis through mitochondrial-dependent and -independent pathways. Oncogene. 2000;20:2122–2133. [PubMed: 11360196]
120.
Brown J M, Wouters B G. Apoptosis, p53, and tumor cell sensitivity to anticancer agents. Cancer Res. 1999;59:1391–1399. [PubMed: 10197600]
121.
Estrov Z, Thall P F, Talpaz M. et al. Caspase 2 and caspase 3 protein levels as predictors of survival in acute myelogenous leukemia. Blood. 1998;92:3090–3097. [PubMed: 9787143]
122.
Faderl S, Estrov Z. The clinical significance of caspase regulation in acute leukemia. Leuk Lymphoma. 2001;40:471–481. [PubMed: 11426520]
123.
Chhanabhai M, Krajewski S, Krajewska M. et al. Immunohistochemical analysis of interleukin-1beta-converting enzyme/Ced-3 family protease, CPP32/Yama/caspase-3, in Hodgkin's disease. Blood. 1997;90:2451–2455. [PubMed: 9310497]
124.
Koomagi R, Volm M. Relationship between the expression of caspase-3 and the clinical outcome of patients with non-small cell lung cancer. Anticancer Res. 2000;20:493–496. [PubMed: 10769711]
125.
Svingen P A, Karp J E, Krajewski S. et al. Evaluation of Apaf-1 and procaspases-2, -3, -7, -8, and 9 as potential prognostic markers in acute leukemia. Blood. 2000;96:3922–3931. [PubMed: 11090079]
Copyright © 2000-2013, Landes Bioscience.
Bookshelf ID: NBK6027

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...