You are on page 1of 13

Clinical Biochemistry 37 (2004) 605 617

Review

Apoptotic markers in cancer


S. Holdenrieder and P. Stieber *
Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, D-81377 Munich, Germany Accepted 4 May 2004

Abstract In cancer, apoptotic processes occur both spontaneously and induced by antitumor therapies. Qualitative and quantitative changes in cancer cell death along with proliferative alterations are essential determinants in the pathogenesis and progression of malignant disease and its responsiveness to therapy. Besides detecting apoptosis by invasive means in tumor tissue, apoptotic products can be quantified in the circulation. Although circulating apoptotic products usually lack organ and tumor specificity, they contribute in the assessment of disease extent or aggressiveness. The ease of drawing blood facilitates the serial measurement of circulating apoptotic markers to monitor antitumor treatment and predict early response to therapy. This review describes the features of apoptotic and necrotic cell death along with the role the balance between the rates of cell death and cell proliferation plays in the progression of malignancy. The intracellular pathways mediating apoptosis are next summarized. The focus then shifts to the apoptotic markers found in the circulation and their diagnostic, prognostic, predictive, and management utility in cancer. D 2004 The Canadian Society of Clinical Chemists. All rights reserved.
Keywords: Cell death; Apoptosis; Cancer; Tumor; Diagnosis; Prognosis; Therapy; Monitoring; Prediction; sFas; CD95; Cytokeratins; CYFRA 21-1; TPA; TPS; DNA; Nucleosomes

History of cell death Although studies on cell death were reported to be performed by Aristotle and later by Galen, who described the regression of larval and fetal structures during ontogenesis, it took until the 19th century for cell death processes to gain the interest of pathologists [1,2]. In 1842, Carl Vogt [3] discovered without the benefit of histochemical methods that cells can undergo programmed cell death. Further specifications and definitions of cell death were introduced by Virchow in 1858 (e.g., degeneration, softening, necrosis, mortification) [1,2], by Flemming in 1885 (spontaneous cell death, chromatolysis) [4], and by Graeper in 1914 (chromatolysis as a counterpart to mitosis) [5]. After World War II, embryologists continued the research on cellular demise and characterized the degradation of embryonic tissue as a result of single cell death [6]. In 1971, John F. Kerr [7] observed specific alterations of dying liver cells after occlusion of the vena portae in a rat model that he named shrinkage necrosis. One year later, he introduced with Andrew Wyllie and Sir Alastair Currie the term apoptosis (Greek: falling of the leaves) to describe this phenomenon and founded the field of modern cell death research [8].

Features of cell death Apoptosis typically affects single cells that are aged, dysfunctional, or damaged by external stimuli. It is an active, energy-requiring process leading to a well-regulated degradation of the cell. Early pathomorphological features are chromatin condensation and marginalization in the nucleus, DNA fragmentation into mono- and oligonucleosomal units, cellular shrinkage, karyorrhexis, packing of organelles, and dilatation of the endoplasmatic reticulum. Initially, mitochondria, lysosomes, and cellular membranes remain functionally intact. Later, budding of cellular membrane is observed leading to the packaging of cellular components into vesicles that are released as apoptotic bodies and phagocytyzed by macrophages and neighboring cells [1,9,10]. Loss of phospholipid asymmetry in the plasma membrane plays an important role in the opsoniza-

* Corresponding author. Institute of Clinical Chemistry, University Hospital Munich-Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany. Fax: +49-89-7095-6298. E-mail address: Petra.Stieber@med.uni-muenchen.de (P. Stieber).

0009-9120/$ - see front matter D 2004 The Canadian Society of Clinical Chemists. All rights reserved. doi:10.1016/j.clinbiochem.2004.05.003

606

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

tion of apoptotic bodies and subsequent phagocytosis [11 13]. Under physiologic conditions, a well-organized recycling system regulates the recognition, incorporation, and lysosomal ingestion of the cellular remnants avoiding inflammatory reactions in the tissue [8,10,14]. The early apoptotic events occur within minutes while the final stages of lysosomal degradation are finished within hours, the time course depending on cell type and tissue [1,9,10,15]. Necrosis characteristically affects groups of cells or whole tissue after extended damage induced by external stimuli like trauma, ischemia, and high dose irradiation. Loss of function of mitochondria and endoplasmatic reticulum leads to a dramatic breakdown of energy supply resulting in cellular, nuclear, and organellar swelling, blebbing of the membrane, and final rupture of the plasma membrane with release of lysosomal enzymes into the extracellular space. Neighboring cells and tissues in the vicinity are attacked by released proteases leading to extended inflammatory reactions [10]. In the nucleus, chromatin flocculation and nonspecific degradation of DNA by proteases is observed [16,17]. The whole process of necrosis is terminated within 12 24 h [1]. Characteristic features of apoptosis and necrosis are summarized in Table 1.
Table 1 Morphological and biochemical characteristics of apoptosis and necrosis Apoptosis Prevalence Localization Grade of activity Pathomechanism Affects single cells Margins of lesions or single cells Active, energy requiring ATP-dependent activation of proteases and endonucleases Loss of water Shrinkage Shrinkage Karyorrhexis Loss of nucleoli Nuclear condensation and marginalization Fragmentation into pieces of 50-300 kbp Fragmentation in multiples of 180 bp by endonucleases Functional Structurally intact Decreased transmembrane potential Budding Packaging of cellular content into membranederived vesicles Apoptotic bodies Phagocytosis by neighboring cells and tissue macrophages No Necrosis Affects groups of cells Center of extended lesions Passive, energy independent Dysfunctional ubiquitin system Dysfunctional ion pump Influx of sodium and water Swelling Swelling

Cell Nucleus

Chromatin

Flocculation Nonspecific fragmentation

Organelles Mitochondria

Membrane

Elimination

Dysfunctional Swelling Dysfunctional ubiquitin system Blebbing Loss of impermeability Rupture Release of cellular content Migration of macrophages Yes

Inflammation

In some cases, apoptosis and necrosis cannot be unambiguously distinguished by these characteristics. For example, cells with morphological necrotic features may show specific DNA fragmentation [18,19], while processes with apoptotic morphology may occur lacking the typical DNA fragmentation [19 21]. A variety of cell death modalities have been described that are intermediate in nature and appearance between necrosis and apoptosis [17,22 25]. As they were observed after both internal and external induction of cell death, it is hypothesized that the severity more than the mode of the stimuli influences the manner of cellular demise [1,17,24,26]. Other influencing factors are the cell type and its predilection for a certain mode of cell death. Whereas cells with high energy levels choose active apoptosis, those with a low energetic level frequently will undergo passive, necrotic cell death [27 29]. The intracellular calcium concentration and pH are also believed to play a role [30,31]. In respect to terminology of cell death, a clarification seems to be reasonable. Often, the terms apoptosis and programmed cell death are used incorrectly as synonymsprobably as the result of confounding two programs that contribute to physiological cell death: (A) a trigger program regulating the time point of cellular suicide, and (B) an execution program regulating the mode of cellular degradation. Programmed cell death describes the genetic triggering of cell death without any specification of the mode of cell death (program A). In contrast, apoptosis is defined as a cell death modality characterized by specific morphological and biochemical features that can be induced by genetic or external stimuli (program B). Depending on the trigger and the mode of cell death, programmed cell death and apoptosis can occur along with or apart from each other while indicating distinct parts of physiological cell death [1]. The temporal disconnect between morphological and biochemical changes further complicates establishing the exact mode of cell death. Processes leading to cell death and those that are the consequences have to be discriminated. In many cases, dying cells reach a point of no return early after the onset of a lethal stimulus. However, the morphological features become evident only hours later. Irreversible changes of the nucleus and cellular plasma characteristic of necrosis take place mainly postmortem [1,15,17]. These aspects have been integrated by Majno and Joris [1] in a new concept of cell death: oncosis (cellular swelling) and apoptosis (cellular shrinkage) are considered to be opposing modalities of cell death while necrosis is restricted to refer to morphological postmortem changes. However, additional forms of cell death like autophagic cell death, paraptosis, and cytoplasmatic cell death add further complexity [22,23]. A global view of cell death processes induced by physiological (intrinsic) and accidental (extrinsic) stimuli must take into account that apoptosis and oncosis coexist with varying mixtures of the

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

607

Fig. 1. Integrated cell death model indicating the complex interaction of cell death stimulus, trigger, modality, and postmortem status.

two observed. An integrated model of cell death modalities is shown in Fig. 1.

Cell death in physiological and pathological processes Homeostasis of cell proliferation and cell death plays an essential role in the maintenance of the function of organs and organisms [10,32]. These processes are characteristically unbalanced without adverse consequences during embryonic development, during establishment of immunologic tolerance, and during regeneration [33 39]. However, imbalance can also lead to adverse consequences such as the clonal expansion of malignancy when cells become resistant to apoptosis or proliferate excessively. Conversely, excessive cell death and/or diminished proliferation may be deleterious. Such imbalance occurs in degenerative diseases like AIDS, ischemia, toxin-mediated or autoimmune diseases, and in some types of cancer [34,38,40,41]. Cell death in cancer A more detailed view reveals the complexity of the processes leading to dyshomeostasis. The relative rates of cell death and cell proliferation may vary as malignancy progresses [38,42,43]. Initially, cell death may increase in parallel to cell proliferation, thereby leading to no net increase in cellular number. Should cell death mechanisms become attenuated, tumor will grow and invade local structures. Angiogenesis when induced will further support the rate of tumor proliferation. Concurrently, cell death could be further lessened or even enhanced. At later stages in the course of the malignancy, the rate of cell death may increase mirroring the presence of evermore dysfunctional cells [44,45]. In patients with malignant tumors, therapeutic strategies aim at equilibrating the imbalance between proliferation and degeneration. Depending on type and dosage of the chemotherapeutic drugs, the modality of radiotherapy, and the sensitivity of the tissue, cellular damage mostly results in arrest of the cell cycle and in response to insufficient repair to induction of active apoptotic cell death [46 49]. Quantifying cell death and cellular proliferation can provide information about the process of carcinogenesis

and the response to antitumor treatment. During physiological apoptosis, most of the cell death products are effectively removed by macrophages and neighboring cells and can be found in the circulation only in small amounts [10,13,50]. However, if the recycling system is impaired or overloaded by the malignant pathological condition, cell death products can accumulate in appreciable quantity in the circulation [51 53] and become potentially useful in the diagnosis, prognosis, and monitoring of disease. As many factors are involved in the process of cell death, it is crucial to choose the most cancer- and organ-specific biomarkers for these purposes. Further challenges are the stability of these factors in vivo and in vitro in serum or plasma, the confounding effects of renal insufficiency, infections, and hepatic failure on biomarker concentration, and the transient appearance of single markers and staggered temporal pattern of panels of multiple markers.

Factors of cell death Generally, intracellular components that are not secreted physiologically and appear in the extracellular space only after disintegration of the plasma membrane are suited for the quantification of cell death. Factors specifically taking part in apoptotic processes could be expected to deliver useful information regarding the mode of cell death. Apoptotic markers are involved at various levels. Receptors on the surface accept the death signal and transmit it across the cellular membrane. Specific intracellular enzymes are activated in order to coordinate the well-orchestrated breakdown of cellular structures. Sets of inhibitors and enhancers modulate the apoptotic signal. Finally, specific death substrates are degraded and externalized in a defined way. Receptors Various receptors are known to transmit the apoptotic signal. Most of them are members of the TNF family, including Fas/Apo1/CD95, TNFR1, DR3, DR4, and DR5. These receptors are glycosylated transmembrane proteins

608

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

consisting of an extracellular component of two to six cysteine rich domains and an intracellular part containing death domains. After binding of specific ligands or stimulating antibodies, the receptors are activated by trimerization that promotes the association of intracellular proteins with the death domains to form the death-inducing signaling complex (DISC) [35,54 56]. Some receptors like Fas/Apo1/CD95 transduce the signal directly to specific apoptotic effector caspases, while others like TNFR1 use alternative intracellular pathways via NF nB and JNK/AP-1,

which are also involved in immune response, differentiation, and proliferation [54,55]. Enzymes and pathways The intracellular transmission of the apoptotic signal is regulated by a well-organized system of initiator and effector caspases (cysteine-dependent aspartate-specific proteases). These proteases activate further procaspases by cutting specifically after an aspartate sequence. Initia-

Fig. 2. Summary of apoptotic pathways illustrating receptor activation, formation of the death-inducing signaling complex (DISC), and activation of initiator ). Alternatively to the direct involvement of the caspase cascade, the signal caspases like caspase-8 and of downstream effector caspases like caspase-3 ( can be enhanced by an amplification circle including mitochondrial release of cytochrome c and activation of caspase-9 in the apoptosome or induce additional mechanisms like the release of the apoptosis-inducing factor (AIF). All pathways merge in the degradation of cell death substrates like cytokeratins and nucleosomal fragmentation of chromatin. Inhibitors ( ) regulate various steps of the cascade and enable fine-tuning of the apoptosis process (see text).

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

609

tor procaspases bind to the DISC where they are autocatalytically transformed into their active form. After a cascade of proteolytic activation of further proteases, the signal finally reaches the effector caspases, which attack defined cellular molecules leading to the typical biochemical and morphological features of apoptosis [47,56,57]. With respect to the Fas/Apo1/CD95-receptor-mediated induction of apoptosis, the formation of the DISC and the activation of effector caspases are observed in all kinds of cells. However, differences occur in (1) the direct activation of the caspase cascade, (2) the involvement of the mitochondria and the release of cytochrome c, and (3) the release of apoptosis-inducing factor by the mitochondria [56,58,59]. Generally, apoptosis can proceed: (a) With rapid formation of the DISC and strong activation of initiator caspase 8, which is able to activate directly the caspase cascade including effector caspase 3 as well as mitochondria. (b) With slow DISC formation and weak activation of caspase 8. In this case, the apoptotic process requires the further activation of mitochondria by the molecule, BID, which results in the release of cytochrome c, cytoplasmatic aggregation of cytochrome c with apoptosis activating factor 1 (APAF-1), ATP and procaspase 9 in the apoptosome, and subsequent activation of the caspases 3 and 8. Caspase 8 further activates BID, thereby establishing an amplifying loop in the apoptoticsignaling pathway [56,58,59]. (c) With release by the mitochondria of apoptosis-inducing factor (AIF), which is capable of provoking morphologic characteristics of apoptosis in conjunction with and independently of the activation of caspases [60].

Inhibitors All essential steps of these pathways are regulated by inhibitors and enhancers that modulate the apoptotic process according to the demands and the possibilities of specific cells. Due to its central position in the transduction of the apoptotic signal, DISC is an important modulation site. FADD-like ICE inhibitory proteins (FLIPs) compete with procaspase 8 for the binding sites at death effector domains on the DISC and have been reported to inappropriately impair apoptosis in tumor cells and in virus-infected cells [58,64]. Other sites of modulation include the initiator caspases, the mitochondrial pathway, and the effector caspases themselves. Cytokine response modifier A (CrmA) effectively blocks caspases 1 and 8 [65 67]. Members of the bcl 2 family inhibit the stimulation of mitochondria by proapoptotic Bax, the release of cytochrome c and AIF into cytoplasm, and the recruiting of procaspase 9 to the apoptosome [34,68 70]. Inhibitors of effector caspases are named inhibitors of apoptosis (IAPs) and include survivin, XIAP, NAIP, and c-IAP-1 and 2. They bind to caspases 3, 7, and to the apoptosome and exhibit significant differences in their inhibiting potential [71 74], enabling effective finetuning of the apoptotic signal. This system is further optimized by IAP-regulating proteins like second mitochondria-derived activator of caspases (Smac) and direct IAPbinding protein with low pI (DIABLO) [47,75,76]. The most important components of the apoptotic pathways are illustrated in Fig. 2.

Methods of detecting cell death Many qualitative and quantitative methods are available to detect cell death by assaying the cell itself and through identifying apoptotic products released into the circulation (Table 2). Cellular- and serum-based tests show significant differences in sensitivity, specificity, and cost effectiveness. Originally, morphologic observations led to the characterization of various types of cell death [8,14]. Additional use of dyes enabled reliable quantitative determinations as well as functional tests of the plasma membrane and organelles. More elegant options for high throughput investigations were established by cytometry and fluorescent markers, which enable the interpretation of volume, structural, and functional changes such as the maintenance of phospholipid asymmetry in the plasma membrane. Further techniques detect cell-death-specific fragmentation of chromatin by DNA electrophoresis, cytometry, filtration assays, quantitative real-time PCR, and recognition of DNA breaks with various fluorescent markers. Finally, improved understanding of the regulating processes and signal pathways involved in cell death has led to new tests that measure the activity of caspases and the presence of specific substrates [17,77,78].

Substrates The various pathways of intracellular signal transduction merge into the activation of effector caspases with caspase 3 playing a particularly key role. These caspases (1) cleave structural components of the cytoskeleton and the nuclear membrane like actin, cytokeratins, and lamins; (2) cause phosphatidylserine to be exposed on the outside of the cellular membrane promoting phagocytation by macrophages and neighboring cells; (3) counteract the apoptosis-inhibiting effect of Bcl 2 und Bcl-xL proteins; (4) inhibit genes that regulate the repair of DNA lesions during cell cycle like mdm-2 and the Rb-gene; (5) inactivate enzymes responsible for stability, integrity, and repair of DNA, like poly-(ADP-ribose) polymerase PARP, DNAPK, and DNA replication factor 140; and (6) cleave ICAD (inhibitor of the caspase-activated DNase) and activate CAD (caspase-activated DNase), which causes the characteristic oligonucleosomal fragmentation of the chromatin [34,61 63].

610

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

Table 2 Methods for detection of apoptosis at cellular level and in blood Methods Cellular level Morphology Electron microscopy Light microscopy Cytometry Light scatter Integrity of plasma membrane Uptake of dyes Release of cellular enzymes Function of organelles AnnexinV-phosphatidylserine DNA fragmentation Agarose gel electrophoresis Fractioning Filtration Inverse field gel electrophoresis TUNEL ISEL Cytometry Polymerase chain reaction Activity of enzymes Inhibition of enzymes Cell death products Serum and plasma Receptors and ligands Components of cytoskeleton Intracellular enzymes Components of nucleus Cell death products and regulators Examples Qualitative Quantitative Cost

+++ + ++ + Trypane blue, propidium, iodide, FDA LDH, adenylate cyclase Rhodamine 123, acridine orange + + + ++ + + + + ++ ++ ++ +++ ++ ++ ++ + + + + +

+ ++ ++ ++ ++ ++ + ++ + + + ++ ++ ++ ++ ++ ++ ++ ++ ++ ++ ++ ++

+ ++ ++ ++ ++ ++ ++ ++ ++ ++ ++ ++ + + ++ + + ++ ++ ++ ++ ++ ++

Caspases, DNases ATA, CMK, FMK PARP, cytokeratins, nucleosomes Fas, FasL Cytokeratins, lamins NSE, LDH Matrix proteins, DNA, nucleosomes Caspases, cytochrome c, APAF1, PARP, ICAD, DNA, nucleosomes, survivin, IAPs, Bcl 2, p53

Qualitative and quantitative potential of the methods are indicated gradually ( low; + intermediate; ++ good; +++ very good), similarly costs ( very expensive; + expensive; ++ intermediate; +++ low priced).

These components and products of cell death can also be measured in serum and plasma, as well as in other body fluids like urine, cerebrospinal fluid, pleural effusions, and ascites. In addition, assays have been developed that detect and quantify apoptosis receptors and their ligands, elements of the cytoskeleton including cytokeratins and lamins, intracellular enzymes like LDH and NSE, free DNA and nucleosomes originating from the nucleus, caspases, their enhancers and inhibitors, and substrate breakdown products. Though blood investigations often lack organ and cell death specificity, because blood is easily drawn, circulating markers lend themselves to serial measurement that aids in monitoring the course of disease management.

sFas and its ligand FasL, cytokeratins, and circulating DNA fragments. Although several assays exist to measure caspase activity, to quantify substrates like PARP, and to determine inhibitors of apoptosis proteins (IAPs) like survivin [79,80], little has been published regarding the clinical utility of circulating levels in cancer. Fas-receptor and Fas-ligand Primarily, it has been the antiapoptotic soluble form of the fas-receptor (sFas) and the proapoptotic fas-ligand (FasL) that have been investigated in serum (Table 3). In contrast to healthy persons, patients with various kinds of cancer showed elevated levels of sFas in colon cancer [81], gastric cancer [82], liver cancer [83], breast cancer [84,85], gynecological cancers [86,87], renal cancer [88,89], melanoma [90,91], and non-Hodgkins lymphoma [92]. In some of these tumor types, sFas correlated with tumor stage [82,84,86,88,91] and with progressive disease [93]. However, several studies have reported that sFas is not elevated in cancer [94,95] and not correlated to stage [94,96]. Soluble Fas ligand (sFasL) has been reported to be elevated in gastric cancer [97], bladder cancer [98], and melanoma [90] and to have an association with tumor stage [98].

Circulating apoptotic markers in cancer Among the various apoptosis assays in blood, several have been evaluated clinically to determine their usefulness for diagnosis, prognosis, therapy monitoring, therapy prediction, and detection of recurrent disease in patients with cancer disease. The most frequent markers investigated in cancer patients have been the soluble forms of the apoptosis receptor

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617 Table 3 Studies on soluble Fas receptor and Fas ligand Author Kushlinskii et al. [81] Liang et al. [82] Peng et al. [83] Sheen-Chen et al. [84] Nonomura et al. [88] Mouawad et al. [90] Pignataro et al. [95] Ichikura et al. [97] Ueno et al. [85] Munker et al. [94] Osorio et al. [93] Marker sFas sFas sFas sFas sFas sFas, sFasL sFas, sFasL sFasL sFas sFas sFas Cancer Colon cancer Gastric cancer HCC Breast cancer Renal cancer Melanoma Laryngeal cancer Gastric cancer Breast cancer NHL B-CLL n 33 42 36 57 31 45 26 112 233 66 83 Control Healthy Healthy Liver cirrhosis Benign Healthy Healthy Healthy Healthy Healthy Healthy Healthy 34 35 155 118 20 21 n 33 30 49 12 Results elevated in cancer elevated in cancer elevated in cancer elevated in cancer elevated in cancer elevated in cancer not elevated in cancer elevated in cancer elevated in cancer not elevated in cancer elevated in progressive disease elevated in cancer elevated in cancer elevated in cancer elevated in cancer elevated in cancer elevated in cancer not elevated in cancer elevated in cancer Correlation with stage no yes Prognosis univariate Prognosis multivariate

611

yes yes

no no yes

high sFas poor PFS and OS no correlation with PFS high sFas poor OS

sFas independent for PFS and OS

sFas not independent

Konno et al. [86] Hara et al. [96] Niitsu et al. [92] Kimura et al. [89] Ugurel et al. [91] Hefler et al. [87] Tsutsumi et al. [101] Mizutani et al. [98] Bewick et al. [99] Mizutani et al. [100] Kanda et al. [102]

sFas sFas sFas sFas sFas sFas sFasL sFasL sFas sFas, sFasL sFasL

Gynecological cancers NHL NHL Renal cancer Melanoma Ovarian cancer Gastric cancer Bladder cancer Breast cancer Bladder cancer Multiple Myeloma

64 67

Healthy Healthy Healthy

24 36

yes no

high sFAS poor OS high sFAS poor OS high sFAS poor OS high sFAS poor OS

sFas independent for OS

72 125 52 166 163 94

Healthy Healthy Healthy, benign Healthy Healthy

17 30 65 43

yes

high sFas poor PFS and OS high sFas poor PFS and OS high sFasL poor OS high sFasL poor PFS high sFas poor PFS and OS high sFas and sFasL poor PFS no

sFas independent for OS sFas independent for OS sFas independent for PFS not for OS sFas independent for PFS and OS sFasL independent for OS

yes

sFas independent for PFS and OS

35

no

Summary of studies dealing on the diagnostic and prognostic value of soluble Fas receptor (sFas) and Fas ligand (sFasL) illustrating the markers investigated, type of cancer diseases, control groups, number of patients (n), diagnostic results, correlation with stage, univariate and multivariate prognostic results concerning overall survival (OS), and progression-free survival (PFS).

The prognostic value of both markers has been investigated in a variety of cancers. In univariate analyses, high sFas levels were related to poor overall survival (OS) in breast cancer [85,99], gynecological cancers [86,87], melanoma [91], renal cancer [89], non-Hodgkins lymphoma [92,96], and B-CLL [93] and associated with poor progression-free survival (PFS) in breast cancer [85,99], ovarian cancer [87], bladder cancer [100], and melanoma [91]. sFasL revealed prognostic information for PFS in bladder cancer [98,100] and gastric cancer [101]. Other studies report no prognostic value for sFas in non-Hodgkins

lymphoma [94] and for sFasL in multiple myeloma [102]. In multivariate analyses, sFas proved to be an independent prognostic factor only for breast cancer [85,99], gynecological cancers [86,87], melanoma [91], renal cancer [89], and non-Hodgkins lymphoma [92]. Cytokeratins Cytokeratins are intracellular structural proteins quite specific to epithelial cells that are cleaved and released during apoptosis [103 105]. Although their soluble frag-

612

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

ments are not tumor specific, they have been quantified in a number of studies to establish their utility in tumor diagnosis, prognosis, therapy monitoring, and detection of recurrent disease. The fragments measured have included CYFRA 21-1 (cytokeratin 19-fragments), which is the most widely used and specific of the cytokeratin markers in clinical practice [106 108], tissue polypeptide antigen (TPA, cytokeratin 8-, 18-, and 19-fragments), and tissue polypeptide-specific antigen (TPS, cytokeratin 18-fragments) [103]. Recently, a new cytokeratin fragment specific to early apoptosis has been identified [109]. It arises from cleavage of cytokeratin 18 by caspase 8. Such cleavage unmasks an epitope that is specifically detected by the M30-antibody. Further studies suggest that different portions of circulating cytokeratin 18 may be useful
Table 4 Studies on circulating DNA fragments Author Leon et al. [157] Marker DNA Cancer Various cancers GI cancers Lung cancer Lung cancer Various cancers Breast cancer Various cancers Various cancers Breast cancer n 173 199 45 68 185 96 220 418 125 Control Healthy n

to distinguish between apoptotic and necrotic processes [110]. Circulating levels of CYFRA 21-1, TPA, and TPS show some association with tumor stage and the rate of cancer cell lysis during therapy. Diagnostic and differential diagnostic value have been demonstrated in squamous cell cancers of the lung and the bladder with CYFRA 21-1 exhibiting the best profile of sensitivity and specificity [111 117]. Serum elevation has also been reported in small cell lung cancer, cervical, nasopharyngeal, esophageal, gastric, cholangiocellular, liver, colorectal, breast, and ovarian cancer [118 134]. Nonmalignant causes of cytokeratin elevations include renal insufficiency and severe infections with high rates of cell death [107,108,117,135,136]. Prognostic value of CYFRA 21-1 has been found in nonsmall cell and small

Results

Correlation Prognosis with stage yes

Therapy n RT

Monitoring therapy

Shapiro et al. [158] DNA Maebo [159] DNA

Fournie et al. [160] DNA Holdenrieder et al. [162] Kuroi et al. [161] Holdenrieder et al. [166] Holdenrieder et al. [165] Kuroi et al. [164] Nucleosomes Nucleosomes Nucleosomes Nucleosomes Nucleosomes

Sozzi et al. [167] Trejo-Becerril et al. [169] Sozzi et al. [171] Silva et al. [177] Lo et al. [163] Lo et al. [180]

DNA

Lung cancer Nucleosomes Cervical cancer DNA Lung cancer DNA Breast cancer EBV-DNA Nasopharyngeal cancer EBV-DNA Nasopharyngeal cancer EBV-DNA EBV-DNA

81 11 100 147 57 25

55 elevated in cancer Healthy, 275 elevated benign in cancer Healthy, 113 elevated benign in cancer Healthy elevated in cancer Healthy, 80 elevated infections in cancer Healthy 111 elevated in cancer Healthy, 90 elevated infections in cancer Healthy, 172 elevated benign in cancer Healthy, 212 elevated in cancer benign, other ca Healthy 32 elevated in cancer Healthy 14 elevated in cancer Healthy 100 elevated in cancer Healthy 35 elevated in cancer Healthy 43 elevated in cancer elevated in recurrent cancer

173 yes

yes yes yes (univariately) CT, RT no yes CT, RT no CT 36 yes, early peak 18 yes, early peak 38 yes 11 yes, early peak 35 yes no (only tendency) 24 yes, early peak

no

no

FU CT

no yes (univariately) yes

FU

RT FU

15 yes 17 yes

Lo et al. [181] Lo et al. [176]

Nasopharyngeal 10 cancer Nasopharyngeal 91, 139 cancer Lymphoma 26 Healthy

RT elevated in early recur. cancer 35 elevated in cancer yes (uni- or multivariately) yes (univariately) CT, RT

10 early peak

Lei et al. [178]

EBV-DNA

15 yes

Summary of studies dealing on the value of circulating DNA fragments for diagnosis, prognosis, and therapy monitoring. Markers investigated, type of cancer diseases, control groups, number of patients (n), diagnostic results, correlation with stage, prognostic results, kinetic investigations during therapies (CT = chemotherapy, RT = radiotherapy, FU = follow-up), and results of therapy monitoring are indicated.

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617

613

cell cancer of the lung [137 141], mesothelioma [142], esophageal cancer [143], and cervical cancer [144]. CYFRA 21-1 has additional proven utility in monitoring treatment in lung cancer [145,146], head and neck cancer [147,148], and cervical cancer [149], in predicting response to therapy in lung cancer [146,150,151], and in predicting recurrent disease in lung cancer [152,153]. The cytokeratin marker M30 was increased in sera of breast cancer patients but failed to show prognostic value [154]. Circulating DNA fragments During apoptosis, chromatin is characteristically cleaved into mono- and oligonucleosomal fragments that are released into the circulation after disintegration of the cellular membrane [10,14,51,53]. In serum and plasma, circulating nucleic acids are conserved by their close association with histone protein from further digestion by endonucleases [155,156]. Elevated levels of circulating DNA fragments have been found in the blood of patients with most types of malignancy (Table 4) including lung, gastrointestinal, colorectal, breast, gynecological, renal, and nasopharyngeal cancers as well as lymphoma [157 171]. However, diagnostic value is limited because nonmalignant conditions associated with high cellular turnover such as those arising from severe bacterial infections also cause elevation of DNA fragments in the circulation [162,165]. Additional qualitative investigations detecting characteristic mutations, alterations of microsatellites, and epigenetic changes might increase the specificity for tumor DNA and improve the diagnostic power for cancer disease [172 175]. The amount of circulating DNA has prognostic significance in various cancers [160,176 178]. The same applies to some of the tumorspecific features of the circulating DNA [168,173,174]. Concerning the monitoring of therapy, patients responding to chemo- and radiotherapy showed declining values whereas those with insufficient response exhibited constantly high or even increasing values. These observations were valid for patients with various solid tumors as well as for lymphoma [157,159,164,165,167,169,179,180]. Several studies have focused on measuring the initial increase of circulating DNA fragments shortly after application of antitumor therapy [162,164,165,169]. A similar phenomenon has been described for EBV-DNA in the plasma of patients with nasopharyngeal cancer during radiotherapy [181]. The extent of these early changes of circulating DNA fragments in the serum of patients with lung cancer during chemotherapy is reported to predict the response to therapy [182].

they can contribute in assessing the extent and aggressiveness of disease. Additionally, many of them show prognostic or predictive information in various cancers. As they are easily and noninvasively accessible in blood, they offer the possibility for serial investigations. The temporal rise of circulating apoptotic markers during chemo- and radiotherapy shows high potential to objectively assess disease management and to predict treatment response in advance of current methods. Large-scale studies of the apoptotic markers evaluated in conjunction with multiple clinical parameters and other biochemical markers are encouraged to more fully establish the utility of the apoptotic markers in cancer management.

References
[1] Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol 1995;146:3 15. [2] Clarke PGH, Clarke S. Nineteenth century research on naturally occurring cell death and related phenomena. Anat Embryol 1996; 193:81 99. [3] Vogt C. Untersuchungen u ber die Entwicklungsgeschichte der Geburtshelferkro te (Alytes obstetricans). Solothurn 1842. ber die Bildung von Richtungsfiguren in Sa [4] Flemming W. U ugethiereiern beim Untergang Graafscher Follikel. Arch Anat Entwicklgesch 1885;221 44. [5] Graeper L. Eine neue Anschauung u ber physiologische Zellausschaltung. Arch Zellforsch 1914;12:373 94. [6] Gluecksmann A. Cell deaths in normal vertebrate ontogeny. Biol Rev 1951;26:59 86. [7] Kerr JF. Shrinkage necrosis: a distinct mode of cellular death. J Pathol 1971;105:13 20. [8] Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972;26:239 57. [9] Arends MJ, Morris RG, Wyllie AH. Apoptosis. The role of the endonuclease. Am J Pathol 1990;136:593 608. [10] Kerr JF, Winterford CM, Harmon BV. Apoptosis. Its significance in cancer and cancer therapy. Cancer 1994;73:2013 26. [11] Fadock VA, Voelker DR, Campbell PA, Cohen JJ, Bratton DL, Henson PM. Exposure of phosphatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages. J Immunol 1992;148:2207 16. [12] Verhoven B, Schlegel RA, Williamson P. Mechanisms of phosphatidylserine exposure, a phagocyte recognition signal, on apoptotic T lymphocytes. J Exp Med 1995;182:1597 601. [13] Hengartner MO. Apoptosis: corraling the corpses. Cell 2001;104: 325 8. [14] Wyllie AH. Cell death: the significance of apoptosis. Int Rev Cytol 1980;68:251 306. [15] Kanduc D, Mittelman A, Serpico R, Sinigaglia E, Sinha AA, Natale C, et al. Cell death: apoptosis versus necrosis (review). Int J Oncol 2002;21:165 70. [16] Cotter TG, Lennon SV, Glynn JG, Martin SJ. Cell death via apoptosis and its relationship to growth, development and differentiation of both tumour and normal cells. Anticancer Res 1990;10:1153 60. [17] Darzynkiewicz Z, Juan G, Li X, Gorczyca W, Murakami T, Traganos F. Cytometry in cell necrobiology: analysis of apoptosis and accidental cell death (necrosis). Cytometry 1997;27:1 20. [18] Collins RJ, Harmon BV, Gobe GC, Kerr JF. Internucleosomal DNA cleavage should not be the sole criterion for identifying apoptosis. Int J Radiat Biol 1992;61:451 3.

Conclusion Circulating apoptotic markers can be detected in serum and plasma and are often elevated in patients with cancer. Many of them lack organ and tumor specificity, which limits their diagnostic and differential diagnostic value. However,

614

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617 angiogenesis and tumor cell apoptosis during colorectal carcinogenesis. Clin Cancer Res 1999;5:135 42. Braithwaite KL, Rabbitts PH. Multi-step evolution of lung cancer. Semin Cancer Biol 1999;9:255 65. Sionov RV, Haupt Y. The cellular response to p53: the decision between life and death. Oncogene 1999;18:6145 57. Hengartner MO. The biochemistry of apoptosis. Nature 2000; 407:770 6. Rich T, Allen RL, Wyllie AH. Defying death after DNA damage. Nature 2000;407:777 83. Schaer P. Spontaneous DNA damage, genome instability, and cancerWhen DNA replication escapes control. Cell 2001;104: 329 32. Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature 2000;407:784 8. Lichtenstein AV, Melkonyan HS, Tomei LD, Umansky SR. Circulating nucleic acids and apoptosis. Ann N Y Acad Sci 2001; 945:239 49. Stroun M, Maurice P, Vasioukhin V, Lyautey J, Lederrey C, Lefort F, et al. The origin and mechanism of circulating DNA. Ann N Y Acad Sci 2000;906:161 8. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001;61:1659 65. Nagata S. Apoptosis by death factor. Cell 1997;88:355 65. Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science 1998;281:1305 8. Krammer PH. CD95s deadly mission in the immune system. Nature 2000;407:789 95. Slee EA, Adrain C, Martin SJ. Serial killers: ordering caspase activation events in apoptosis. Cell Death Differ 1999;6:1067 74. Sartorius U, Schmitz I, Krammer P. Molecular mechanisms of deathreceptor-mediated apoptosis. ChemBioChem 2001;2:20 9. Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ, et al. Two CD95 (APO-1/Fas) signaling pathways. EMBO J 1998;17:1675 87. Joza N, Susin SA, Daugas E, Stanford WL, Cho SK, Li CYJ, et al. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death. Nature 2001;410:549 54. Nicholson DW. Caspase structure, proteolytic substrates, and function during apoptotic cell death. Cell Death Differ 1999;6:1028 42. Samali A, Zhivotovsky B, Jones D, Nagata S, Orrenius S. Apoptosis: cell death defined by caspase activation. Cell Death Differ 1999; 6:495 6. Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, Nagata S. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 1998;391:43 50. Muzio M, Chinnaiyan AM, Kischkel FC, ORourke K, Shevchenko A, Ni J, et al. FLICE, a novel FADD-homologous ICE/ ced-3-like protease, is recruited to the CD95 (Fas/Apo-1) deathinducing signaling complex. Cell 1996;85:817 27. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998;281: 1309 13. Ekert PG, Silke J, Vaux DL. Caspase inhibitors. Cell Death Differ 1999;6:1081 6. Budihardjo I, Oliver H, Lutter M, Luo X, Wang X. Biochemical pathways of caspase activation during apoptosis. Annu Rev Cell Dev Biol 1999;15:269 90. Adams JM, Cory S. The Bcl-2 protein family: arbiters of cell survival. Science 1998;281:1322 6. Kroemer G. The proto-oncogene Bcl-2 and its role in regulating apoptosis. Nat Med 1997;3:614 9. Reed JC. Bcl-2 family proteins: strategies for overcoming chemoresistance in cancer. Adv Pharmacol 1997;41:501 32. Thornberry NA, Lazebnik Y. Caspases: enemies within. Science 1998;281:1312 6.

[19] Stewart BW. Mechanisms of apoptosis: integration of genetic, biochemical, and cellular indicators. J Natl Cancer Inst 1994;86: 1286 96. [20] Cohen GM, Sun XM, Snowden RT, Dinsdale D, Skilleter DN. Key morphological features of apoptosis may occur in the absence of internucleosomal DNA fragmentation. Biochem J 1992;286: 331 4. [21] Oberhammer F, Wilson JW, Dive C, Morris ID, Hickman JA, Wakeling AE, et al. Apoptotic death in epithelial cells: cleavage of DNA to 300 and/or 50 kb fragments prior to or in the absence of internucleosomal fragmentation. EMBO J 1993;12:3679 84. [22] Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science 2000;290:1717 21. [23] Sperandio S, de Bell I, Bredesen DE. An alternative, nonapoptotic form of programmed cell death. Proc Natl Acad Sci U S A 2000;97: 14376 81. [24] Leist M, Ja a tela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev, Mol Cell Biol 2001;2:1 10. [25] Lockshin RA, Osborne B, Zakeri Z. Cell death in the third millennium. Cell Death Differ 2000;7:2 7. [26] Nicotera P, Leist M, Single B, Volbracht C. Execution of apoptosis: converging or diverging pathways? Biol Chem 1999;380:1035 40. [27] Richter C, Schweizer M, Cossarizza A, Franceschi C. Control of apoptosis by the cellular ATP level. FEBS Lett 1996;378:107 10. [28] Nicotera P, Leist M, Ferrando-May E. Intracellular ATP, a switch in the decision between apoptosis and necrosis. Toxicol Lett 1998; 102 103:139 42. [29] Leist M, Single B, Castoldi AF, Kuhnle S, Nicotera P. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J Exp Med 1997;185:1481 6. [30] Matsuyama S, Llopis J, Deveraux QL, Tsien RY, Reed JC. Changes in intramitochondrial and cytosolic pH: early events that modulate caspase activation during apoptosis. Nat Cell Biol 2000;2: 318 25. [31] Leist M, Nicotera P. Calcium and neuronal death. Rev Physiol Biochem Pharmacol 1998;132:79 125. [32] Cummings MC, Winterford CM, Walker NI. Apoptosis. Am J Surg Pathol 1997;21:88 104. [33] Prindull G. Apoptosis in the embryo and tumorigenesis. Eur J Cancer 1995;31:116 23. [34] Granville DJ, Carthy CM, Hunt DWC, McManus BM. Apoptosis: molecular aspects of cell death and disease. Lab Invest 1998; 78:893 913. [35] Krammer PH. CD95 (APO-1/Fas)-mediated apoptosis: live and let die. Adv Immunol 1999;71:163 210. [36] Ucker DS. Death and dying in the immune system. Adv Pharmacol 1997;41:179 218. [37] Desmouliere A, Badid C, Bochaton-Piallat ML, Gabbiani G. Apoptosis during wound healing, fibrocontractive diseases and vascular wall injury. Int J Biochem Cell Biol 1997;29:19 30. [38] Fadeel B, Orrenius S, Zhivotovsky B. Apoptosis in human disease: a new skin for the old ceremony? Biochem Biophys Res Commun 1999;266:699 717. [39] Warner HR, Hodes RJ, Pocinki K. What does cell death have to do with aging? J Am Geriatr Soc 1997;45:1140 6. [40] Robertson JD, Fadeel B, Zhivotovsky B, Orrenius S. Centennial nobel conference on apoptosis and human disease. Cell Death Differ 2002;9:468 75. [41] Saikumar P, Dong Z, Mikhailov V, Denton M, Weinberg JM, Venkatachalam MA. Apoptosis: definition, mechanisms, and relevance to disease. Am J Med 1999;107:489 506. [42] Reed JC. Dysregulation of apoptosis in cancer. J Clin Oncol 1999;17:2941 53. [43] Zoernig M, Hueber AO, Baum W, Evan G. Apoptosis regulators and their role in tumorigenesis. Biochim Biophys Acta 2001;1551: F1 37. [44] Aotake T, Lu CD, Chiba Y, Muraoka R, Tanigawa N. Changes of

[45] [46] [47] [48] [49]

[50] [51]

[52]

[53]

[54] [55] [56] [57] [58] [59]

[60]

[61] [62]

[63]

[64]

[65] [66] [67]

[68] [69] [70] [71]

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617 [72] Ja a ttela M. Escaping cell death: survival proteins in cancer. Exp Cell Res 1999;248:30 43. [73] Deveraux QL, Stennicke HR, Salvesen GS, Reed JC. Endogenous inhibitors of caspases. J Clin Immunol 1999;19:388 98. [74] Goyal L. Cell death inhibition: keeping caspases in check. Cell 2001;104:805 8. [75] Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000;102:33 42. [76] Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000;102:43 53. [77] Darzynkiewicz Z, Li X, Gong J. Assays of cell viability: discrimination of cells dying by apoptosis. Methods Cell Biol 1994;41: 15 38. [78] Mesner PW, Kaufmann SH. Methods utilized in the study of apoptosis. Adv Pharmacol 1997;41:57 87. [79] Hentze H, Schoebel F, Lund S, Kehl M, Ertel W, Wendel A, et al. In vivo and in vitro evidence for extracellular caspase activity released from apoptotic cells. Biochem Biophys Res Commun 2001;283: 1111 7. [80] Smith SD, Wheeler MA, Plescia J, Colberg JW, Weiss RM, Altieri DC. Urine detection of survivin and diagnosis of bladder cancer. JAMA 2001;285:324 8. [81] Kushlinskii NE, Britvin TA, Abbasova SG, Perevoshchikov AG, Prorokov VV, Kostanyan IA, et al. Soluble Fas antigen in the serum of patients with colon cancer. Bull Exp Biol Med 2001;131:361 3. [82] Liang QL, Pan DC, Yin ZM, Liu GX, Yang Q, Xie JR, et al. Clinical value of serum soluble Apo-1/Fas for predicting biological behaviors and prognosis of gastric carcinoma. Ai Zheng 2002;21:174 6. [83] Peng Z, Tang H, Ling Y, Han G. Apoptosis and Fas system are significantly involved in the process of liver cirrhosis converting into hepatocellular carcinoma. J Tongji Med Univ 2001;21:126 9. [84] Sheen-Chen SM, Chen HS, Eng HL, Chen WJ. Circulating soluble Fas in patients with breast cancer. World J Surg 2003;27:10 3. [85] Ueno T, Toi M, Tominaga T. Circulating soluble Fas concentration in breast cancer patients. Clin Cancer Res 1998;5:3529 33. [86] Konno R, Takano T, Sato S, Yajima A. Serum soluble fas level as a prognostic factor in patients with gynecological malignancies. Clin Cancer Res 2000;6:3576 80. [87] Hefler L, Mayerhofer K, Nardi A, Reinthaller A, Kainz C, Tempfer C. Serum soluble Fas levels in ovarian cancer. Obstet Gynecol 2000;96:65 9. [88] Nonomura N, Nishimura K, Ono Y, Fukui T, Harada Y, Takaha N, et al. Soluble Fas in serum from patients with renal cell carcinoma. Urology 2000;55:151 5. [89] Kimura M, Tomita Y, Imai T, Saito T, Katagiri A, Tanikawa T, et al. Significance of serum-soluble CD95 (Fas/APO-1) on prognosis in renal cell cancer patients. Br J Cancer 1999;80:1648 51. [90] Mouawad R, Khayat D, Soubrane C. Plasma Fas ligand, an inducer of apoptosis, and plasma soluble Fas, an inhibitor of apoptosis, in advanced melanoma. Melanoma Res 2000;10:461 7. [91] Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased soluble CD95 (sFas/CD95) serum level correlates with poor prognosis in melanoma patients. Clin Cancer Res 2001;7:1282 6. [92] Niitsu N, Sasaki K, Umeda M. A high serum soluble Fas/APO-1 level is associated with a poor outcome of aggressive non-Hodgkins lymphoma. Leukemia 1999;13:1434 40. [93] Osorio LM, Aguilar-Santelises M, De Santiago A, Hachiya T, Mellstedt H, Jondal M. Increased serum levels of soluble Fas in progressive B-CLL. Eur J Haematol 2001;66:342 6. [94] Munker R, Younes A, Cabanillas F, Andreeff M. Soluble CD95 in the serum of patients with low and intermediate grade malignant lymphomas: absence of prognostic correlations. Leuk Lymphoma 1997;27:517 21. [95] Pignataro L, Arisi E, Sambataro G, Corsi MM. Soluble Fas (sFas)

615

[96]

[97]

[98]

[99]

[100]

[101]

[102]

[103] [104]

[105]

[106]

[107]

[108] [109]

[110]

[111]

[112]

[113]

and soluble Fas ligand (sFas-L) balance in laryngeal carcinoma before and after surgical treatment. J Surg Oncol 2003;83:112 5. Hara T, Tsurumi H, Takemura M, Goto H, Yamada T, Sawada M, et al. Serum-soluble fas level determines clinical symptoms and outcome of patients with aggressive non-Hodgkins lymphoma. Am J Hematol 2000;64:257 61. Ichikura T, Majima T, Uchida T, Okura E, Ogawa T, Mochizuki H. Plasma soluble Fas ligand concentration: decrease in elderly men and increase in patients with gastric carcinoma. Oncol Rep 2001; 8:311 4. Mizutani Y, Hongo F, Sato N, Ogawa O, Yoshida O, Miki T. Significance of serum soluble Fas ligand in patients with bladder carcinoma. Cancer 2001;92:287 93. Bewick M, Conlon M, Parissenti AM, Lee H, Zhang L, Gluck S, et al. Soluble Fas (CD95) is a prognostic factor in patients with metastatic breast cancer undergoing high-dose chemotherapy and autologous stem cell transplantation. J Hematother Stem Cell Res 2001;10:759 68. Mizutani Y, Yoshida O, Ukimura O, Kawauchi A, Bonavida B, Miki T. Prognostic significance of a combination of soluble Fas and soluble Fas ligand in the serum of patients with Ta bladder cancer. Cancer Biother Radiopharm 2002;17:563 7. Tsutsumi S, Kuwano H, Shimura T, Morinaga N, Mochiki E, Asao T. Circulating soluble Fas ligand in patients with gastric carcinoma. Cancer 2000;89:2560 4. Kanda Y, Ara C, Chizuka A, Yamamoto R, Hamaki T, Suguro M, et al. Lack of correlation between clinical characteristics and serum soluble Fas ligand levels in patients with multiple myeloma. Leuk Lymphoma 2001;40:351 6. Buccheri G, Ferrigno D. Lung tumor markers of cytokeratin origin: an overview. Lung Cancer 2001;34:S65 9. Sheard MA, Vojtesek B, Simickova M, Valik D. Release of cytokeratin-18 and -19 fragments (TPS and CYFRA 21-1) into the extracellular space during apoptosis. J Cell Biochem 2002;85: 670 7. Wu F, Fujita J, Murota M, Li JQ, Ishida T, Nishioka M, et al. CYFRA 21-1 is released in TNF-alpha-induced apoptosis in the hepatocellular carcinoma cell line HuH-7. Int J Oncol 2002;21:441 5. Bodenmueller H, Ofenloch-Haehnle B, Lane EB, Dessauer A, Bottger V, Donie F. Lung cancer-associated keratin 19 fragments: development and biochemical characterisation of the new serum assay Enzymun-Test CYFRA 21-1. Int J Biol Markers 1994;9: 75 81. Stieber P, Hasholzner U, Bodenmueller H, Nagel D, Sunder-Plassmann L, Dienemann H, et al. CYFRA 21-1. A new marker in lung cancer. Cancer 1993;72:707 13. Stieber P. Sensible use of tumor markers. J Lab Med 2001;25: 327 36. Leers MPG, Koelgen W, Bjoerklund V, Bergman T, Tribbick G, Persson B, et al. Immunocytochemical detection and mapping of a cytokeratin 18 neo-epitope exposed during early apoptosis. J Pathol 1999;187:567 72. Kramer G, Erdal H, Mertens HJ, Nap M, Mauermann J, Steiner G, et al. Differentiation between cell death modes using measurements of different soluble forms of extracellular cytokeratin 18. Cancer Res 2004;64:1751 6. Stieber P, Dienemann H, Hasholzner U, Muller C, Poley S, Hofmann K, et al. Comparison of cytokeratin fragment 19 (CYFRA 21-1), tissue polypeptide antigen (TPA) and tissue polypeptide specific antigen (TPS) as tumour markers in lung cancer. Eur J Clin Chem Clin Biochem 1993;31:689 94. Stieber P, Dienemann H, Hasholzner U, Fabricius PG, Schambeck C, Weinzierl M, et al. Comparison of CYFRA 21-1, TPA and TPS in lung cancer, urinary bladder cancer and benign diseases. Int J Biol Markers 1994;9:82 8. Pujol JL, Grenier J, Parrat E, Lehmann M, Lafontaine T, Quantin X, et al. Cytokeratins as serum markers in lung cancer: a comparison

616

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617 of CYFRA 21-1 and TPS. Am J Respir Crit Care Med 1996;154: 725 33. Giovanella L, Ceriani L, Bandera M, Rimoldi R, Beghe B, Roncari G. Tissue polypeptide specific antigen (tps) and cytokeratin 19 fragment (CYFRA 21.1) immunoradiometric assay in non small cell lung cancer evaluation. Q J Nucl Med 1995;39:285 9. Molina R, Filella X, Auge JM, Fuentes R, Bover I, Rifa J, et al. Tumor markers (CEA, CA 125, CYFRA 21-1, SCC and NSE) in patients with non-small cell lung cancer as an aid in histological diagnosis and prognosis. Comparison with the main clinical and pathological prognostic factors. Tumour Biol 2003;24:209 18. Pujol JL, Grenier J, Daures JP, Daver A, Pujol H, Michel FB. Serum fragment of cytokeratin subunit 19 measured by CYFRA 21-1 immunoradiometric assay as a marker of lung cancer. Cancer Res 1993;53:61 6. Schalhorn A, Fuerst H, Stieber P. Tumor markers in lung cancer. J Lab Med 2001;25:353 61. Molina R, Agusti C, Filella X, Jo J, Joseph J, Gimenez N, et al. Study of a new tumor marker, CYFRA 21-1, in malignant and nonmalignant diseases. Tumour Biol 1994;15:318 25. Giovanella L, Piantanida R, Ceriani L, Bandera M, Novario R, Bianchi L, et al. Immunoassay of neuron-specific enolase (NSE) and serum fragments of cytokeratin 19 (CYFRA 21.1) as tumor markers in small cell lung cancer: clinical evaluation and biological hypothesis. Int J Biol Markers 1997;12:22 6. Yazigi R, Castillo R, Aliste G, Garrido J, Opazo A, Prado S, et al. Cyfra 21-1 marker in carcinoma of the cervix. Int J Gynecol Cancer 2000;10:203 6. Nasu K, Etoh Y, Yoshimatsu J, Matsu T, Narahara H, Miyakawa I. Serum levels of cytokeratin 19 fragments in cervical cancer. Gynecol Obstet Invest 1996;42:267 70. Ayude D, Gacio G, Paez de la Cadena M, Pallas E, Martinez-Zorzano VS, de Carlos A, et al. Combined use of established and novel tumour markers in the diagnosis of head and neck squamous cell carcinoma. Oncol Rep 2003;10:1345 50. Deng YF, Chen P, Lin YZ, Le JZ, Wu XL, Yu MQ, et al. Analytical and clinical evaluation of CYFRA 21-1 by electrochemiluminescent immunoassay in head and neck squamous cell carcinoma. J Laryngol Otol 2003;117:190 4. Tai CJ, Liu FY, Liang JA, Yang SN, Tsai MH, Kao CH. Comparison of CYFRA 21-1 and tissue polypeptide specific antigen (TPS) for detecting nasopharyngeal carcinoma. Anticancer Res 2002;22: 3793 6. Lee JK, Hsieh JF, Tsai SC, Ho YJ, Sun SS, Kao CH. Comparison of CYFRA 21-1 and squamous cell carcinoma antigen in detecting nasopharyngeal carcinoma. Ann Otol Rhinol Laryngol 2001;110: 775 8. Brockmann JG, St Nottberg H, Glodny B, Heinecke A, Senninger NJ. CYFRA 21-1 serum analysis in patients with esophageal cancer. Clin Cancer Res 2000;6:4249 52. Nakata B, Chung YS, Kato Y, Ogawa M, Ogawa Y, Inui A, et al. Clinical significance of serum CYFRA 21-1 in gastric cancer. Br J Cancer 1996;73:1529 32. Kashihara T, Ohki A, Kobayashi T, Sato T, Nishizawa H, Ogawa K, et al. Intrahepatic cholangiocarcinoma with increased serum CYFRA 21-1 level. J Gastroenterol 1998;33:447 53. Uenishi T, Kubo S, Hirohashi K, Tanaka H, Shuto T, Yamamoto T, et al. Cytokeratin-19 fragments in serum (CYFRA 21-1) as a marker in primary liver cancer. Br J Cancer 2003;88:1894 9. Yamamoto H, Miyake Y, Noura S, Ogawa M, Yasui M, Ikenaga M, et al. Tumor markers for colorectal cancer. Gan To Kagaku Ryoho 2001;28:1299 305. Giovanella L, Ceriani L, Giardina G, Bardelli D, Tanzi F, Garancini S. Serum cytokeratin fragment 21.1 (CYFRA 21.1) as tumour marker for breast cancer: comparison with carbohydrate antigen 15.3 (CA 15.3) and carcinoembryonic antigen (CEA). Clin Chem Lab Med 2002;40:298 303. [132] Rodriguez CA, Cruz JJ, Martin T, Gomez A, Olaverri A, Hernandez M. Serum CYFRA 21-1 is one of the most reliable tumor markers for breast carcinoma. Cancer 2002;95:670 1. [133] Nakata B, Ogawa Y, Ishikawa T, Ikeda K, Kato Y, Nishino H, et al. Serum CYFRA 21-1 is one of the most reliable tumor markers for breast carcinoma. Cancer 2000;89:1285 90. [134] Hasholzner U, Baumgartner L, Stieber P, Meier W, Hofmann K, Fateh-Moghadam A. Significance of the tumour markers CA 125 II, CA 72-4, CASA and CYFRA 21-1 in ovarian carcinoma. Anticancer Res 1994;14:2743 6. [135] Tramonti G, Ferdeghini M, Donadio C, Norpoth M, Annichiarico C, Bianchi R, et al. Renal function and serum concentration of five tumor markers (TATI, SCC, CYFRA 21-1, TPA, and TPS) in patients without evidence of neoplasia. Cancer Detect Prev 2000; 24:86 90. [136] Nakahama H, Tanaka Y, Fujita Y, Fujii M, Sugita M. CYFRA 21-1 and ProGRP, tumor markers of lung cancer, are elevated in chronic renal failure patients. Respirology 1998;3:207 10. [137] Pujol JL, Boher JM, Grenier J, Quantin X. Cyfra 21-1, neuron specific enolase and prognosis of non-small cell lung cancer: prospective study in 621 patients. Lung Cancer 2001; 31:221 31. [138] Reinmuth N, Brandt B, Semik M, Kunze WP, Achatzy R, Scheld HH, et al. Prognostic impact of Cyfra21-1 and other serum markers in completely resected non-small cell lung cancer. Lung Cancer 2002;36:265 70. [139] Kasimir-Bauer S, Schleucher N, Weber R, Neumann R, Seeber S. Evaluation of different markers in non-small cell lung cancer: prognostic value of clinical staging, tumour cell detection and tumour marker analysis for tumour progression and overall survival. Oncol Rep 2003;10:475 82. [140] Takei Y, Minato K, Tsuchiya S, Takise A, Nakano H, Ezawa K, et al. CYFRA 21-1: an indicator of survival and therapeutic effect in lung cancer. Oncology 1997;54:43 7. [141] Pujol JL, Quantin X, Jacot W, Boher JM, Grenier J, Lamy PJ. Neuroendocrine and cytokeratin serum markers as prognostic determinants of small cell lung cancer. Lung Cancer 2003;39:131 8. [142] Schouwink H, Korse CM, Bonfrer JM, Hart AA, Baas P. Prognostic value of the serum tumour markers Cyfra 21-1 and tissue polypeptide antigen in malignant mesothelioma. Lung Cancer 1999;25: 25 32. [143] Tsuchiya Y, Onda M, Miyashita M, Sasajima K. Serum level of cytokeratin 19 fragment (CYFRA 21-1) indicates tumour stage and prognosis of squamous cell carcinoma of the oesophagus. Med Oncol 1999;16:31 7. [144] Bonfrer JM, Gaarenstroom KN, Kenter GG, Korse CM, Hart AA, Gallee MP, et al. Prognostic significance of serum fragments of cytokeratin 19 measured by Cyfra 21-1 in cervical cancer. Gynecol Oncol 1994;55:371 5. [145] Ebert W, Muley T. CYFRA 21-1 in the follow-up of inoperable nonsmall cell lung cancer patients treated with chemotherapy. Anticancer Res 1999;19:2669 72. [146] Vollmer RT, Govindan R, Graziano SL, Gamble G, Garst J, Kelley MJ, et al. Serum CYFRA 21-1 in advanced stage non-small cell lung cancer: an early measure of response. Clin Cancer Res 2003;9: 1728 33. [147] Kuropkat C, Lippert BM, Werner JA. Follow-up with serum Cyfra 21-1 in patients with squamous cell carcinomas of the head and neck. Oncology 2002;63:280 5. [148] Pradier O, Hille A, Schmiberger H, Hess CF. Monitoring of therapy in head and neck patients during the radiotherapy by measurement of Cyfra 21-1. Cancer Radiother 2002;6:15 21. [149] Pras E, Willemse PH, Canrinus AA, de Bruijn HW, Sluiter WJ, ten Hoor KA, et al. Serum squamous cell carcinoma antigen and CYFRA 21-1 in cervical cancer treatment. Int J Radiat Oncol Biol Phys 2002;52:23 32. [150] Ray P, Quantin X, Grenier J, Pujol JL. Predictive factors of tumor

[114]

[115]

[116]

[117] [118]

[119]

[120]

[121]

[122]

[123]

[124]

[125]

[126]

[127]

[128]

[129]

[130]

[131]

S. Holdenrieder, P. Stieber / Clinical Biochemistry 37 (2004) 605617 response and prognostic factors of survival during lung cancer chemotherapy. Cancer Detect Prev 1998;22:293 304. Muley T, Dienemann H, Ebert W. Increased CYFRA 21-1 and CEA levels are negative predictors of outcome in p-stage I NSCLC. Anticancer Res 2003;23:4085 93. Sun SS, Hsieh JF, Tsai SC, Ho YJ, Lee JK, Kao CH. Cytokeratin fragment 19 and squamous cell carcinoma antigen for early prediction of recurrence of squamous cell lung carcinoma. Am J Clin Oncol 2000;23:241 3. Yeh JJ, Liu FY, Hsu WH, Wang JJ, Ho ST, Kao A. Monitoring cytokeratin fragment 19 (CYFRA 21-1) serum levels for early prediction of recurrence of adenocarcinoma and squamous cell carcinoma in the lung after surgical resection. Lung 2002;180:273 9. Ueno T, Toi M, Biven K, Bando H, Ogawa T, Linder S. Measurement of an apoptotic product in the sera of breast cancer patients. Eur J Cancer 2003;39:769 74. Rumore P, Muralidhar B, Lin M, Lai C, Steinman CR. Hemodialysis as a model for studying endogenous plasma DNA: oligonucleosome-like structure and clearance. Clin Exp Immunol 1992;90: 56 62. Chan KCA, Zhang J, Chan ATC, Lei KIK, Leung SF, Chan LYS, et al. Molecular characterization of circulating EBV DNA in the plasma of nasopharyngeal carcinoma and lymphoma patients. Cancer Res 2003;63:2028 32. Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977;37:646 50. Shapiro B, Chakrabarty M, Cohn EM, Leon SA. Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer 1983;51:2116 20. Maebo A. Plasma DNA level as a tumor marker in primary lung cancer. Nihon Kyobu Shikkan Gakkai Zasshi 1990;28:1085 91. Fournie GJ, Courtin JP, Laval F, Chale JJ, Pourrat JP, Pujazon MC, et al. Plasma DNA as a marker of cancerous cell death. Investigations in patients suffering from lung cancer and in nude mice bearing human tumours. Cancer Lett 1995;91:221 7. Kuroi K, Tanaka C, Toi M. Plasma nucleosome levels in node-negative breast cancer patients. Breast Cancer 1999;6:361 4. Holdenrieder S, Stieber P, Foerg T, Kuehl M, Schulz L, Busch M, et al. Apoptosis in serum of patients with solid tumours. Anticancer Res 1999;19:2721 4. Lo YMD, Chan LY, Lo KW, Leung SF, Zhang J, Chan AT, et al. Quantitative analysis of cell-free Epstein Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res 1999; 59:1188 91. Kuroi K, Tanaka C, Toi M. Clinical significance of plasma nucleosomes levels in cancer patients. Int J Oncol 2001;19:143 8. Holdenrieder S, Stieber P, Bodenmueller H, Busch M, Fertig G, Fuerst H, et al. Nucleosomes in serum of patients with benign and malignant diseases. Int J Cancer 2001;95:114 20. Holdenrieder S, Stieber P, Bodenmueller H, Fertig G, Fuerst H, Schmeller N, et al. Nucleosomes in serum as a marker for cell death. Clin Chem Lab Med 2001;39:596 605.

617

[151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159] [160]

[161] [162]

[163]

[164] [165]

[166]

[167] Sozzi G, Conte D, Mariani L, Lo Vullo S, Roz L, Lombardo C, et al. Analysis of circulating tumor DNA in plasma at diagnosis and during follow-up of lung cancer patients. Cancer Res 2001; 61:4675 8. [168] Ziegler A, Zangemeister-Wittke U, Stahel RA. Circulating DNA: a new diagnostic gold mine? Cancer Treat Rev 2002;28:255 71. [169] Trejo-Becerril C, Perez-Cardenas E, Trevino-Cuevas H, TajaChayeb L, Garcia-Lopez P, Segura-Pacheco B, et al. Circulating nucleosomes and response to chemotherapy: an in vitro, in vivo and clinical study on cervical cancer patients. Int J Cancer 2003;104:663 8. [170] Anker P, Mulcahy H, Stroun M. Circulating nucleic acids in plasma and serum as a noninvasive investigation for cancer: time for largescale clinical studies? Int J Cancer 2003;103:149 52. [171] Sozzi G, Conte D, Leon M, Ciricione R, Roz L, Ratcliffe C, et al. Quantification of free circulating DNA as a diagnostic marker in lung cancer. J Clin Oncol 2003;21:3902 8. [172] Anker P, Stroun M. Tumor-related alterations in circulating DNA, potential for diagnosis, prognosis and detection of minimal residual disease. Leukemia 2001;15:289 91. [173] Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 2003;349:2042 54. [174] Sidransky D. Emerging molecular markers of cancer. Nat Rev Cancer 2002;2:210 9. [175] Andriani F, Conte D, Mastrangelo T, Leon M, Ratcliffe C, Roz L, et al. Detecting lung cancer in plasma with the use of multiple genetic markers. Int J Cancer 2004;108:91 6. [176] Lo YMD, Chan AT, Chan LY, Leung SF, Lam CW, Huang DP, et al. Molecular prognostication of nasopharyngeal carcinoma by quantitative analysis of circulating Epstein Barr virus DNA. Cancer Res 2000;60:6878 81. [177] Silva JM, Silva J, Sanchez A, Garcia JM, Dominguez G, Provencio M, et al. Tumor DNA in plasma at diagnosis of breast cancer patients is a valuable predictor of disease-free survival. Clin Cancer Res 2002;8:3761 6. [178] Lei KI, Chan LY, Chan WY, Johnson PJ, Lo YMD. Diagnostic and prognostic implications of circulating cell-free Epstein Barr virus DNA in natural killer/T-cell lymphoma. Clin Cancer Res 2002; 8:29 34. [179] Holdenrieder S, Stieber P, Bodenmueller H, Busch M, v Pawel J, Schalhorn A, et al. Circulating nucleosomes in serum. Ann N Y Acad Sci 2001;945:93 102. [180] Lo YMD, Chan LY, Chan AT, Leung SF, Lo KW, Zhang J, et al. Quantitative and temporal correlation between circulating cell-free Epstein Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res 1999;59:5452 5. [181] Lo YMD, Leung SF, Chan LY, Chan AT, Lo KW, Johnson PJ, et al. Kinetics of plasma Epstein Barr virus DNA during radiation therapy for nasopharyngeal carcinoma. Cancer Res 2000;60:2351 5. [182] Holdenrieder S, Stieber P, v Pawel J, Raith H, Nagel D, Feldmann K, Seidel D. Circulating nucleosomes predict the response to chemotherapy in patients with advanced non small cell lung cancer. Clin Cancer Res 2004 [submitted for publication].

You might also like