9
Abstract Metastatic spread is a major factor in the prog- nosis of cancer patients. Early detection and eradication of circulating tumour cells prior to the development of metastases could help to improve the outcome of patients after tumour resection. Disseminated tumour cells have been detected in different compartments of the body us- ing cytological and immunostaining methods and, more recently, using different molecular biological techniques. The most frequently studied body compartments are the bone marrow, peritoneal cavity, blood and lymph nodes, but other body fluids such as urine, bile, pancreatic juice and sputum have also been analysed. At all of these sites, tumour cells have been detected. However, the specificity and sensitivity of the methods and their prognostic impact are still being debated. This review discusses the accura- cy of the detection methods and the prognostic value of detecting disseminated tumour cells in the bone marrow, blood and peritoneal lavage of patients with colorectal, gastric and pancreatic carcinomas. Keywords Disseminated tumour cells · Immunocytochemical detection · Molecular detection · Prognosis · Gastrointestinal carcinoma · Pancreatic carcinoma Introduction Early tumour diagnosis, improved surgical treatment and multimodal therapeutic concepts have helped to reduce the mortality of patients with gastrointestinal and pancreatic carcinoma. Nevertheless, the survival rates of these patients are still highly unsatisfactory. The prognosis depends on local tumour recurrence and/or the occurrence of distant metastases. Staging systems, such as the TNM classifica- tion, have been developed to help estimate the probability of further tumour progression after treatment. Detection of disseminated tumour cells at a distance from the primary tumour before the development of metastases or local tu- mour recurrence may help to refine the TNM system [22]. In recent years, numerous immunocytochemical and molecular studies have shown that micrometastases and disseminated tumour cells can be demonstrated not only in lymph nodes that had appeared tumour free on conven- tional histology [37], but also in such body compartments as the bone marrow, peritoneal cavity and blood [15, 16, 25, 54, 55, 59]. Not only have increasing numbers of compartments been analysed; studies have been extended to other malignancies that seldom demonstrate metastatic spread to the bone, such as oesophageal, pancreas or pri- mary hepatic tumours [32]. From these data, it has been concluded that conventional staging most likely underes- timates the true tumour stage. However, because the tech- niques used to identify disseminated tumour cells have not yet been standardised, the detection of disseminated tumour cells was not included in the most recent TNM classification. It does, however, recommend documenting any relevant findings. If isolated tumour cells are detect- ed immunocytochemically in lymph nodes, the UICC suggests adding an “i+” in parentheses along with the N stage. If molecular findings indicate the presence of tu- mour cells, a “mol+” should be added [23]. Disseminated tumour cells are detected by means of immunocytochemical or molecular methods designed to recognise certain tumour-associated antigens and/or cell lineage-specific markers, such as cytokeratins (CKs) [15, 16, 43, 54]. These markers are not tumour specific, how- ever, but may also be expressed by non-neoplastic cells. If both the immunocytochemical and the molecular ana- lyses are to be valid, it is therefore of the utmost impor- tance to avoid generating “false-positive” data, i.e. re- cognising non-neoplastic cells instead of tumour cells. However, a high degree of sensitivity must be reached to reduce “false-negative” results. Another crucial issue is the biological significance of the disseminated tumour cells that are detected. Are these I. Vogel ( ) · H. Kalthoff Molecular Oncology Research Laboratory, Department of General and Thoracic Surgery, University of Kiel, Arnold-Heller-Strasse 7, 24105 Kiel, Germany e-mail: [email protected] Tel.: +49-431-5974481, Fax: +49-431-597-1939 Virchows Arch (2001) 439:109–117 DOI 10.1007/s004280100476 REVIEW ARTICLE Ilka Vogel · Holger Kalthoff Disseminated tumour cells Their detection and significance for prognosis of gastrointestinal and pancreatic carcinomas Received: 9 May 2001 / Accepted: 9 May 2001 / Published online: 7 July 2001 © Springer-Verlag 2001

Disseminated tumour cells

  • Upload
    holger

  • View
    222

  • Download
    5

Embed Size (px)

Citation preview

Page 1: Disseminated tumour cells

Abstract Metastatic spread is a major factor in the prog-nosis of cancer patients. Early detection and eradicationof circulating tumour cells prior to the development ofmetastases could help to improve the outcome of patientsafter tumour resection. Disseminated tumour cells havebeen detected in different compartments of the body us-ing cytological and immunostaining methods and, morerecently, using different molecular biological techniques.The most frequently studied body compartments are thebone marrow, peritoneal cavity, blood and lymph nodes,but other body fluids such as urine, bile, pancreatic juiceand sputum have also been analysed. At all of these sites,tumour cells have been detected. However, the specificityand sensitivity of the methods and their prognostic impactare still being debated. This review discusses the accura-cy of the detection methods and the prognostic value ofdetecting disseminated tumour cells in the bone marrow,blood and peritoneal lavage of patients with colorectal,gastric and pancreatic carcinomas.

Keywords Disseminated tumour cells ·Immunocytochemical detection · Molecular detection ·Prognosis · Gastrointestinal carcinoma · Pancreaticcarcinoma

Introduction

Early tumour diagnosis, improved surgical treatment andmultimodal therapeutic concepts have helped to reduce themortality of patients with gastrointestinal and pancreaticcarcinoma. Nevertheless, the survival rates of these patientsare still highly unsatisfactory. The prognosis depends onlocal tumour recurrence and/or the occurrence of distantmetastases. Staging systems, such as the TNM classifica-

tion, have been developed to help estimate the probabilityof further tumour progression after treatment. Detection ofdisseminated tumour cells at a distance from the primarytumour before the development of metastases or local tu-mour recurrence may help to refine the TNM system [22].

In recent years, numerous immunocytochemical andmolecular studies have shown that micrometastases anddisseminated tumour cells can be demonstrated not onlyin lymph nodes that had appeared tumour free on conven-tional histology [37], but also in such body compartmentsas the bone marrow, peritoneal cavity and blood [15, 16,25, 54, 55, 59]. Not only have increasing numbers ofcompartments been analysed; studies have been extendedto other malignancies that seldom demonstrate metastaticspread to the bone, such as oesophageal, pancreas or pri-mary hepatic tumours [32]. From these data, it has beenconcluded that conventional staging most likely underes-timates the true tumour stage. However, because the tech-niques used to identify disseminated tumour cells havenot yet been standardised, the detection of disseminatedtumour cells was not included in the most recent TNMclassification. It does, however, recommend documentingany relevant findings. If isolated tumour cells are detect-ed immunocytochemically in lymph nodes, the UICCsuggests adding an “i+” in parentheses along with the Nstage. If molecular findings indicate the presence of tu-mour cells, a “mol+” should be added [23].

Disseminated tumour cells are detected by means ofimmunocytochemical or molecular methods designed torecognise certain tumour-associated antigens and/or celllineage-specific markers, such as cytokeratins (CKs) [15,16, 43, 54]. These markers are not tumour specific, how-ever, but may also be expressed by non-neoplastic cells.If both the immunocytochemical and the molecular ana-lyses are to be valid, it is therefore of the utmost impor-tance to avoid generating “false-positive” data, i.e. re-cognising non-neoplastic cells instead of tumour cells.However, a high degree of sensitivity must be reached toreduce “false-negative” results.

Another crucial issue is the biological significance ofthe disseminated tumour cells that are detected. Are these

I. Vogel (✉ ) · H. KalthoffMolecular Oncology Research Laboratory,Department of General and Thoracic Surgery, University of Kiel,Arnold-Heller-Strasse 7, 24105 Kiel, Germanye-mail: [email protected].: +49-431-5974481, Fax: +49-431-597-1939

Virchows Arch (2001) 439:109–117DOI 10.1007/s004280100476

R E V I E W A RT I C L E

Ilka Vogel · Holger Kalthoff

Disseminated tumour cellsTheir detection and significance for prognosis of gastrointestinaland pancreatic carcinomas

Received: 9 May 2001 / Accepted: 9 May 2001 / Published online: 7 July 2001© Springer-Verlag 2001

Page 2: Disseminated tumour cells

cells able to proliferate and give rise to metastases by in-teracting with the environment, or are they in a state ofdormancy, arrested in the G0 phase [58] and waiting toproliferate at a later point in time? An important answerto these questions will probably come from studies onthe prognostic relevance of disseminated tumour cellsdetected in the bone marrow, peritoneal cavity and blood.This article reviews these issues by discussing the vari-ous detection methods used so far and the prognostic rel-evance of the results gained from these studies.

Detection methods

Sensitivity and specificity

The detection rate of disseminated tumour cells is affected by vari-ous factors, including number of analysed cells, collection andtreatment of the sample, cell separation protocol, selection of anti-bodies and primers, and evaluation techniques.

The number of cells investigated clearly determines the detec-tion rate in a given patient, although the sensitivity of the variousassays is high, ranging from one tumour cell in 106 to one in 107

mononuclear cells [27, 52, 66]. It has also been demonstrated thatmultiple samples taken from different sites result in higher detec-tion rates than single samples [53].

All methods rely on the recognition of antigens or gene tran-scripts that are expressed only by tumour cells and not by the sur-rounding cells. It is often difficult to find such a specific antigen,but, even if there is a marker supposed to be expressed by the tu-mour cells, antigen expression in the disseminated tumour cellscan vary greatly [33], because antigens expressed by the primarytumour may have been downregulated or lost. For some markers[prostate-specific antigen (PSA), mucins], modulation due to hor-monal influence has been demonstrated [28]. This problem mightbe overcome by the parallel use of multiple markers.

False-positive results can also occur regardless of the methodsapplied [immunostaining or reverse-transcription polymerase chainreaction (RT-PCR)], if epithelial markers are used, due to contami-nation with skin cells or release of epithelial cells in benign prolif-erative diseases [24, 52, 72]. The isolation of mononuclear cells[34] may lead to loss of tumour cells and result in false-negativedata due to small sample volumes.

Immunostaining approaches

Several studies have clearly demonstrated that immunocytochem-istry is superior to conventional cytology at detecting isolated tu-mour cells in the bone marrow [42, 61]. Immunostaining methodsfor detecting disseminated epithelial tumour cells in mesenchymalcompartments, such as the bone marrow, employ antibodies direct-ed against a variety of epithelial cell markers. Pan-specific anti-bodies against CK seem to have a higher specificity than antibod-ies directed against the epithelial mucin family [14, 61] (Fig. 1).However, in addition to increased sensitivity, a higher number offalse-positive results due to non-specific labelling have been ob-served. The main reasons were (i) expression of some epithelialantigens by a few haematological cells [5], (ii) endogenous alka-line phosphatase in cases where anti-alkaline phosphatase (AP-AAP) complexes were used [3] and (iii) phagocytosed stainablematerial in granulocytes [48].

Molecular biological approaches

Chromosomal abnormalities can be detected in haematologicalmalignancies using breakpoint-specific PCR tests. In solid tu-mours, this molecular approach is not applicable, because the chro-

mosomal abnormalities are much more heterogeneous and com-plex. However, in solid tumours, it is possible to identify dissemi-nated tumour cells by detecting RNA specific to the tissue of ori-gin of these cells [6]. The method of choice is the RT-PCR, whichhas been further refined by many investigators so that it is applica-ble to the detection of disseminated tumour cells.

When disseminated tumour cells are analysed using RNA-based assays for epithelial cells, it has to be assumed that cells ofnon-haematopoietic origin normally do not circulate in the pe-ripheral blood or bone marrow. The target RNA should not be ex-pressed by haematopoietic cells, but exclusively by the dissemi-nated tumour cells that are to be identified. Alternatively, a two-step procedure of enrichment and/or derichment methods needs tobe included. Studies of target RNAs for tumours of gastrointesti-nal origin have mainly focused on tissue-specific markers, suchas CKs (CK19, CK20; Fig. 2) and mucins, or on tumour-associat-ed antigens such as carcinoembryonic antigen (CEA) [15, 16, 25,56, 59]. The PCR assays can detect a single target cell in up to100 million irrelevant cells in spiking experiments [10, 66]. Di-rect comparison of molecular methods using immunostaining ap-proaches showed that the PCR method is potentially 100 timesmore sensitive [64]. Moreover, by means of the PCR technique,target nucleic acids can be amplified by a factor of 106–109 with-in less than 1 h. The high sensitivity and specificity of these as-says can be limited, however, by DNA contamination [35], “ille-gitimate” expression (transcription of any gene in any cell type)of epithelial or tumour-specific antigens in haematopoietic cells[5, 11, 29], upregulation of target mRNA expression by inflam-mation or hormonal induction of gene expression [28], the exis-tence of pseudogenes, which can lead to false-positive results inhealthy individuals [28, 29, 47] or in patients with benign diseas-es [16], or the degradation of RNA during the various steps of theassay protocol [31].

The detection rates achieved for venous blood samples usingRT-PCR are lower than corresponding analyses in bone marrowsamples. Possible explanations for this phenomenon are (i) quali-tative and quantitative differences between cells in the bone mar-row and venous blood, (ii) differences in survival or immune es-cape possibilities in different compartments and (iii) methodolog-ical aspects, such as loss of tumour cells due to masking withthrombocytes or clotting problems.

110

Fig. 1 Disseminated tumour cell in the bone marrow of a patientwith colorectal carcinoma. Using the alkaline phosphatase withanti-alkaline phosphatase (APAAP) technique, staining was per-formed with a cocktail of the following antibodies: C1-P83 [carci-noembryonic (CEA)], Ca 19–9, KL-1 (cytokeratin); counterstain-ing with haemalum

Page 3: Disseminated tumour cells

Characterisation of disseminated tumour cells

In addition to detecting disseminated tumour cells, it is necessaryto characterise them in more detail, especially because of thera-peutic implications. Features that are of interest in this respect arethe immunogenic potential of the cells, their proliferation activity,and their expression of growth factors and adhesion molecules.The identification of these features on disseminated tumour cellsrequires double labelling of the cells, which has been performedusing various techniques. Studies using these methods revealeddownregulation of major histocompatibility complex (MHC)class-I antigen in colorectal and gastric carcinomas [51, 62], neo-expression of intercellular adhesion molecule (ICAM)-1 and mel-anoma cell adhesion molecule (MUC-18) and downregulation ofdesmosomal protein [53], reduced expression of Ki67 (prolifera-tion-associated antigen) in gastrointestinal carcinoma [51], re-duced expression of PCNA (proliferating cell nuclear antigen) inprostatic cancer [45], expression of growth factor receptors(transferrin, c-erbB2) in metastatic breast cancer [51], expressionof E-cadherin (adhesion molecule) in breast and gastric cancer[13] and expression of the urokinase plasminogen activator (uPA)receptor [1].

New methods have been developed in recent years by combin-ing either immunostaining methods or immunofluorescence within situ hybridisation (FISH) [30, 39, 44] or single-cell PCR tech-niques [33]. With these techniques, it was possible to prove themalignancy of cells by detecting identical chromosomal abnormal-ities to those in the primary tumour [45]. These techniques havealso been used to demonstrate the heterogeneity of disseminatedtumour cells found at different sites in a single patient [33]. Cul-tures of disseminated cells have been successfully established invitro, increasing the cell number available for further phenotypicand genotypic analysis [49, 58].

Results of the clinical studies and prognostic impact

Colorectal carcinoma

Immunocytochemical analyses of bone marrow samplesfrom patients with colorectal carcinoma were initiallyperformed by Schlimok et al. [61] with a monoclonal an-tibody called CK2, which specifically reacts with CK18

[9]. Further studies with the same antibody revealed de-tection rates in the bone marrow between 16% and 32%.Other studies using combinations of antibodies founddisseminated cells in the bone marrow in up to 74% ofthe cases [4], whereas PCR tests yielded positive resultsin 31–84% of the patients [26]. Specificity was evaluatedin all studies by analysing samples from patients withoutany evidence of carcinoma or from healthy subjects.Most of the studies had a false-positive rate of below10% [27, 52, 65, 78]. Using multivariate analysis, Linde-mann [38] showed that detection of disseminated tumourcells in the bone marrow with the monoclonal antibodyCK2 is an independent prognostic factor for survival(Table 1). Leinung et al. [36] used the pan-specific CKantibody A45-B/B3, which detects a common epitope ona variety of CK types, including CK8, CK18 and CK19[68], and also found that the detection of disseminatedtumour cells had a significant influence on survival. Oth-er authors combined different antibodies directed againstCKs and/or tumour-associated antigens, but none ofthem performed multivariate analyses to demonstrateprognostic relevance [4, 27, 65]. To date, none of thestudies analysing peritoneal washings has demonstratedthat the detection of disseminated tumour cells in pa-tients with colorectal carcinoma is an independent prog-nostic marker [4, 27, 65] (Table 1).

Most molecular biological studies have been per-formed on bone marrow and/or venous blood from pa-tients with colorectal carcinoma. CK genes, especiallythe CK19 and CK20 genes, have been used as markergenes [7, 10, 12, 17, 59, 66, 75, 77, 78]. The gene of theCEA was also tested in several studies [26, 59, 69].Since haematopoietic cells can “illegitimately” expresstumour-associated antigens or epithelial cell-specific an-tigens [17, 79] and pseudogenes may give rise to PCRproducts identical to those of marker genes, the RT-PCRassays resulted in a large number of false-positive sig-nals [17]. So far, the CK20 gene seems to be the bestmarker, although some false-positive results have beenobserved with this also [75, 76]. Our analyses of 295 pa-tients with curative resections showed for the first timethat the detection of disseminated tumour cells in thebone marrow and/or venous blood is an independentprognostic factor for overall survival in patients with co-lorectal carcinoma [75] (Table 2). It was clearly demon-strated that tumour cells are present in the bone marrowand blood in colorectal carcinoma patients. Detection

111

Fig. 2 Cytokeratin 20 (CK20)-specific nested reverse-transcrip-tion polymerase chain reaction (RT-PCR) after serial dilution ofRNA from the pancreatic tumour cell line A818–4 (cells per milli-litre). Separation of samples was done by agarose-gel electropho-resis and staining with ethidium bromide. The corresponding con-trol reaction was performed as GAPDH RT-PCR (359-bp product)using a duplex approach. ex RT-PCR product (788 bp) using theexternal primers, in nested PCR product (485 bp), –RT amplifica-tion without reverse transcription, K water control, M molecularweight marker

Page 4: Disseminated tumour cells

rates were higher in patients whose tumours were in anadvanced stage than in those with early tumour stages,although disseminated tumour cells were already detect-able in up to 20% of patients with early tumour stages[75]. Only a few other studies have also focused on the

prognostic relevance of tumour cell detection (Table 2).They confirmed that disseminated cells are of prognosticvalue for these patients, but the results need to be estab-lished by multicenter clinical trials under standardisedconditions.

112

Table 1 Prognostic relevance of immunocytochemical studies on bone marrow (BM) and/or peritoneal lavage (PL) obtained from pa-tients with colorectal carcinoma prior to tumour resection

+: relevant to prognosis, –: not relevant to prognosis, ∅ : not done; univar.: univariate analysis; multivar.: multivariate analysis

Table 2 Prognostic relevance of molecular biological studies (PCR) on bone marrow (BM) and/or peripheral blood (BL) obtained frompatients with colorectal carcinoma prior to tumour resection

+: relevant to prognosis, –: not relevant to prognosis, ∅ : not done; univar.: univariate analysis; multivar.: multivariate analysis

Page 5: Disseminated tumour cells

Gastric carcinoma

In immunocytological studies of patients with gastriccarcinoma, the same monoclonal antibodies have gener-ally been used to detect disseminated tumour cells in thebone marrow and the peritoneal cavity as in colorectalcarcinoma patients. Detection rates in the bone marrowranged between 25% and 82% [1, 4, 27]. Overall, thisseems to be slightly higher than in colorectal carcinoma.Even higher detection rates (32–72%) were observed inthe peritoneal cavity [4, 27, 65]. The explanation for thismight be the higher rate of peritoneal carcinomatosis atthe time of the primary operation in gastric carcinomacompared with colorectal carcinoma.

One study, published by Jauch in 1996 [24], includeda multivariate analysis. It demonstrated that the presenceof three or more cells in the bone marrow was of prog-nostic significance for disease-free survival, but only inpatients with T1/2 tumours (UICC classification; Ta-ble 3). Heiss et al. [21] showed that tumour cell dissemi-nation in the bone marrow was an additional prognosticfactor in patients who are in early tumour stages (UICC

I/II), are lymph node-negative (N0) and express PAI-1(plasminogen activator inhibitor type 1).

Molecular analyses have been performed with CEA,CK19 and CK20 as markers [7, 25, 59, 67]. To date, theonly studies to demonstrate in univariate analyses a sig-nificant difference in overall survival for patients withtumour cells in the bone marrow and blood were doneby our group (Table 4). These studies included 22 bonemarrow samples and 58 venous blood samples [67]. It israther striking that the detection rate in the bone marrowwas low relative to immunocytochemical studies, al-though advanced stages were included [67]. The likelyexplanation for this is that CK20 is not expressed at theprotein level in every gastric carcinoma [43]. It is there-fore necessary to evaluate other markers that might bemore consistently expressed in gastric carcinomas.

Pancreatic carcinoma

Still fewer studies have dealt with patients with pancreat-ic ductal carcinoma. This may be a reflection of the poor

113

Table 3 Prognostic relevance of immunocytochemical studies on bone marrow (BM) and/or peritoneal lavage (PL) obtained from pa-tients with gastric carcinoma prior to tumour resection

+: relevant to prognosis, –: not relevant to prognosis, ∅ : not done; univar.: univariate analysis; multivar.: multivariate analysis

Table 4 Prognostic relevance of molecular biological studies (PCR) on bone marrow (BM) and/or peripheral blood (BL) obtained frompatients with gastric carcinoma prior to tumour resection

+: relevant to prognosis, –: not relevant to prognosis, ∅ : not done; univar.: univariate analysis; multivar.: multivariate analysis

Page 6: Disseminated tumour cells

prognosis for these patients and the lower incidence ofthe disease than with colorectal carcinoma. Immunocyto-chemical analyses of bone marrow samples have beenperformed using cocktails of antibodies, mostly includ-ing Ca 19–9 as a tumour-associated antigen [60, 70, 71,74]. In view of the higher number of advanced-stage pa-tients in the respective series, detection rates of dissemi-nated cells of almost 40–60% have been found [60]. Inunivariate analyses, most of the studies demonstratedshorter survival in patients in whom tumour cells weredetected in the bone marrow, while our investigationswere the only ones to demonstrate that detection of cellsin the peritoneal lavage has an impact on patient progno-sis [74] (Table 5).

Molecular analyses have been performed in only asmall number of cases [2, 50, 57, 66, 67]. Neither detec-tion in the bone marrow nor detection in the blood hasbeen evaluated for prognostic impact. The only study sofar to test mutant- Ki-ras (that is known to occur fre-quently in ductal carcinomas but also in hyperplasticduct lesions of the pancreas [40]) as a marker in theblood was on a series of ten patients [50]. Bilchik et al.[2] reported a PCR combination of three markers: MET(hepatocyte growth factor receptor gene c-met), Gal-NAc-T (β1,4-N-acetyl-galactosaminyl-transferase) andβ-hCG (β-human chorionic gonadotropin) in an analysisof 33 patients. Their detection rate was 100% in stage-IV patients and 87.5% in stage-II and -III patients, if allthree markers were combined. Obviously, these markerswill need to be evaluated in a larger series.

Conclusion and perspectives

Our evaluation of the studies on colorectal, gastric andpancreatic ductal carcinomas indicates that detection ofdisseminated tumour cells in different compartments maylead to more accurate tumour staging. Further studies will

be required, however, to verify the independent prognos-tic impact in each of the compartments analysed so far.Such studies should try to evaluate methodological as-pects, such as methods for enriching mononuclear or tu-mour cells, and develop highly specific and sensitive de-tection systems (including methods, marker and primerchoices). Before these methods can be incorporated intoclinical routine, they need to be standardised.

Further studies should focus, in particular, on (semi)-quantitative RT-PCR, which additionally allows the am-plification rate to be standardised. PCR reactions usingmultiple markers may overcome tumour cell heterogene-ity and false-positive results and would therefore increasethe sensitivity and specificity. The enrichment of tumourcells, e.g. by magnetic beads, could solve the problem offalse-positive results by reducing the background noise.Initial analyses using this method have been very promis-ing [10, 46].

If future studies confirm the results published so far,patients with proven disseminated tumour cells whohave no other clinical or radiological evidence of diseaseafter resection of the primary tumour should receive ad-juvant therapy, because conventional staging underesti-mates the lymphatic and haematogenous spread of thetumour. As recurrences are sometimes seen after a periodof years, it is likely that disseminated cells can persist ina dormant state for prolonged periods. There is some evi-dence that these cells cannot be reached by means ofconventional chemotherapy but might be better targetedby antibody-based adjuvant cancer therapies [41, 73].

One problem that has to be considered is the heteroge-neity of solid tumours, since it reduces the likelihood thatall disseminated cells will be removed. An individualisedcharacterisation of the tumour cells might be one solu-tion, but this does not appear practicable in daily routine.The use of a cocktail of antibodies is more promising. Fur-ther analyses that consider methodological aspects, cellcharacterisation, the prognostic impact and new adjuvant

114

Table 5 Prognostic relevance of immunocytochemical studies on bone marrow (BM) and/or peritoneal lavage (PL) obtained from pa-tients with ductal pancreatic carcinoma prior to tumour resection

+: relevant to prognosis, –: not relevant to prognosis, ∅ : not done; univar.: univariate analysis; multivar.: multivariate analysis

Page 7: Disseminated tumour cells

therapeutic approaches are necessary and will certainlylead to improvements in the treatment of gastrointestinaland pancreatic carcinoma in the near future.

Acknowledgements The authors thank Prof. Klöppel for criticalreading the manuscript. They are also thankful to Mrs. Kay Degeand Heinke Brütting for English editing and typing the manu-script.

References

1. Allgayer H, Heiss MM, Riesenberg R, Grutzner KU, TarabichiA, Babic R, Schildberg FW (1997) Urokinase plasminogen ac-tivator receptor (uPA-R): one potential characteristic of meta-static phenotypes in minimal residual disease. Cancer Res 57:1394–1399

2. Bilchik A, Miyashiro M, Kelly M, Kuo C, Fujiwara Y, Naka-mori S, Monden M, Hoon DS (2000) Molecular detection ofmetastatic pancreatic carcinoma cells using a multimarker re-verse transcriptase-polymerase chain reaction assay. Cancer 88:103–104

3. Borgen E, Beiske K, Trachsel S, Nesland JM, Kvalheim G,Herstad TK, Schlichting E, Quist H, Naume B (1998) Immuno-cytochemical detection of isolated epithelial cells in bone mar-row: non-specific staining and contribution by plasma cells di-rectly reactive to alkaline phosphatase. J Pathol 185:427–434

4. Broll R, Lembcke K, Stock C, Zingler M, Duchrow M, Schim-melpenning H, Strik M, Müller G, Kujath P, Bruch HP (1996)Tumorzelldissemination in das Knochenmark und in die Peri-tonealhöhle. Langenbecks Arch Chir 381: 51–58

5. Brugger W, Bühring HJ, Grünebach F, Vogel W, Kraul S,Müller R, Brümmendorf TH, Ziegler BL, Rappold I, BrossartP, Scheding S, Kranz L (1999) Expression of MUC-1 epitopeson normal bone marrow: implications for the detection of mi-crometastatic tumour cells. J Clin Oncol 17:1535–1544

6. Burchill SA, Bradbury MF, Pittman K, Southgate J, Smith B,Selby P (1995) Detection of epithelial cancer cells in peripher-al blood by reverse transcriptase-polymerase chain reaction.Br J Cancer 71:278–281

7. Burchill SA, Lewis IJ, Selby P (1999) Improved methods us-ing the reverse transcriptase-polymerase chain reaction to de-tect tumour cells. Br J Cancer 79:971–977

8. Cohen AM, Garin-Chesa P, Hanson M, Weyhrauch K, KemenyN, Fong Y, Paty P, Welt S, Old L (1998) In vitro detection ofoccult bone marrow metastases in patients with colorectal can-cer hepatic metastases. Dis Colon Rectum 41:1112–1115

9. Debus E, Weber K, Osborn M (1982) Monoclonal cytokeratinantibodies that distinguish simple from stratified squamous epi-thelia: characterization on human tissue. EMBO J 1:1641–1647

10. Denis MG, Lipart C, Leborgne J, LeHur PA, Galmiche JP, De-nis M, Ruud E, Truchaud A, Lustenberger P (1997) Detectionof disseminated tumor cells in peripheral blood of colorectalcancer patients. Int J Cancer 74:540–544

11. Desol G, Gatter KC, Stein H, Erber WN, Pulford KAF, ZinnekB, Mason DY (1984) Human lymphoid cells express epithelialmembrane antigen. Implications for diagnosis of human neo-plasms. Lancet 11:1123–1129

12. Funaki NO, Tanaka J, Sugiyama T, Ohshio G, Nonaka A,Yotsumoto F, Furutani M, Imamura M (2000) Perioperativequantitative analysis of cytokeratin 20 mRNA in peripheralvenous blood of patients with colorectal adenocarcinoma. On-col Rep 7:21–76

13. Funke I, Fries S, Rolle M, Heiss MM, Untch M, Bohmert H,Schildberg W, Jauch KW (1996) Comparative analyses ofbone marrow micrometastases in breast and gastric cancer. IntJ Cancer 65:755–761

14. Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, Duhig T,Peat N, Burchell J, Pemberton L, Lalini EN, Wuilson D (1996)Molecular cloning and expression of human tumor-associatedpolymorphic epithelial mucin. J Biol Chem 265:15286–15293

15. Ghossein RA, Bhattacharya S, Rosain J (1999) Molecular de-tection of micrometastases and circulating tumor cells in solidtumors. Clin Cancer Res 5:1950–1960

16. Goeminne JC, Guillaume T, Symann M (2000) Pitfalls in thedetection of disseminated non-hematological tumor cells. AnnOncol 11:785–792

17. Gunn J, McCall JL, Yun K, Wright PA (1996) Detection of mi-crometastases in colorectal cancer patients by K19 and K20reverse-transcription polymerase chain reaction. Lab Invest 75:611–616

18. Hardingham JE, Kotasek D, Sage RE, Eaton MC, Pascoe VH,Dobrovic A (1995) Detection of circulating tumor cells in co-lorectal cancer by immunobead-PCR is a sensitive prognosticmarker for relapse of disease. Mol Med 1:789–794

19. Hardingham JE, Hewett PJ, Sage RE, Finch JL, Nuttall JD,Kotasek D, Dobrovic A (2000) Molecular detection of blood-borne epithelial cells in colorectal cancer patients and in pa-tients with benign bowel disease. Int J Cancer 89:8–13

20. Heiss MM, Allgayer H, Gruetzner KU, Funke I, Babic R,Jauch KW, Schildberg FW (1995) Individual development anduPA-receptor expression of disseminated tumor cells in bonemarrow: a reference to early systemic disease in solid cancer.Nat Med 1:1035–1039

21. Heiss MM, Allgayer H, Gruetzner KU, Babic R, Jauch KW,Schildberg FW (1997) Clinical value of extended biologicstaging by bone marrow micrometastases and tumor-associat-ed proteases in gastric cancer. Ann Surg 226:736–745

22. Hermanek P, Sobin LH (eds) (1998) UICC TNM Classifica-tion of malignant tumors, 5th edn. Springer, Berlin HeidelbergNew York

23. Hermanek P, Hutter RVP, Sobin LH, Wittekind C (1999) Clas-sification of isolated tumor cells and micrometastasis. Cancer86:2668–2673

24. Jauch KW, Heiss MM, Gruetzner U, Funke I, Pantel K, BabicR, Eissner HJ, Riethmüller G, Schildberg FW (1996) Prognos-tic significance of bone marrow micrometastases in patientswith gastric cancer. J Clin Oncol 14:1810–1817

25. Johnson PW, Burchill SA, Selby PJ (1995) The molecular de-tection of circulating tumor cells. Br J Cancer 72:268–276

26. Jonas S, Windeatt S, O-Boateng A, Fordy C, Allen-Mersh TG(1996) Identification of carcinoembryonic antigen-producingcells circulating in the blood of patients with colorectal carci-noma by reverse transcriptase polymerase chain reaction. Gut39:717–721

27. Juhl H, Stritzel M, Wroblewski A, Henne-Bruns D, Kremer B,Schmiegel W, Neumaier M, Wagener C, Schreiber HW, Kalt-hoff H (1994) Immunocytological detection of micrometastat-ic cells: comparative evaluation of findings in the peritonealcavity and the bone marrow of gastric, colorectal and pancre-atic cancer patients. Int J Cancer 57:330–335

28. Jung R, Krüger W, Hosch S, Holweg M, Kröger N, GutensohnK, Wagener C, Neumaier M, Zander AR (1998) Specificity ofreverse transcriptase polymerase chain reaction assays de-signed for the detection of circulating cancer cells is influencedby cytokines in vivo and in vitro. Br J Cancer 78:1194–1198

29. Jung R, Petersen K, Krüger W, Wolf M, Wagener C, Zander A,Neumaier M (1999) Detection of micrometastasis by cytokera-tin 20 RT-PCR is limited due to stable background transcrip-tion in granulocytes. Br J Cancer 81:870–873

30. Kell MR, Winter DC, O´Sullivan GC, Shanahan F, RedmondHP (2000) Biological behaviour and clinical implications onmicrometastases. Br J Surg 87:1629–1639

31. Keilholz U, Willhauck M, Rimoldi D, Brasseur F, Dummer W,Rass K, de Vries T, Blaheta J, Voit C, Lethe B, Burchill S (1998)Reliability of reverse transcription-polymerase chain reaction(RT-PCR)-based assays for the detection of circulating tumourcells: a quality-assurance initiative of the EORTC MelanomaCooperative Group. Eur J Cancer 34:750–753

32. Kienle P, Weitz J, Klaes R, Koch M, Brenner A, Lehnert T,Herfarth C, von Knebel Doeberitz M (2000) Detection of iso-lated disseminated tumor cells in bone marrow and blood sam-ples of patients with hepatocellular carcinoma. Arch Surg 135:213–218

115

Page 8: Disseminated tumour cells

33. Klein CA, Schmidt-Kittler O, Sachardt JA, Pantel K, SpeicherMR, Riethmüller G (1999) Comparative genomic hybridization,loss of heterozygosity, and DNA sequence analysis of singlecells. Proc Natl Acad Sci U S A 96:4494–4499

34. Krüger W, Jung R, Kröger N, Gutensohn K, Fiedler W, Neu-maier M, Jänicke F, Wagener C, Zander AR (2000) Sensitivityof assays designed for the detection of disseminated epithelialtumor cells is influenced by cell separation methods. ClinChemistry 46:435–436

35. Kwok S, Higuchi R (1989) Avoiding false positives with PCR.Nature 339:237–238

36. Leinung S, Würl P, Weiss CL, Röder I, Schönfelder M (2000)Cytokeratin-positive cells in bone marrow in comparison withother prognostic factors in colon carcinoma. LangenbecksArch Surg 385:337–343

37. Liefers GJ, Cleton-Jansen AM, van der Velde CJ, Hermans J,van Krieken JH, Cornelisse CJ, Tollenaar RA (1998) Micro-metastases and survival in stage II colorectal cancer. NewEngl J Med 339:223–228

38. Lindemann F, Schlimok G, Dirschedl P, Witte J, RiethmüllerG (1992) Prognostic significance of micrometastatic tumorcells in bone marrow of colorectal cancer patients. Lancet 340:685–689

39. Litle VR, Warren RS, Moore D II, Pallavicini MG (1997) Mo-lecular cytogenetic analysis of cytokeratin 20-labeled cells inprimary tumors and bone marrow aspirates from colorectalcarcinoma patients. Cancer 79:1664–1670

40. Lüttges J, Reinecke-Lüthge A, Möllmann B, Menke MAOH,Clemens A, Klimpfinger M, Sipos B, Klöppel G (1999) Ductchanges and K-ras mutations in the disease free pancreas:analysis of type, age relation and spatial distribution. Vir-chows Arch 435:461–468

41. Maxwell-Amstrong CA, Durrant LG, Scholefield JH (1999)Immunotherapy for colorectal cancer. Am J Surg 177:344–348

42. Molino A, Colombatti M, Bonetti F, Zaradini M, Pasini F,Perini A, Pelosi G, Tridente G, Veneri D, Cetto GL (1991) Acomparative analysis of three different techniques for the de-tection of breast cancer cells in bone marrow. Cancer 67:1032–1036

43. Moll R, Löwe A, Laufer J, Franke WW (1992) Cytokeratin 20in human carcinomas. A new histodiagnostic marker detectedby monoclonal antibodies. Am J Pathol 140:427–447

44. Müller P, Weckermann D, Riethmüller G, Schlimock G (1996)Detection of genetic alterations in micometastatic cells in bonemarrow of cancer patients by fluorescence in situ hybridiza-tion. Cancer Genet Cytogenet 88:8–16

45. Müller P, Caroll P, Bowers E, Moore D II, Cher M, Presti J,Wessman M, Pallavincini M (1998) Low frequency epithelialcells in marrow aspirates from prostate cancer patients are cy-togenetically aberrant. Cancer 83:538–546

46. Naume B, Borgen E, Nesland JM, Beiske K, Gilen E, RenolenA, Ravnas G, Quist H, Karesen R, Kvalheim G (1998) In-creased sensitivity for detection of micrometastatases in bonemarrow/peripheral blood stem-cell products from breast-can-cer patients by negative immunomagnetic separation. Int JCancer 78:556–560

47. Neumaier M, Gerhard M, Wagener C (1995) Diagnosis of mi-crometastases by the amplification of tissue-specific genes.Gene 159:43–47

48. Nicholson AG, Graham AN, Pezzella F, Agneta G, GoldstrawP, Pastorino U (1997) Does the use of immunohistochemistryto identify micrometastases provide useful information in thestaging of node-negative non-small cell lung carcinomas?Lung Cancer 18:231–240

49. Noack F, Schmitt M, Bauer J, Helmecke D, Kruger W, Thor-ban S, Sandherr M, Kuhn W, Graeff H, Harbeck N (2000) Anew approach to phenotyping disseminated tumor cells: meth-odological advances and clinical implications. Int J Biol Mark-ers 15:100–104

50. Nomoto S, Nakao A, Kasai Y, Harada A, Nonami T, Takagi H(1996) Detection of ras gene mutations in perioperative pe-ripheral blood with pancreatic adenocarcinoma. Jpn J CancerRes 87:793–797

51. Pantel K, Schlimok G, Kutter D, Schaller G, Genz T, Wie-becke B, Backmann R, Funke I, Riethmüller G (1991) Fre-quent down-regulation of major histocompatibility class I anti-gen expression on individual micrometastatic carcinoma cells.Cancer Res 51:4712–4715

52. Pantel K, Schlimok G, Angstwurm M, Weckermann D, SchmausW, Gath H, Passlick B, Izbicki JR, Riethmüller G (1994) Meth-odological analysis of immunocytochemical screening for dis-seminated epithelial tumor cells in bone marrow. J Hematother3:165–173

53. Pantel K, Schlimock G, Angstwurm M, Passlick B, Izbicki JR,Johnson JP, Riethmüller G (1995) Early metastasis of humansolid tumors: expression of cell adhesion molecules. Ciba FoundSymp 189:157–170

54. Pantel K, Cote RJ, Fodstad Ø (1999) Detection and clinicalimportance of micrometastatic disease. J Natl Cancer Inst 91:1113–1124

55. Passlick B, Thetter O, Pantel K, Kubuschok B, Pichlmeier U,Schweiberer L, Izbicki JR (1995) Bedeutung der regionalenlymphatischen Tumorzelldisseminierung bei Patienten mit res-ektablem nicht-kleinzelligem Bronchialcarcinom. Chirurg 66:780–786

56. Pelkey TJ, Frierson HF Jr, Bruns DE (1996) Molecular andimmunological detection of circulating tumor cells and micro-metastases from solid tumors. Clin Chem 42:1369–1381

57. Piva MG, Navaglia F, Basso D, Fogar P, Roveroni G, Gallo N,Zambon C-F, Pedrazzoli S, Plebani M (2000) CEA mRNA iden-tification in peripheral blood is feasible for colorectal, but not forgastric or pancreatic cancer staging. Oncology 59:323–328

58. Putz E, Witter K, Offner S, Stosiek P, Zippelius A, Johnson J,Zahn R, Riethmüller G, Pantel K (1999) Phenotypic character-istics of cell lines derived from disseminated cancer cells inbone marrow of patients with solid epithelial tumors: estab-lishment of working models for human micrometastases. Can-cer Res 59:241–248

59. Raj GV, Moreno JG, Gomella LG (1998) Utilization of poly-merase chain reaction technology in the detection of solid tu-mors. Cancer 82:1419–1442

60. Roder JD, Thorban S, Pantel K, Siewert JR (1999) Microme-tastases in bone marrow: prognostic indicators for pancreaticcancer. World J Surg 23:888–891

61. Schlimok G, Funke I, Holzmann B, Göttlinger G, Schmidt G,Häuser H, Swierkot S, Warnecke HH, Schneider B, Koprow-ski H, Riethmüller G (1987) Micrometastatic cells in bonemarrow: in vitro detection with anitcytokeratin and in vivo la-beling with anti-17-1A monoclonal antibodies. Proc Natl AcadSci U S A 84:8672–8676

62. Schlimok G, Funke I, Bock B, Schweiberer B, Witte J, Riethmüller G (1990) Epithelial tumor cells in bone marrow ofpatients with colorectal cancer: immunocytochemical detec-tion, phenotypic characterization, and prognostic significance.J Clin Oncol 8:831–837

63. Schlimok G, Funke I, Pantel K, Strobel F, Lindemann F, WitteJ, Riethmüller G (1991) Micrometastatic tumor cells in bonemarrow of patients with gastric cancer: methodological as-pects of detection and prognostic significance. Eur J Cancer27:1461–1465

64. Schoenfeld A, Kruger KH, Gomm J, Sinnett HD, Gazet JC,Sacks N, Bender HG, Luqmani Y, Coombes RC (1997) Thedetection of micrometastases in the peripheral blood and bonemarrow of patients with breast cancer using immunohisto-chemistry and reverse transcriptase polymerase chain reactionfor keratin 19. Eur J Cancer 33:845–861

65. Schott A, Vogel I, Krueger U, Kalthoff H, Schreiber HW,Schmiegel W, Henne-Bruns D, Kremer B, Juhl H (1998) Iso-lated tumor cells are frequently detectable in the peritonealcavity of gastric and colorectal cancer patients and serve as anew prognostic marker. Ann Surg 227:372–379

66. Soeth E, Röder C, Juhl H, Krüger U, Kremer B, Kalthoff H(1996) The detection of disseminated tumor cells in bone mar-row from colorectal-cancer patients by a cytokeratin-20-spe-cific nested reverse-transcriptase-polymerase-chain reaction isrelated to the stage of disease. Int J Cancer 69:278–282

116

Page 9: Disseminated tumour cells

67. Soeth E, Vogel I, Röder C, Juhl H, Marxsen J, Krüger U, Hen-ne-Bruns D, Kremer B, Kalthoff H (1997) Comparative analysisof bone marrow and venous blood isolates from gastrointestinalcancer patients for the detection of disseminated tumor cells us-ing reverse transcription PCR. Cancer Res 57:3106–3110

68. Stosiek P, Kasper M, Karsten U, Goertchen R (1991) Detec-tion of cancer metastasis in regional lymph nodes: compara-tive histological and immunohistological investigations withthe broad-range anticytokeratin monoclonal antibody A45-B/B3. Neoplasma 38:43–47

69. Taniguchi T, Makino M, Suzuki K, Kaibara N (2000) Prognos-tic significance of reverse transcriptase-polymerase chain re-action measurement of carcinoembryonic antigen mRNA lev-els in tumor drainage blood and peripheral blood of patientswith colorectal carcinoma. Cancer 89:970–976

70. Thorban S, Roder JD, Pantel K, Siewert JR (1996) Epithelialtumor cells in bone marrow of patients with pancreatic carci-noma detected by immunocytochemical staining. Eur J Cancer32A:363–365

71. Thorban S, Roder JD, Siewert JR (1999) Detection of micro-metastasis in bone marrow of pancreatic cancer patients. AnnOncol 10[Suppl 4]:111–113

72. Traweek ST, Liu J, Battifora H (1993) Keratine gene expres-sion in non-epithelial tissues. Detection with polymerase chainreaction. Am J Pathol 142:1111–1118

73. Vermorken JB, Claessen AM, van Tinteren H, Gall HE, Ezin-ga R, Meijer S, Scheper RJ, Meijer CJ, Bloemena E, RansomJH, Hanna MG Jr, Pinedo HM (1999) Active specific immu-notherapy for stage II and stage III human colon cancer: a ran-domised trial. Lancet 353:345–350

74. Vogel I, Krüger U, Marxsen J, Soeth E, Kalthoff H, Henne-Bruns D, Kremer B, Juhl H (1999) Disseminated tumor cellsin pancreatic cancer patients detected by immunocytology: anew prognostic factor. Clin Cancer Res 5:593–599

75. Vogel I, Soeth E, Röder C, Kremer B, Henne-Bruns D, Kalt-hoff H (2000) Multivariate analysis reveals RT-PCR-detectedtumour cells in the blood and/or bone marrow of patients withcolorectal carcinoma as an independent prognostic factor. EurJ Clin Oncol 26:281

76. Wharton RQ, Jonas SK, Glover C, Khan ZA, Klokouzas A,Quinn H, Henry M, Allen-Mersh TG (1999) Increased detec-tion of circulating tumor cells in the blood of colorectal carci-noma patients using two reverse transcription assays and mul-tiple blood samples. Clin Cancer Res 5:4158–4163

77. Wyld DK, Selby P, Perren TJ, Jonas SK, Allen-Mersh TG,Wheeldon J, Burchill SA (1998) Detection of colorectal can-cer cells in peripheral blood by reverse-transcriptase poly-merase chain reaction for cytokeratin 20. Int J Cancer 79:288–293

78. Yamaguchi K, Takagi Y, Aoki S, Futamura M, Saji S (2000)Significant detection of circulating cancer cells in the blood byreverse transcriptase-polymerase chain reaction during colo-rectal cancer resection. Ann Surg 232:58–65

79. Zippelius A, Kufer P, Honold G, Köllermann MW, ObernederR, Schlimok G, Riethmüller G, Pantel K (1997) Limitations ofreverse transcriptase-polymerase chain reaction analyses fordetection of micrometastatic epithelial cancer cells in bonemarrow. J Clin Oncol 15:2701–2708

117