Occult tumor cells in lymph nodes as a predictor for tumor relapse in pancreatic adenocarcinoma

Share Embed


Descrição do Produto

Langenbecks Arch Surg (2008) 393:359–365 DOI 10.1007/s00423-007-0215-0

ORIGINAL ARTICLE

Occult tumor cells in lymph nodes as a predictor for tumor relapse in pancreatic adenocarcinoma Peter Scheunemann & Nikolas H. Stoecklein & Alexander Rehders & Minu Bidde & Sylvia Metz & Matthias Peiper & Claus F. Eisenberger & Jan Schulte am Esch & Wolfram T. Knoefel & Stefan B. Hosch

Received: 27 February 2007 / Accepted: 16 July 2007 / Published online: 18 August 2007 # Springer-Verlag 2007

Abstract Background and aims Occurrence of tumor relapse is frequent in patients with pancreatic cancer despite the absence of residual tumor detectable at primary surgery and in histopathological examination. Therefore, it has to be assumed that current tumor staging procedures fail to identify minimal amounts of disseminated tumor cells, which might be precursors of subsequent metastatic relapse. The aim of this study was to assess the prognostic impact of minimal tumor cell spread detected in lymph nodes classified as “tumor-free” in routine histopathologic evaluation. Materials and methods A total of 154 “tumor-free” lymph nodes from 59 patients with pancreatic cancer who underwent intentionally curative tumor resection were examined by immunohistochemistry for disseminated tumor cells. Results Fifty (32.5%) of the “tumor-free” lymph nodes obtained from 36 (61%) patients displayed disseminated tumor cells. Multivariate survival analysis revealed that the presence of disseminated tumor cells in “tumor-free” lymph nodes is an independent prognostic factor for both a significantly reduced relapse-free survival (p=0.03) and overall survival (p=0.02). P. Scheunemann (*) : N. H. Stoecklein : A. Rehders : M. Peiper : C. F. Eisenberger : J. Schulte am Esch : W. T. Knoefel : S. B. Hosch Department of General Surgery, Heinrich-Heine University, University Hospital Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany e-mail: [email protected] M. Bidde : S. Metz Department of General Surgery, University Hospital Hamburg–Eppendorf, Hamburg, Germany

Conclusions The frequent occurrence and prognostic impact of immunohistochemically identifiable disseminated tumor cells in lymph nodes of patients with operable pancreatic cancer supports the need for a refined staging system of excised lymph nodes, which should include immunohistochemical examination. Keywords Micrometastasis . Lymph nodes . Pancreatic cancer . Minimal residual disease . Immunohistochemistry

Introduction Ductal adenocarinoma of the pancreas are highly aggressive malignancies with an unchanged poor prognosis and is currently the forth-leading cause of cancer-related death in Western countries [1]. Although postoperative mortality has declined and resection rates have improved considerably, the reported 5-year survival rates still range only between 5 and 36% [2–5]. Early metastatic relapse after the complete resection of an apparently localized primary tumor indicates an occult tumor cell dissemination or micrometastatic disease that cannot be detected by current staging procedures. Therefore, more sensitive immunohistochemical assays have been developed that are based on the detection of epithelial cell- or tumor-associated markers and are able to detect minimal disseminated tumor cells in lymph nodes classified as “tumor-free” by conventional histopathologic examination [6–19]. An increasing number of published studies indicates that these early tumor cell deposits in “tumor-free” lymph nodes appeared to be strong and independent predictors of tumor relapse in several carcinoma entities [7, 9–11, 14–19]. However, in contrast to

360

carcinoma of the esophagus, stomach, or breast, little is known about the clinical significance of occult-disseminated tumor cells in lymph nodes in pancreatic cancer. In pancreatic cancer, the strongest predictor for longterm survival after complete tumor resection (R0) is the absence of nodal tumor involvement verified by routine histopathology (pN0). Nevertheless, most of these patients die from tumor relapse within 3 years after surgery. In an initial study on 18 patients with R0-resected pancreatic adenocarcinoma, we demonstrated that immunohistochemically detected tumor cells in “tumor-free” lymph nodes were of strong prognostic impact [10]. To verify these results, we immunohistochemically analyzed “tumor-free” lymph nodes of 41 additional patients with resectable pancreatic carcinoma and evaluated the prognostic value of our findings on in total 59 patients, including the first 18 patients with actualized follow-up.

Materials and methods Patients This study was approved by the ethics committee of the chamber of physicians in Hamburg. Informed consent was obtained from all the patients before their inclusion in the study. Lymph nodes were prospectively collected from 59 consecutive patients with pancreatic ductal adenocarcinoma without overt metastases (M0) who underwent intentional curative pancreatic resection at the University Hospital Hamburg during November 1992 and June 2002. In each patient, reconstruction was performed with a pancreatojejunostomy according to Whipple–Kausch [20]. None of the 59 study patients received neoadjuvant or adjuvant therapy. The clinical and histopathological data of the 59 patients analyzed in this study are summarized in Table 1. For survival analyses, patients without R0 tumor resection (n=4) and patients who died during the hospital stay (n=4) were excluded. In addition, six patients were lost to follow-up. The median postoperative observation period of the remaining 45 patients was 11 months (range, 3 to 122 months). Lymph node preparation and immunohistochemical detection of tumor cells During the systematic lymphadenectomy, lymph nodes that were macroscopically inconspicuous were randomly collected from five different node groups: the superior node group including the superior pancreatic nodes, the inferior node group including the inferior pancreatic and superior mesenteric nodes, the anterior node group including the anterior pancreaticoduodenal nodes, the posterior node group including the posterior pancreaticoduodenal, celiac

Langenbecks Arch Surg (2008) 393:359–365 Table 1 Characteristics of patients and tumors Variable

Number of patients

Number (%) of patients with EpCCAM+ cells in “tumor-free” lymph nodesa

p value

All patients Male Female Median age 62 years (33–83) 18 Monthsb Distant metastasis-free All patients’ 4/15 events (%) 33 Monthsb a b

(40)

Patients with EpCAM+ cells

p valuea

24/30 (80)

0.013

11 (46.7)

23/30 (76.7)

0.018

,8

Cum. overall survival

Survival

EpCAM-pos. (n=30)

12/30 (40)

0.43

0,0 0

20

40

60

80

100

120

140

14 Months after operation

(26.7)

16/30 (53.3)

0.037

Fig. 2 Overall survival of patients with and without EpCAM-positive cells in lymph nodes

11

Log-rank test Median value

ences were not statistically significant for both the relapsefree survival (p=0.15) and overall survival (p=0.07). Discussion Despite improvement of surgical treatment, prognosis of patients with pancreatic carcinoma has not changed remarkably over the last decades [3, 22–25]. Lymph node metastasis identified on routine histopathological examination is one of the most important prognostic factors in patients with pancreatic cancer [22]. Our study provides evidence that isolated tumor cells or small tumor cell clusters detected in lymph nodes using a sensitive immu-

nohistochemical assay are independent prognostic factors for relapse-free and overall survival in pancreatic cancer. Remarkably, the presence of EpCAM-positive cells in lymph nodes predicted for distant metastatic relapse but not for local recurrence. This finding suggests that these cells might be indicators for a generalized occult metastatic disease with a high load of residual tumor cells, rather than indicators of an advanced locoregional disease that might be controlled by radical local tumor resection with tumorfree resection margins. We are aware that the number of analyzed patients is too low to draw firm conclusions. In addition, the lacking significance of EpCAM-positive cells in lymph nodes in the subgroup survival analysis is an argument for cautious interpretation. However, the observation that both pN0 as well as pN1 patients with EpCAMpositive cells in their lymph nodes showed an clearly 1,0

Cum. metastatic relapse-free survival

1,0

,8

Cum. relapse-free survival

EpCAM-neg. (n=15)

,4

,2

7 (40)

p=0.013

,6

n=0.018

,6

EpCAM-neg. (n=15)

,4

,2

EpCAM-pos. (n=30)

,9

,8

,7

,6

p=0.037 EpCAM-neg. (n=15)

,5

,4

EpCAM-pos. (n=30)

,3

0,0 0

20

40

60

80

100

120

140

Months after operation

Fig. 1 Relapse-free survival of patients with and without EpCAMpositive cells in lymph nodes

0

20

40

60

80

100

120

140

Months after operation

Fig. 3 Distant metastases-free survival of patients with and without EpCAM-positive cells in lymph nodes

Langenbecks Arch Surg (2008) 393:359–365

363 1,0

Table 4 Subgroup analysis of the prognostic impact of EpCAMpositive cells in lymph nodes according to the histopathological lymph node classification

Overall pN0 patients’ events (%) Monthsb pN1 patient’ events (%) Monthsb Relapse-free pN0 patient’ events (%) Monthsb pN1 patient’ events (%) Monthsb a b

Patients without EpCAM+ cells

Patients with EpCAM+ cells

3/10 (30)

11/15 (80)

>24 3/5 (60)

18 13/15 (86.7)

p valuea

0.071

0.132

Cum. overall survival

,8

Survival

EpCAM-neg. (n=10)

,6

p=0.071

,4

EpCAM-pos. (n=15)

,2

33

10

4/10 (40)

10/15 (66.7)

0.150

>18 3/5 (60)

11 13/15 (86.7)

0.071

33

6

0

20

40

60

80

100

120

140

Months after operation

Log-rank test Median value

reduced postoperative outcome compared to patients without such cells raises our hopes that these cells are truly relevant prognostic parameters. Furthermore, our findings are consistent to that observed in a recent study analyzing a total of 148 “tumor-free” lymph nodes of 48 pN0-pancreatic cancer patients [26] by immunohistochemistry with mAb Ber-EP4. In this study, EpCAM-positive cells could be detected in a similar rate of analyzed patients (58%) and the presence of these cells was also of independent prognostic significance on relapse-free survival and overall survival. Occult lymph node “micrometastasis” is a well-known phenomenon, and several reports have been published concerning detection of these cells by extensive serial sectioning and conventionally haematoxylin and eosin (HE) staining of apparently “tumor-free” lymph nodes, mainly in Table 5 Results of Cox regression survival analysis

Relapse-free survival pT3–4 vs pTis–2 pN1 vs pN0 Grade III vs grade I–II Immunohistochemistry (EpCAM+ vs EpCAM−) Overall survival pT3–4 vs pTis–2 pN1 vs pN0 Grade III vs grade I–II Immunohistochemistry (EpCAM+ vs EpCAM−)

0,0

Relative risk

95% confidence interval

p value

0.6 2.8 1.1 2.6

0.232–1.361 1.176–6.772 0.467–2.529 1.115–6.283

0.2 0.02 0.9 0.03

0.7 3.0 1.7 3.2

0.273–1.624 1.301–7.029 0.713–3.904 1.233–8.216

0.4 0.01 0.2 0.02

Fig. 4 Overall survival of patients staged as pN0 with and without EpCAM-positive cells in lymph nodes

patients with breast cancer [27–29]. One study, initiated by the International Breast Cancer Study Group in 1990 [30], could demonstrate that extensive serial sectioning of lymph nodes previously judged as “tumor-free” in routine histopathology was suitable to detect “micrometastases” in 83 of 921 (9%) “node-negative” breast carcinoma patients by conventional HE staining. Furthermore, detection of these nodal “micrometastases” with subsequent conversion from “node-negative” to “node-positive” status was correlated with a significantly poorer prognosis. However, this timeconsuming method analyzing extensive numbers of consecutive sections is not feasible as a routine procedure. Sensitive immunohistochemical methods using mAb for identification of minimal disseminated tumor cells in lymph nodes might be a more reasonable approach. Most studies examining pathohistologically “tumor-free” lymph nodes in carcinoma patients have used antibodies against a broad spectrum of cytokeratins (CK) [6–8, 19, 31–34]. However, a previously reported irregular CK expression (e.g., CK18) in normal lymphatic reticulum cells [35] can lead to falsepositive results. Therefore, we used the mAb Ber-EP4, which is more specific for epithelial cell detection in lymph nodes [9, 11, 17] because it does not react with mesenchymal tissue [17, 21]. However, general limitations of immunohistochemical assays are the somewhat subjective criteria of the observer who distinguishes between real tumor cells, stained normal cells (if present), and laboratory artifacts (false-positive events). In addition, evaluation of two sections each cut from three different lymph node levels as performed in our study might be possibly not representative for the entire lymph node and could therefore introduce further sampling errors. The positive correlation between our immunohistochemical findings and the prognostic data suggests, however, that our approach provides clinical relevant information.

364

As an alternative to immunohistochemistry, nucleic acidbased detection of minimal disseminated tumor cells has recently received considerable attention [12, 15, 16, 18, 36– 39], which allows hypothetically the evaluation of an entire lymph node. However, the specificity of these ultrasensitive nucleic acid-based molecular assays is limited by the lack of any morphological correlate, the heterogeneity of genetic alterations, and the absence of suitable detection markers on messenger ribonucleic acid or deoxyribonucleic acid levels exclusively found in pancreatic cancer cells [40–43]. As a consequence, the immunohistochemical or nucleic acidbased detection of disseminated tumor cells in lymph nodes has not been incorporated in the current International Union Against Cancer tumor staging nomenclature. Thus, there is an urgent need for standardization of the current protocols before “micrometastatic” lymph node staging can be implemented into clinical practice.

Conclusion Our data indicate that immunohistochemical assessment of lymph nodes can be used to refine the staging system for pancreatic cancer and help to identify patients at a high risk of tumor recurrence, which cannot be cured by surgery alone. The detection of the earliest manifestations of tumor cell dissemination is an extremely promising approach, which might enable us to identify suitable candidates for adjuvant treatment strategies (e.g., with humanized therapeutic anti-EpCAM antibodies). Furthermore, in the past 10 years, new immunologic and molecular analytic procedures have been developed to further characterize early disseminated cancer cells [9, 44–47]. Improved methods for analysis on the single-cell level [48–52] may help to identify new target molecules on disseminated tumor cells, thus optimizing the therapeutic options for the individual patients with pancreatic cancer. Immunohistochemical analysis of lymph nodes should therefore be included as a stratification criteria to these trials.

References 1. Smeenk HG, Tran TC, Erdmann J, van Eijck CH, Jeekel J (2005) Survival after surgical management of pancreatic adenocarcinoma: does curative and radical surgery truly exist? Langenbecks Arch Surg 390:94–103 2. Peters JH, Carey LC (1991) Historical review of pancreaticoduodenectomy. Am J Surg 161:219–225 3. Reinders ME, Allema JH, van Gulik TM, Karsten TM, de Wit LT, Verbeek PC, Rauws EJ, Gouma DJ (1995) Outcome of microscopically nonradical, subtotal pancreaticoduodenectomy (Whipple’s resection) for treatment of pancreatic head tumors. World J Surg 19:410–414 discussion 414–415

Langenbecks Arch Surg (2008) 393:359–365 4. Safioleas MC, Moulakakis KG (2004) Pancreatic cancer today. Hepatogastroenterology 51:862–868 5. Schmidt CM, Powell ES, Yiannoutsos CT, Howard TJ, Wiebke EA, Wiesenauer CA, Baumgardner JA, Cummings OW, Jacobson LE, Broadie TA, Canal DF, Goulet RJ Jr, Curie EA, Cardenes H, Watkins JM, Loehrer PJ, Lillemoe KD, Madura JA (2004) Pancreaticoduodenectomy: a 20-year experience in 516 patients. Arch Surg 139:718–725 discussion 725–727 6. Bonavina L, Ferrero S, Midolo V, Buffa R, Cesana B, Peracchia A (1999) Lymph node micrometastases in patients with adenocarcinoma of the esophagogastric junction. J Gastrointest Surg 3:468–476 7. Calaluce R, Miedema BW, Yesus YW (1998) Micrometastasis in colorectal carcinoma: a review. J Surg Oncol 67:194–202 8. Greenson JK, Isenhart CE, Rice R, Mojzisik C, Houchens D, Martin EW Jr (1994) Identification of occult micrometastases in pericolic lymph nodes of Duke’s B colorectal cancer patients using monoclonal antibodies against cytokeratin and CC49. Correlation with long-term survival. Cancer 73:563–569 9. Hosch S, Kraus J, Scheunemann P, Izbicki JR, Schneider C, Schumacher U, Witter K, Speicher MR, Pantel K (2000) Malignant potential and cytogenetic characteristics of occult disseminated tumor cells in esophageal cancer. Cancer Res 60:6836–6840 10. Hosch SB, Knoefel WT, Metz S, Stoecklein N, Niendorf A, Broelsch CE, Izbicki JR (1997) Early lymphatic tumor cell dissemination in pancreatic cancer: frequency and prognostic significance. Pancreas 15:154–159 11. Izbicki JR, Hosch SB, Pichlmeier U, Rehders A, Busch C, Niendorf A, Passlick B, Broelsch CE, Pantel K (1997) Prognostic value of immunohistochemically identifiable tumor cells in lymph nodes of patients with completely resected esophageal cancer. N Engl J Med 337:1188–1194 12. Kano M, Shimada Y, Kaganoi J, Sakurai T, Li Z, Sato F, Watanabe G, Imamura M (2000) Detection of lymph node metastasis of oesophageal cancer by RT-nested PCR for SCC antigen gene mRNA. Br J Cancer 82:429–435 13. Komukai S, Nishimaki T, Watanabe H, Ajioka Y, Suzuki T, Hatakeyama K (2000) Significance of immunohistochemically demonstrated micrometastases to lymph nodes in esophageal cancer with histologically negative nodes. Surgery 127:40–46 14. Kubuschok B, Passlick B, Izbicki JR, Thetter O, Pantel K (1999) Disseminated tumor cells in lymph nodes as a determinant for survival in surgically resected non-small-cell lung cancer. J Clin Oncol 17:19–24 15. Liefers GJ, Cleton-Jansen AM, van de Velde CJ, Hermans J, van Krieken JH, Cornelisse CJ, Tollenaar RA (1998) Micrometastases and survival in stage II colorectal cancer. N Engl J Med 339:223–228 16. Lindblom A (1998) Improved tumor staging in colorectal cancer. N Engl J Med 339:264–265 17. Passlick B, Izbicki JR, Kubuschok B, Nathrath W, Thetter O, Pichlmeier U, Schweiberer L, Riethmuller G, Pantel K (1994) Immunohistochemical assessment of individual tumor cells in lymph nodes of patients with non-small-cell lung cancer. J Clin Oncol 12:1827–1832 18. Sidransky D (1997) Nucleic acid-based methods for the detection of cancer. Science 278:1054–1059 19. Trojani M, de Mascarel I, Coindre JM, Bonichon F (1987) Micrometastases to axillary lymph nodes from invasive lobular carcinoma of breast: detection by immunohistochemistry and prognostic significance. Br J Cancer 56:838–839 20. Jones L, Russell C, Mosca F, Boggi U, Sutton R, Slavin J, Hartley M, Neoptolemos JP (1999) Standard Kausch–Whipple pancreatoduodenectomy. Dig Surg 16:297–304 21. Latza U, Niedobitek G, Schwarting R, Nekarda H, Stein H (1990) Ber-EP4: new monoclonal antibody which distinguishes epithelia from mesothelial. J Clin Pathol 43:213–219

Langenbecks Arch Surg (2008) 393:359–365 22. Gall FP, Kessler H, Hermanek P (1991) Surgical treatment of ductal pancreatic carcinoma. Eur J Surg Oncol 17:173–181 23. Gudjonsson B (1995) Carcinoma of the pancreas: critical analysis of costs, results of resections, and the need for standardized reporting. J Am Coll Surg 181:483–503 24. Klempnauer J, Ridder GJ, Pichlmayr R (1995) Prognostic factors after resection of ampullary carcinoma: multivariate survival analysis in comparison with ductal cancer of the pancreatic head. Br J Surg 82:1686–1691 25. Roder JD, Stein HJ, Huttl W, Siewert JR (1992) Pylorus-preserving versus standard pancreatico-duodenectomy: an analysis of 110 pancreatic and periampullary carcinomas. Br J Surg 79:152–155 26. Bogoevski D, Yekebas EF, Schurr P, Kaifi JT, Kutup A, Erbersdobler A, Pantel K, Izbicki JR (2004) Mode of spread in the early phase of lymphatic metastasis in pancreatic ductal adenocarcinoma: prognostic significance of nodal microinvolvement. Ann Surg 240:993–1000 discussion 1000–1001 27. Friedman S, Bertin F, Mouriesse H, Benchabat A, Genin J, Sarrazin D, Contesso G (1988) Importance of tumor cells in axillary node sinus margins (‘clandestine’ metastases) discovered by serial sectioning in operable breast carcinoma. Acta Oncol 27:483–487 28. Fisher ER, Swamidoss S, Lee CH, Rockette H, Redmond C, Fisher B (1978) Detection and significance of occult axillary node metastases in patients with invasive breast cancer. Cancer 42:2025–2031 29. Pickren JW (1961) Significance of occult metastases. A study of breast cancer. Cancer 14:1266–1271 30. International (Ludwig) Breast Cancer Study Group (1990) Prognostic importance of occult axillary lymph node micrometastases from breast cancers. Lancet 335:1565–1568 31. Glickman JN, Torres C, Wang HH, Turner JR, Shahsafaei A, Richards WG, Sugarbaker DJ, Odze RD (1999) The prognostic significance of lymph node micrometastasis in patients with esophageal carcinoma. Cancer 85:769–778 32. Matsumoto M, Natsugoe S, Nakashima S, Sakamoto F, Okumura H, Sakita H, Baba M, Takao S, Aikou T (2000) Clinical significance of lymph node micrometastasis of pN0 esophageal squamous cell carcinoma. Cancer Lett 153:189–197 33. Natsugoe S, Mueller J, Stein HJ, Feith M, Hofler H, Siewert JR (1998) Micrometastasis and tumor cell microinvolvement of lymph nodes from esophageal squamous cell carcinoma: frequency, associated tumor characteristics, and impact on prognosis. Cancer 83:858–866 34. Pantel K, Cote RJ, Fodstad O (1999) Detection and clinical importance of micrometastatic disease. J Natl Cancer Inst 91:1113–1124 35. Franke WW, Moll R (1987) Cytoskeletal components of lymphoid organs. I. Synthesis of cytokeratins 8 and 18 and desmin in subpopulations of extrafollicular reticulum cells of human lymph nodes, tonsils, and spleen. Differentiation 36:145–163 36. Gerhard M, Juhl H, Kalthoff H, Schreiber HW, Wagener C, Neumaier M (1994) Specific detection of carcinoembryonic antigen-expressing tumor cells in bone marrow aspirates by polymerase chain reaction. J Clin Oncol 12:725–729 37. Gudemann CJ, Weitz J, Kienle P, Lacroix J, Wiesel MJ, Soder M, Benner A, Staehler G, Doeberitz MV (2000) Detection of hematogenous micrometastasis in patients with transitional cell carcinoma. J Urol 164:532–536 38. Kijima F, Natsugoe S, Takao S, Aridome K, Baba M, Yoshifumi M, Eizuru Y, Aikou T (2000) Detection and clinical significance of lymph node micrometastasis determined by reverse transcriptionpolymerase chain reaction in patients with esophageal carcinoma. Oncology 58:38–44

365 39. Mori M, Mimori K, Inoue H, Barnard GF, Tsuji K, Nanbara S, Ueo H, Akiyoshi T (1995) Detection of cancer micrometastases in lymph nodes by reverse transcriptase-polymerase chain reaction. Cancer Res 55:3417–3420 40. Bostick PJ, Chatterjee S, Chi DD, Huynh KT, Giuliano AE, Cote R, Hoon DS (1998) Limitations of specific reverse-transcriptase polymerase chain reaction markers in the detection of metastases in the lymph nodes and blood of breast cancer patients. J Clin Oncol 16:2632–2640 41. Jung R, Petersen K, Kruger W, Wolf M, Wagener C, Zander A, Neumaier M (1999) Detection of micrometastasis by cytokeratin 20 RT-PCR is limited due to stable background transcription in granulocytes. Br J Cancer 81:870–873 42. Ko Y, Klinz M, Totzke G, Gouni-Berthold I, Sachinidis A, Vetter H (1998) Limitations of the reverse transcription-polymerase chain reaction method for the detection of carcinoembryonic antigen-positive tumor cells in peripheral blood. Clin Cancer Res 4:2141–2146 43. Zippelius A, Kufer P, Honold G, Kollermann MW, Oberneder R, Schlimok G, Riethmuller G, Pantel K (1997) Limitations of reverse-transcriptase polymerase chain reaction analyses for detection of micrometastatic epithelial cancer cells in bone marrow. J Clin Oncol 15:2701–2708 44. Pantel K, Angstwurm M, Kutter D, Braun S, Karg O, Izbicki JR, Werner U, Schlimok G, Riethmuller G (1992) Downregulation and neo-expression of cell surface antigens on individual micrometastatic carcinoma cells. In: Rabes H, Peters PE, Munk K (eds) Metastasis: basic research and its clinical applications. Contributions to oncology, vol. 44. Karger, Basel, pp 283–293 45. Pantel K, Hosch SB (2001) Molecular profiling of micrometastatic cancer cells. Ann Surg Oncol 8:18S–21S 46. Pantel K, Otte M (2001) Occult micrometastasis: enrichment, identification and characterization of single disseminated tumour cells. Semin Cancer Biol 11:327–337 47. Pantel K, Schlimok G, Braun S, Kutter D, Lindemann F, Schaller G, Funke I, Izbicki JR, Riethmuller G (1993) Differential expression of proliferation-associated molecules in individual micrometastatic carcinoma cells. J Natl Cancer Inst 85:1419–1424 48. Dietmaier W, Hartmann A, Wallinger S, Heinmoller E, Kerner T, Endl E, Jauch KW, Hofstadter F, Ruschoff J (1999) Multiple mutation analyses in single tumor cells with improved whole genome amplification. Am J Pathol 154:83–95 49. Klein CA, Blankenstein TJ, Schmidt-Kittler O, Petronio M, Polzer B, Stoecklein NH, Riethmuller G (2002) Genetic heterogeneity of single disseminated tumour cells in minimal residual cancer. Lancet 360:683–689 50. Klein CA, Schmidt-Kittler O, Schardt JA, Pantel K, Speicher MR, Riethmuller G (1999) Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells. Proc Natl Acad Sci USA 96:4494–4499 51. Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M, Ahr A, Blankenstein TJ, Kaufmann M, Diebold J, Arnholdt H, Muller P, Bischoff J, Harich D, Schlimok G, Riethmuller G, Eils R, Klein CA (2003) From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci USA 100:7737–7742 52. Schutze K, Lahr G (1998) Identification of expressed genes by laser-mediated manipulation of single cells. Nat Biotechnol 16:737–742

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.