Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-19T17:39:59.763Z Has data issue: false hasContentIssue false

The role of disseminated and circulating tumour cells in breast cancer

Published online by Cambridge University Press:  17 February 2006

D. Auer
Affiliation:
Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria.
A. Schneitter
Affiliation:
Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria.
C. Marth
Affiliation:
Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria.
S. Braun
Affiliation:
Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria.

Abstract

Occult dissemination of tumour cells in patients with operable breast cancer is a crucial step in formation of metastasis, yet conventional tumour staging does not reveal it. To identify individual tumour cells that have successfully escaped from the primary tumour and invaded secondary organs, several research groups established sensitive immunocytochemical and molecular assays. Aside of the well documented prognostic impact of lymph node metastasis and micrometastases, respectively, bone marrow plays a prominent role as a determinant for haematogenous micrometastatic organ involvement. In the past decade, several groups have documented the independent prognostic impact of the presence of bone marrow micrometastases, which, in a recent pooled analysis of individual patient data from over 4000 breast cancer patients with Stage I–III disease, could be confirmed for the entire study population, and, in addition, provided data for challenging hypotheses to be tested in future adjuvant therapy trials of clinically relevant subgroups of breast cancer patients. Although the availability of a simple blood test would be highly desirable, no prognostically relevant data so far exists for early breast cancer patients. Options for the applicability of such approaches to detect disseminated (to bone marrow) and circulating tumour cells (in blood) are ample, both in clinical trial settings and for basic as well as translational research. In this overview we provide a brief summary of the prognostic role and the potential clinical utility of bone marrow micrometastases in breast cancer patients.

Type
Focus On
Copyright
2006 Cambridge University Press

Background

When Steven Paget published his theory of ‘seed and soil’ in 1889 [1], the idea of haematogenous tumour-cell dissemination was born. More than a century later, with the use of molecular tools, new clinical findings have resulted in explanations of hither-to unexplainable phenomena, such as that donor-derived cancer in recipient organ allografts [2] and viable single tumour cells in secondary organs were both the descendants of a known primary tumour [3] and the potential precursors of subsequent metastasis [4].

Currently, the genesis of overt metastasis in breast cancer is based on the idea that tumour cells dissociate from the primary cancer and gain access to circulation either directly into blood vessels or after transit in lymphatic channels. Thus, detection of such cells in patients with newly diagnosed solid tumours has been an appealing strategy to provide evidence of future metastasis [5].

Evidence of malignant nature

Overall, the existence of circulating tumour cells (CTC) and the settlement of these cells in secondary organs, such as liver, bone, and lungs, as disseminated metastatic tumour cells (DTC) is generally accepted. These cells are believed to be rare members among the cellular population of primary tumour cells [6]. This model, viewing CTC and DTC as rare and late events during primary tumour progression, has been challenged by recent expression profiling studies, in which a more ubiquitous ‘metastatic phenotype,’ that can be assessed by gene expression analysis [79].

On the other hand, most readers will not be familiar with this word analyses of numerical chromosomal changes and altered gene expression of single disseminated tumour cells demonstrated that the majority of DTCs have genetic aberrations compatible with malignancy, and therefore, are most likely direct descendants of the primary tumour, although the genetic changes generally were incongruent with the dominant genotype of the corresponding primary tumour [1012].

Prognostic and potential clinical role

The actual presence of tumour cells outside the primary tumour and in organs relevant for subsequent metastasis formation, such as bone and bone marrow, serve three purposes that could be clinically useful (1) as unambiguous evidence for an early occult spread of tumour cells; (2) as a relevant risk factor for subsequent metastases and, thus, a poor prognosis; and (3) as a marker for monitoring treatment efficacy. Finally, and perhaps as importantly in the long run, genotyping and phenotyping of CTC and DTC should provide detailed insight into the metastatic process and permit direct exploration of targeted treatment strategies [5].

Is detection of CTC or DTC prognostic in early-stage breast cancer? The currently available literature regarding the prognostic relevance of the presence of DTC in bone marrow is controversial, and without clear conclusions if viewed globally. However, a substantial number of studies do not meet essential criteria for quality assurance, adequate controls, and/or clinical trial design, and therefore should be excluded from the debate. To date, sufficient data are available from several large studies that unambiguously demonstrate the independent poor prognostic influence of DTC present in bone marrow on outcome in patients with Stage I, II, or III breast cancer [1319] add Cote et al. [20]; Wong et al. [21]. In a recent pooled analysis of individual patient data from over 4700 breast cancer patients with Stage I–III disease, we were able to confirm the independent prognostic impact for the entire study population, and, in addition, provided data for challenging hypotheses to be tested in future adjuvant therapy trials of clinically relevant subgroups of breast cancer patients [22].

Although a blood test specifically designed for patients with Stage I–III breast cancer would be highly desirable, preliminary data suggest that that findings on CTCs and DTCs in peripheral blood and bone marrow, respectively, do not provide congruent results. In contrast to DTC detection in bone marrow of patients with early-stage disease, CTC analysis appears to be less sensitive and less prognostic [23]. On the other hand, a recently reported, highly rigorous study clearly showed that, in breast cancer patients, with metastatic disease the number of CTCs permit prediction of progression-free and overall survival as well as response to treatment [24]. As a major finding of their study, the opportunity to predict response as early as 3–4 weeks after initiation of treatment reflects an important step towards individualized treatment decisions in patients with metastatic disease.

Methodological aspects and potential pit-falls

In contrast to the simplicity of the technology, the influence of confounding variables of the immunocytochemical assay on detection of bone marrow should not be underestimated [2527]. Thorough and critical evaluation of each process step of sample preparation, immunostaining, and analysis is required to avoid misinterpretation. Before unrestricted routine use of the technology, results of an ongoing process of methodological improvement have to be awaited. Thus, the immunocytochemical technology, which ultimately has turned out to be technically demanding, has induced implementation of seemingly easier molecular solutions, such as the reverse-transcriptase polymerase chain reaction technique [2831]. However, the same quality control issues that were raised for immunocytochemistry are pertinent for this technology as well, and there are concerns that many of the reported studies may overestimate the importance of the findings [3235]. As a hallmark and further essential requirement for upcoming studies using molecular techniques for DTC detection, comparison with the benchmark technique of DTC detection (i.e., immunocytochemistry with anticytokeratin antibodies) would be essential.

Conclusion

The prognostic value of DTC in bone marrow of breast cancer patients can be viewed as a statistically valid and clinically useful prognostic marker. Beyond mere prognostic estimation, and perhaps even more important, it may be assumed that presence of DTC can serve as a predictive marker. In order to individualize decision-making on adjuvant therapy and to find out the prognostic relevance of CTCs in comparison to DTCs, we need well designed, highly powered, prospective clinical trials using DTCs and CTCs as candidate surrogate markers for the various clinical settings currently under investigation question, such as secondary adjuvant endocrine treatment and dose-dense or otherwise intensified cytotoxic therapy.

References

Paget S. Distribution of secondary growths in cancer of the breast. Lancet 1889; 1: 571573.Google Scholar
Loh E, Couch FJ, Hendricksen C, et al. Development of donor-derived prostate cancer in a recipient following orthotopic heart transplantation. J Am Med Assoc 1997; 277: 133137.Google Scholar
Klein CA, Schmidt-Kittler O, Schardt JA, Pantel K, Speicher MR, Riethmüller G. Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells. Proc Natl Acad Sci USA 1999; 96: 44944499.Google Scholar
Braun S, Pantel K, Müller P, et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. New Engl J Med 2000; 342: 525533. [Erratum, New Engl J Med 2000; 343: 308.]Google Scholar
Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer 2004; 4: 448456.Google Scholar
Fidler IJ, Kripke ML. Metastasis results from preexisting variant cells within a malignant tumour. Science 1977; 197: 893895.Google Scholar
van't Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002; 415: 530536.Google Scholar
van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. New Engl J Med 2002; 347: 19992009.Google Scholar
Bernards R, Weinberg RA, et al. Metastasis genes: a progression puzzle. Nature 2002; 418: 823824.Google Scholar
Schmidt-Kittler O, Ragg T, Daskalakis A, et al. From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci USA 2003; 100: 77377742.Google Scholar
Klein CA, Blankenstein TJF, Schmidt-Kittler O, et al. Genetic heterogeneity of single disseminated tumour cells in minimal residual cancer. Lancet 2002; 360: 683689.Google Scholar
Klein CA, Seidl S, Petat-Dutter K, et al. Combined transcriptome and genome analysis of single micrometastatic cells. Nat Biotechnol 2002; 20: 392397.Google Scholar
Pierga J-Y, Bonneton C, Vincent-Salomon A, et al. Clinical significance of immunocytochemical detection of tumour cells using digital microscopy in peripheral blood and bone marrow of breast cancer patients. Clin Cancer Res 2004; 10: 13921400.Google Scholar
Wiedswang G, Borgen E, Karesen R, et al. Detection of isolated tumour cells in bone marrow is an independent prognostic factor in breast cancer. J Clin Oncol 2003; 21: 34693478.Google Scholar
Gebauer G, Fehm T, Merkle E, et al. Epithelial cells in bone marrow of breast cancer patients at time of primary surgery: clinical outcome during long-term follow up. J Clin Oncol 2001; 19: 36693674.Google Scholar
Gerber B, Krause A, Muller H, et al. Simultaneous immunohistochemical detection of tumour cells in lymph nodes and bone marrow aspirates in breast cancer and its correlation with other prognostic factors. J Clin Oncol 2001; 19: 960971.Google Scholar
Braun S, Pantel K, Müller P, et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II or III breast cancer. New Engl J Med 2000; 342: 525533.Google Scholar
Mansi JL, Gogas H, Bliss JM, et al. Outcome of primary breast cancer patients with micrometastases: a long-term follow-up. Lancet 1999; 354: 197202.Google Scholar
Diel IJ, Kaufmann M, Costa SD, et al. Micrometastatic breast cancer cells in bone marrow at primary surgery: prognostic value in comparison with nodal status. J Nat Cancer Inst 1996; 88: 16521658.Google Scholar
Cote RJ, Rosen PP, Lesser ML, Old LJ, Osborne MP. Prediction of early relapse in patients with operable breast cancer by detection of occult bone marrow micrometastases. J Clin Oncol 1991; 9: 17491756.Google Scholar
Wong GYC, Yu QQ, Osborne MP. Bone marrow micrometastasis is a significant predictor of long-term relapse-free survival for breast cancer by a non-proportional hazards model [Abstract]. Breast Cancer Res Treat 2003; 82(Suppl 1): S99.Google Scholar
Braun S, Vogl FD, Naume B, Janni W, Osborne MP, Coombes RC, et al. A pooled analysis of bone marrow micrometastasis in breast cancer. NEJM 2005; 353 (8): 793802.Google Scholar
Naume N, Wiedswang G, Borgen E, et al. Clinical significance of isolated tumour cells in peripheral blood in breast cancer patients three years after diagnosis: comparison between analysis of peripheral blood and bone marrow. [Abstract 9554]. Proc Am Soc Clin Oncol 2004; 23: 844.Google Scholar
Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumour cells, disease progression, and survival in metastatic breast cancer. New Engl J Med 2004; 351: 781791.Google Scholar
Borgen E, Naume B, Nesland JM, et al. Standardisation of the immunocytochemical detection of cancer cells in bone marrow and blood: establishment of objective criteria for the evaluation of immunostained cells: the European ISHAGE working group for standardization of tumor cell detection. Cytotherapy 1999; 1: 377388.Google Scholar
Braun S, Müller M, Hepp F, et al. Re: Micrometastatic breast cancer cells in bone marrow at primary surgery: prognostic value in comparison with nodal status. J Natl Cancer Inst 1998; 90: 10991101.Google Scholar
Pantel K, Schlimok G, Angstwurm M, et al. Methodological analysis of immunocytochemical screening for disseminated epithelial tumour cells in bone marrow. J Hematother 1994; 3: 165173.Google Scholar
Stathopoulou A, Vlachonikolis I, Mavroudis D, et al. Molecular detection of cytokeratin-19-positive cells in the peripheral blood of patients with operable breast cancer: evaluation of their prognostic significance. J Clin Oncol 2002; 20: 34043412.Google Scholar
Sidransky D. Nucleic acid-based methods for the detection of cancer. Science 1997; 278: 10541059.Google Scholar
Fields KK, Elfenbein GJ, Trudeau WL, et al. Clinical significance of bone marrow metastases as detected using polymerase chain reaction in patients with breast cancer undergoing high-dose chemotherapy and autologous bone marrow transplantation. J Clin Oncol 1996; 14: 18681876.Google Scholar
Datta YH, Adams PT, Drobyski WR, et al. Sensitive detection of occult breast cancer by the reverse-transcriptase polymerase chain reaction. J Clin Oncol 1994; 12: 475482.Google Scholar
Zippelius A, Lutterbuse R, Riethmuller G, et al. Analytical variables of reverse transcription-polymerase chain reaction-based detection of disseminated prostate cancer cells. Clin Cancer Res 2000; 6: 27412750.Google Scholar
Ruud P, Fodstad O, Hovig E. Identification of a novel CK-19 pseudo-gene that may interfere with reverse-transcriptase polymerase chain reaction assays used to detect micrometastatic tumour cells. Int J Cancer 1999; 80: 119125.Google Scholar
Bostick PJ, Chatterjee S, Chi DD, et al. 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 1998; 16: 26322640.Google Scholar
Zippelius A, Kufer P, Honold G, et al. Limitations of reverse-transcriptase polymerase chain reaction for detection of micrometastatic epithelial cancer cells in bone marrow. J Clin Oncol 1997; 15: 27012708.Google Scholar