Proliferative markers as prognostic and predictive tools in early breast cancer: where are we now?

M. Colozza1, E. Azambuja2, F. Cardoso2,3, C. Sotiriou2,3, D. Larsimont3,4 and M. J. Piccart2,3,*

1 S. C. Oncologia Medica, Azienda Ospedaliera, San Sisto 06132 Perugia, Italy; 2 Medical Oncology Clinic, 3 Translational Research Unit and 4 Department of Pathology, Jules Bordet Institute, Boulevard de Waterloo, 125, 1000 Brussels, Belgium

* Correspondence to: Dr M. Piccart, Jules Bordet Institute, Boulevard de Waterloo, 125, 1000 Brussels, Belgium. Tel: +32-2-541-32-06; Fax: +32-2-538-08-58; E-mail: martine.piccart{at}bordet.be


    Abstract
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
In the last few decades, proliferative markers have been broadly evaluated as prognostic and predictive factors for early stage breast cancer patients. Several papers evaluating one or more markers have been published, often with contradictory results. As a consequence, there is still uncertainty about the role of these proliferative markers. The present paper critically reviews the current knowledge about the following markers: thymidine labeling index, S phase fraction/flow cytometry, Ki 67, thymidine kinase (TK), cyclins E, cyclin D, the cyclin inhibitors p27 and p21, and topoisomerase II{alpha}. For each marker, the prognostic and predictive role was separately analyzed. Only papers published in English in peer-reviewed journals before June 2004 that include at least 100 evaluable patients were selected. In addition, the prognostic and predictive role of the proliferative markers had to be assessed through multivariate analyses. One hundred and thirty-two papers fulfilled these criteria and 159 516 patients were analyzed. Unfortunately, several methodological problems in the research to date prevent us from including any one of these proliferative markers among the standard prognostic and predictive factors. Early incorporation of translational research and new technologies with clinical trials are needed to prospectively validate biological markers and allow their use in clinical practice.

Key words: breast cancer, proliferative markers, prognostic markers, predictive markers


    Introduction
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Invasive breast carcinoma (BC) is the most frequent carcinoma and is the second cause of death from malignant disease among women in the western world. Significant improvements in disease-free survival (DFS) and overall survival (OS) have been obtained with the extensive use of adjuvant systemic therapies, which are largely empirically based. The possibility to have strong prognostic and/or predictive markers is of utmost importance for clinicians in order to identify patients at higher risk of relapse and to select the most appropriate systemic treatment for an individual patient. Prognostic factors are those that predict the risk of recurrence or of death from BC independently of treatment. Predictive factors are those that distinguish between patients who are more or less likely to respond to a given therapy. However, the distinction between the prognostic and predictive value of each marker is not straightforward. The retrospective nature of the great majority of these studies may jeopardize their results. Hundreds of papers evaluating several prognostic and predictive factors have been published in the last 30 years. However, the only validated prognostic factors are tumor (T) size, lymph node (N) status, hormone receptor (HR) status, histologic grade, and age [1Go]; and, as predictive factors: HR status and human epidermal growth factor receptor-2 (HER2) status [2Go] for endocrine and trastuzumab therapy, respectively.

Tumor cell proliferation has been widely investigated in BC for its association with neoplastic growth, progression, and metastatic potential; the present article is a review of the knowledge gathered on tumor cell proliferative markers in the past decade, with a critical assessment of their prognostic and/or predictive value.


    Materials and methods
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
A computerized literature search through Medline was performed, applying the words ‘BC proliferative prognostic markers’ and ‘BC proliferative predictive markers’ and each one of the following: thymidine labeling index, S phase fraction/flow cytometry, Ki 67, thymidine kinase (TK), cyclins E, cyclin D, cyclin inhibitors p27 and p21, and topoisomerase II{alpha}. Articles were also identified by back-referencing from original and relevant review papers. We chose not to include histological grade since it is an already accepted standard marker, and similarly, mitotic index, which is an important component of all histological grading systems and has recently been reviewed [3Go].

Selected for the present review were papers published in English in peer-reviewed journals before June 2004, which included at least 100 evaluable patients and in which the prognostic and predictive role of each marker was assessed through multivariate analyses. When more than one proliferative marker was evaluated, we referred to the most relevant one. The levels of evidence were provided according to the Tumor Marker Grading Utility System proposed by Hayes et al. [4Go] as shown in Table 1, and we arbitrarily decided to consider as large retrospective studies those including ≥200 patients. We cannot exclude the fact that some papers that did not find significant correlations for a certain proliferative markers, have never been published and, therefore, this could hamper our findings.


View this table:
[in this window]
[in a new window]
 
Table 1. Levels of evidence for grading clinical utility of tumor markers

 

    Measurement of cells in the S phase
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Cell kinetics can be evaluated by detecting cells undergoing DNA synthesis. It is possible either to measure the fraction of cells that incorporates labeled pyrimidine bases into newly synthesized DNA or to estimate the percentage of cells in the S phase of the cell cycle (S-phase fraction) by flow cytometry while simultaneously determining DNA content.


    3H-thymidine labeling index
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
3H-thymidine labeling index (TLI) was one of the first methods utilized to evaluate the proliferative activity of BC. The number of tumor cells undergoing DNA synthesis can be measured using in vivo or in vitro assays for 3H-thymidine uptake [5Go], which can be visualized by autoradiography. The TLI expresses the ratio between thymidine-labeled cells and the total number of tumor cells and has been shown to be reliable and stable over time [6Go]. Labeled pyrimidine bases other than [3H] thymidine, such as the halogenated analogue bromodeoxyuridine (BrdU), have also been used successfully [7Go, 8Go]. Of note, incorporated BrdU can be revealed by immunohistochemistry (IHC). The technique, however, has limitations that have hampered its acceptance as a standard method. In fact, fresh tumor tissue is needed and a complex and time-consuming radioactive assay or in vivo administration of labeled substances is required.

Prognostic role
Several papers, mostly retrospective and sometimes based on large series of BC patients, have reported a significant correlation between high TLI and poor clinical outcome in terms of relapse-free survival (RFS)/DFS, distant metastases-free survival (DMFS) and/or OS independently of N status, T size, histological grade, HR status and menopausal status [9Go–17Go]. However, even if the results of these studies have consistently shown the feasibility and the high reproducibility of TLI as a measure of the tumor proliferative activity [18Go, 19Go] it has never been accepted as a standard prognostic marker for the reasons described above.

Predictive role
Three studies have evaluated the predictive role of TLI. In a retrospective study, 285 out of 403 women with more than three N-positive BC randomized to receive, as adjuvant chemotherapy, alternating or sequential courses of doxorubicin and CMF (cyclophosphamide, methotrexate, 5-fluorouracil) were analyzed [20Go]. The median values and ranges of TLI for the two arms were superimposable. Patients with high TLI had a significantly worse 12-year RFS, DMFS and OS and benefited significantly more from the sequential administration of doxorubicin and CMF. In two multicenter prospective phase III adjuvant trials, N-negative BC patients with high TLI, regardless of HR status, were randomized to receive chemotherapy (CT) or no further systemic therapy. In the first trial [21Go], the adjuvant CT consisted of six courses of the ‘classic’ CMF regimen. At a median follow-up of 81 months, a statistically significant increase in DFS was obtained with CT. Similar results were reported in the other trial utilizing an anthracycline-based regimen [cyclophosphamide, epirubicin and 5-fluorouracil (FEC)] for six courses [22Go]. The analysis of relapse sites showed that CT reduced significantly all loco-regional relapses and led to a decrease in the number of distant metastases and contralateral tumors, although these differences were not significant. Despite the achievement of level 1 evidence by these two trials, TLI was not adopted in clinical practice given the technical problems reported above.


    Flow cytometry
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
Flow cytometric measurement of the nuclear DNA content of tumor cells provides simultaneous information about DNA ploidy and proliferative activity as represented by the S-phase fraction (SPF). This technique can be performed on fresh-frozen tumors or on paraffin-embedded tissue. Paraffin blocks provide the advantage of using archival tissue stored for many years, but there is a risk of finding more debris than in frozen or fresh tissue, which results in a relatively high proportion of tumors for which SPF cannot be determined. However, the utilization of fresh-frozen material requires compliance to a number of different technical steps and is therefore cumbersome [23Go].

Prognostic role
Since the first publication in 1987 [24Go], numerous papers have addressed the technological aspects of the method, the relationships of SPF with other standard prognostic factors and its association with clinical outcome. In 1992, the DNA Cytometry Consensus Conference supported by the National Cancer Institute concluded that the literature clearly showed a link between high SPF values and increased risk of recurrence and death for patients with primary BC, but the lack of a standardized procedure to prepare and to analyze tumor samples precluded recommending this method as a routine way to determine prognosis or to select treatment in the adjuvant or metastatic settings [25Go]. Attempts to standardize the method have been made by several groups [23Go, 26Go–30Go], some of which have also prepared guidelines and recommendations and shown the possibility to increase interlaboratory reproducibility [31Go]. Nonetheless, the Tumor Marker Panel of the American Society of Clinical Oncology recently did not include SPF among the standard markers to use for treatment decision-making in BC patients [2Go]. Interestingly, high SPF has been associated with HR-negativity, larger T size, N-positivity and high grade in several reports, and has been found to be a significant independent prognostic factor for early BC patients in 41 out of 49 published papers enrolling a large number of patients (range 131–709) [23Go, 32Go–77Go]. In the majority of the studies, high levels of SPF predicted worse DFS/RFS; in nine this was true only for OS [32Go, 42Go, 58Go, 65Go–68Go, 74Go, 77Go] and in 11 for both DFS and OS [23Go, 33Go, 37Go, 40Go, 51Go, 59Go, 61Go, 69Go]. However, only three studies were prospective [45Go, 49Go, 76Go], and several weaknesses in the design, conduct, and interpretation of the results of these studies hamper the clinical utility of SPF for BC patients. First, the tumor tissue utilized differed across the studies, and while more often paraffin-embedded tissue blocks were processed (26 studies), in others fresh/frozen material was used. Secondly, the characteristics of patients enrolled in the different studies varied according to stage, menopausal status and treatment received. Thirdly, the median follow-up fluctuated from 26 months to 27 years. Fourthly, there was substantial variability in the assay methodology and in the selection of cut-off values. In fact, in some papers, one cut-off point was selected to separate high versus low SPF, while in others, two cut-off points were used in order to provide three categories as suggested by the DNA Cytometry Consensus Conference [25Go].

Predictive role
We found only one retrospective study evaluating the predictive role of SPF for adjuvant tamoxifen therapy in stage I–III BC patients with progesterone receptor (PgR)-positive tumors [78Go]. While with univariate analysis, tamoxifen improved the clinical outcome of patients with high SPF more than in those with low SPF, this effect disappeared with multivariate analysis.


    Tymidine kinase
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Thymidine kinase (TK) is a salvage enzyme implicated in DNA synthesis: it catalyses the phosphorylation of deoxythymidine to deoxythymidine monophosphate. Its activity increases after the G1-S transition checkpoint and then declines rapidly in G2. High levels of TK have been reported in BC patients [79Go–82Go] and refer to the fetal isoform, which is located in the cytoplasm and is cell cycle-regulated [83Go]. TK enzyme activity is determined using a radioenzymatic assay on cytosol and is expressed as mU/mg protein.

Prognostic role
We found only one prospective study [82Go] of 290 N-negative and N-positive BC patients in whom high TK activity was significantly correlated with high grade and PgR-negativity and predicted a worse RFS in the pre/peri-menopausal subset and worse OS in the postmenopausal subgroup.

Predictive role
The predictive value of TK was retrospectively evaluated in two studies [81Go, 84Go], of which one analyzed 1692 BC patients [84Go]. In both studies, high levels were correlated with large T size and HR-negativity [84Go] or PgR-negativity [81Go]. In the first, all N-positive patients received CT with a regimen containing fluorouracil [CMF or CAF (cyclophosphamide, doxorubicin and 5-fluorouracil)] and no endocrine therapy [81Go]. High levels of TK were predictive of worse RFS and OS independently of the treatment, as though the overexpression of this enzyme could allow tumor cells to escape the effects of fluorouracil and methotrexate. In the second study, high TK was predictive of better disease specific survival (DSS) and distant relapse-free interval in N-negative patients treated with anthracycline-based CT (FAC or FEC) in comparison with those untreated [84Go].


    Proliferation associated antigens (Ki-67, MIB 1)
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
Cell proliferation can also be assessed through IHC measurement of the expression of proliferation-associated antigens in tumor tissues. This method is cheap and easy to perform in virtually every pathology department, but it has some shortfalls mainly related to the subjectiveness of the evaluation, the differences in reactivity among the various antibodies available for detecting a given antigen, and other sources of inter-observer variability, such as different reactivity of the detection kits or methods used (e.g. epitope unmasking techniques). Several monoclonal antibodies reacting with different proliferating cell nuclear antigens have been described, such as PCNA, Ki-67 and MIB 1, KiS1 and others. The Ki-67 monoclonal antibody, identified in 1983, reacts with a nuclear protein present exclusively in proliferating cells and the function of which remains unknown. A detailed cell cycle analysis revealed that the antigen was present in the nuclei of cells in all phases as well as in mitosis, while quiescent or resting cells in the G0 phase did not express it. For more detailed information on the protein we refer to two review articles [85Go, 86Go]. Since this protein is present in all proliferating cells (normal and tumor), it became evident that it could be an important marker to evaluate the growth fraction of a given cell population. However, initially the Ki-67 monoclonal antibody could only be used on fresh or frozen tissue, since fixation greatly reduced or abolished immunostaining; this problem could be overcome by the preparation of another equivalent monoclonal antibody MIB-1, which can be evaluated on paraffin sections after antigen retrieval using microwave-processing [87Go]. A good correlation between the two antibodies was demonstrated [88Go, 89Go] so that it was possible to investigate the prognostic potential of the MIB1 protein in retrospective studies using formalin-fixed and paraffin-embedded archived pathologic material. For the other antibodies, conflicting results have been reported [3Go], and therefore they have no role in clinical practice.

Prognostic role
In the last decade, a large number of papers have been published and, even after taking into consideration the inevitable publication bias in favor of positive trials, it is now clear that the Ki-67/MIB 1 protein has a prognostic value for many types of malignant tumors. In BC, all the selected studies [89Go–103Go] have shown a statistically significant correlation with clinical outcome (DFS and/or OS) as reported in Table 2, but all except one [104Go] are retrospective. The number of patients included ranged from 127 to 707, and the populations were heterogeneous, except in three studies [91Go, 96Go, 102Go], which analyzed only N-negative patients. The median follow-up length ranged from 31 months [90Go, 92Go] to 13.5 years [103Go], and other relevant differences among these studies were related to the type of antibody utilized, the cut-off value selected to define high versus low proliferative activity, and the number of cells counted. Although, the Ki-67/MIB 1 protein expressed as Ki-67 index (percentage of cells staining positive) is widely used in the routine assessment of prognostic markers, it is not considered a standard one [1Go, 105Go] due to the lack of an international standardization method for antigen retrieval, staining procedures and scoring methods (semi quantitative and quantitative).


View this table:
[in this window]
[in a new window]
 
Table 2. Studies evaluating Ki-67 as prognostic marker

 
Predictive role
Ki-67 has been retrospectively [106Go–109Go] and prospectively [110Go] assayed in a few trials of primary systemic therapy (PST) in an attempt to identify biological markers of response that could be surrogates for long-term outcome, as shown in Table 3. The only prospective study conducted did not find any significant correlation between high proliferative activity and response to PST, but Ki67 was determined on fine-needle aspirations of the invasive primary tumor obtained before starting CT and repeated before the second cycle [110Go]; furthermore, the chemotherapeutic regimen utilized (mitoxantrone, methotrexate, ± mytomicin C) cannot be considered a standard one, and the follow-up is relatively short. Conflicting results have been reported in the retrospective studies. In one of the two positive studies, for example, the cut-off value chosen was quite high (>40%) [106Go] while, in the other, only a reduction of Ki-67 of ≥36% in two consecutive measures was predictive of response rate (clinical CR or PR) [109Go]. Other differences regarding tumor tissue preservation, monoclonal antibody and scoring system used, chemotherapeutic regimen, evaluation methods of clinical responses, and length of follow-up could partially explain these results. Furthermore, intratumor variations of MIB 1 scoring were reported in the assessment of individual cancers in a limited set of patients, questioning the reliability of monitoring treatment induced changes in proliferation by this technique in the clinic [111Go].


View this table:
[in this window]
[in a new window]
 
Table 3. Studies evaluating Ki-67 as predictive marker

 

    Cyclins E
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
Eukaryotic cells are driven through the cell cycle by successive activation and inactivation of cyclin-dependent kinases (Cdks). The Cdks are regulated by different proteins, including cyclins that bind and activate the Cdks to form a serine/threonine kinase holoenzyme complex. The different cyclins, so called because their concentration rise and fall at specific stages throughout the cell cycle, have a temporally distinct and highly regulated pattern of expression, i.e. they are synthesized and degraded at specific stages of the cell cycle. Cyclin E is the limiting factor for G1 phase progression and S phase entry. It associates with Cdk2 and activates its kinase activity shortly before entry of cells into the S phase. More detailed information on cyclin E can be found in two recently published reviews [112Go, 113Go]. Two proteins, codified by two different genes but with high homology, called cyclin E1 (formerly cyclin E) and cyclin E2 have been identified [114Go–116Go]. Cyclin E2 like cyclin E1 associates with Cdk2 and activates its kinase activity at the G1/S boundary [117Go]. Cyclin E2 shares 47% overall similarity to cyclin E1, and it remains to be determined if structural features outside the conserved regions impart unique functions [117Go]. Recently, several splice variants of cyclin E1, which are not present in normal cells, have also been discovered; importantly, they seem to stimulate cells to progress through the cell cycle much more efficiently than the full length cyclin E1 through a mechanism not yet completely elucidated [113Go, 118Go], but probably different from kinase activation [112Go]. They are generated post translationally through cleavage by elastase, and it is possible that a balance between elastase activity and an endogenous inhibitor of elastase, elafin, controls their expression [119Go]. Cyclin E1 protein, as a proliferative marker, has been analyzed in BC patients, and the determination method more frequently used has been IHC followed by western blot (which allows assessment of both total and isoform-specific expression), while cyclin E mRNA has been measured by real time reverse transcription polymerase chain reaction (RT-PCR). Increased expression of cyclin E1 has been reported in approximately 40% of cases [120Go, 121Go], and of cyclin E2 in 38% of primary ER-negative BC [122Go]. Elevated levels of both cyclins were more frequently found in ER-negative than in ER-positive tumors [123Go].

Prognostic role
The prognostic role of cyclin E (E1) has been retrospectively evaluated in several studies [120Go, 124Go–132Go] shown in Table 4. The majority of these studies included patients with early stage BC, but in two of them [120Go, 124Go], stage IV patients were also entered. Few studies analyzed only N-negative patients [126Go–128Go] and in two of them the same patient population was evaluated with just 59 tamoxifen-treated patients added [126Go]. In some studies no systemic adjuvant treatment was administered, while in others, all patients or only a subgroup of them received CT and/or endocrine therapy. IHC was the most common evaluation method even if the antibodies utilized and the cut-off values were different. Western blotting analysis was performed in two studies [120Go, 124Go], while RT-PCR was chosen in another one [132Go]. Cyclin E was prognostic in seven out of 10 studies [120Go, 125Go–130Go]. Keiomarsi et al. found that the overexpression of cyclin E was accompanied by the appearance of low molecular weight (LMW) isoforms, and both were a reliable prognostic marker in stage I–III BC patients [120Go]. In fact, the hazard ratio for death due to BC in patients with high levels of cyclin E was higher than the hazard ratio associated with any other biological marker examined (seven times higher than the hazard ratio associated with N metastases). All N-negative patients with high levels of cyclin E (12 out of 114) died of BC. In some studies a correlation between cyclin E and high histological grade [126Go–129Go, 131Go] or ER negativity [124Go, 126Go–129Go, 131Go, 132Go] has been reported, while an inverse correlation with p27 was observed in a study where both factors were prognostic [133Go]. No definitive conclusion about the prognostic role of cyclin E can be derived from these studies. In fact, a standardization of evaluation methods and scoring systems and large prospective studies are required. No data on the potential prognostic role of cyclin E2 in BC have been published.


View this table:
[in this window]
[in a new window]
 
Table 4. Studies evaluating cyclin E as prognostic marker

 
Predictive role
The role of cyclin E1 as a predictive marker has been evaluated only in one study by quantitative RT-PCR technique [132Go]. High levels of cyclin E1 were predictive of resistance to tamoxifen adjuvant therapy in 108 node-positive BC patients, independently of ER status.


    Cyclin D1
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
D-type cyclins are other key regulator proteins of the G1 phase progression. There are at least three cyclins in this family with differential effects on the development of the normal mammary gland [123Go]. Cyclins D bind to Cdks 4 and 6 and phosphorylate downstream substrates, mainly pRb, and these complexes can also sequester Cdk inhibitors (p21 and p27) in the G1/S transition. Furthermore, cyclin D1, the first member identified, can have Cdk independent functions and can act as a co-factor for ER{alpha} independently of the ligand [134Go]. Cyclin D1 is the product of a gene (CCND1) located on chromosome 11q13. The protein is synthesized in response to growth factors; its levels reach a maximum in the mid-G1 phase of the cell cycle and then begin to drop. It appears that the association of cyclin D1 to Cdk is crucial to drive cells to the restriction point where the cell is committed to divide [135Go]. Overexpression of cyclin D1 has been observed in many human tumors and is likely to promote cell proliferation and differentiation by shortening the G1/S transition [136Go]. Amplification of the gene has been detected in about 15% of BC, while overexpression of cyclin D1 at mRNA and protein levels is seen in up to 50% of primary BC, mostly ER-positive and well-differentiated tumors [123Go]. The overexpression of cyclin D1 has been evaluated by IHC utilizing different antibodies, or by RT-PCR or northern blot technique.

Prognostic role
As shown in Table 5, seven retrospective studies have analyzed the prognostic role of cyclin D1 in 1509 patients. In two of these studies stage I–IV BC patients were included [137Go, 138Go]; the median follow-up ranged from 75 months to 16.7 years. The most common evaluation method was IHC, with different antibodies and cut-off values utilized. Gene amplification and IHC were both assessed in two studies [139Go, 140Go]. A strong correlation between overexpression of cyclin D1 and HR-positivity has been reported in the majority of trials, but cyclin D1 does not appear to be a strong prognostic marker. In fact, its overexpression has been associated with better RFS in only one study [140Go] and with better RFS and OS in another one [137Go].


View this table:
[in this window]
[in a new window]
 
Table 5. Studies evaluating cyclin D1 as prognostic marker

 
Predictive role
Only one retrospective study has evaluated the predictive role of cyclin D1. At a median follow-up of 18 years, only patients with low/moderate levels of cyclin D1 and ER{alpha}-positive BC tumors benefited from tamoxifen, as though high levels of cyclin D1 affected tamoxifen response [134Go].


    Cdk inhibitors
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
p27
p27 is a CDKI and belongs to the KIP family. The protein binds and inhibits cyclin E–Cdk2 and cyclin A–Cdk2 complexes [141Go–143Go] in the early G1 phase, but it also assembles cyclin D1–Cdks in the cytoplasm and facilitates the import of cyclin D1-complexes into the nucleus. During periods of cell proliferation, p27 remains in storage by binding to Cdk4 or Cdk6 in a non-inhibitory fashion. Anti-proliferative signals, including transforming growth factor-ß (TGF-ß) and cell to cell contact, mobilize the stored p27 so that it can inhibit Cdk2 [144Go]. This all happens in the nucleus because Cdk2 is a nuclear protein, and cytoplasmic localization of p27 would effectively partition it away from this target. Mechanisms of p27 regulation in normal cells and its deregulation in BC have been thoroughly reviewed [145Go].

Prognostic role
In some adjuvant studies, the prognostic role of p27 in BC patients has been retrospectively evaluated by IHC utilizing different monoclonal antibodies [133Go, 146Go–156Go]. Cells were considered positive only when nuclear staining was identified, and the percentage of immunoreactive cells was scored in the majority of studies as low or high using a cut-off value of 50%. The number of patients analyzed ranged from 102 to 830, and they received no adjuvant systemic treatment, CT alone, hormone therapy alone, or both. Low levels of p27 were associated with worse clinical outcome in eight out of 12 studies, but no impact on prognosis was reported in four of them including the two with the largest number of patients [152Go, 153Go, 155Go, 156Go] (Table 6). An inverse relationship was found between p27 levels and histological grade, and a positive one with ER status, in the majority of studies. Furthermore, an inverse correlation with cyclin E levels [121Go, 133Go] was observed, whereas higher concentrations of p27 were associated with increased concentrations of cyclin D1 and low tumor grade [151Go, 152Go, 157Go] or cyclin D expression and bcl-2 positivity [154Go]. A statistically significant correlation between low p27 protein expression and HER2 overexpression (158–160) could validate the hypothesis that one pathway for HER2 oncogenic activity seems to rely on down regulation of the cell-cycle regulator p27 [161Go]. Interestingly, a decrease in the p27 levels has been reported in a trastuzumab-resistant HER2-positive cell line, and the resistance could be reverted by proteasome inhibitors such as MG 132, which induced p27 expression [162Go]. An inverse correlation with p53 was shown in three out of five studies where it was analyzed [147Go, 154Go, 163Go]. In addition, in 202 Askenazi Jewish BC patients [149Go], BRCA 1/2 mutations were associated with low levels of p27, and both were identified as independent prognostic factors. The failure of some studies to find a prognostic value for p27 might reflect differences in tumor fixation, methods of staining and scoring, and also the prolonged storage time of the archival tumor blocks utilized in several studies [145Go]. Therefore, it is crucial to define a uniform methodology for tumor processing, staining and scoring and to evaluate p 27 in large prospective trials.


View this table:
[in this window]
[in a new window]
 
Table 6. Studies evaluating p27 as prognostic marker

 
Predictive role
The predictive role of p27 has been retrospectively evaluated in two adjuvant studies shown in Table 7. In the first, the analysis focused on a group of premenopausal HR-positive BC patients with stage I–II disease enrolled into a randomized trial comparing goserelin plus tamoxifen to CT with CMF, and for whom a tissue sample was available [164Go]. High expression of p27 was observed in about 50% of patients in both arms and was found to be predictive of better RFS and OS, at a median follow-up of 5.5 years. The protein was also an independent predictor of responsiveness to endocrine therapy. Moreover, in patients with high p27 expression, endocrine therapy was superior to CT, although the benefit was statistically significant only for RFS. An unfavorable outcome was observed in patients with low p27 expression regardless of the adjuvant systemic treatment given.


View this table:
[in this window]
[in a new window]
 
Table 7. Studies evaluating p27 as predictive marker

 
In the other study [159Go], p27 immunoreactivity was analyzed in N-negative and N-positive BC patients enrolled in the International Breast Cancer Study Group (IBCSG) Ludwig trial V, but it did not show any significant prognostic value in terms of DFS and OS. However, in the N-negative population, the benefit from one course of perioperative CT (CMF i.v.) was confined exclusively to patients with tumors showing low levels of p27. In this subgroup of patients, low levels of p27 were associated with HER2 overexpression so, both factors should probably be analyzed to predict response to drugs that act on EGF/HER pathways. The usefulness of p27 in predicting response to systemic treatment needs to be investigated in large prospective trials.


    p21WAF1/CIP1
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
The p21WAF1 (wild type p53 activated fragment 1) is a nuclear protein, also known as cyclin-dependent kinase-interacting protein 1 (CIP1) or senescent cell-derived inhibitor 1 (SDI1), that has been implicated in the mechanisms of cell-cycle arrest that allow cell DNA repair, differentiation and apoptosis. In response to DNA damaging agents, wild type p53 induces the expression of p21, which blocks the progression of the cell cycle at the G1/S transition by inhibiting the activity of Cdk2 and Cdk4/6; it can also serve as an assembly factor for cyclin D–Cdk4 complexes, increasing the efficiency of complex formation and Cdk4 activity. The integrity of G1 and G2 checkpoints requires the nuclear localization of p21WAF1. It has been shown that in cancer cells and in cell lines, p21WAF1 can localize in the cytoplasm where it inhibits apoptosis by binding and inhibiting the apoptosis signal-regulating kinase 1. In addition, other factors can induce p21 expression and cell cycle arrest independently from p53, such as insulin-like growth factors [165Go], epidermal growth factor [166Go], or HER2 expression [167Go]. Upregulation of p21WAF1 occurs through the phosphatidylinositol 3-kinase/Akt (PI-3K/Akt) signaling. In HER2 overexpressing breast cancer cell lines, p21WAF1 is transcriptionally upregulated but also dislocated into the cytoplasm through a mechanism whereby Akt binds and phosphorylates p21WAF1 in its nuclear-localization signal [167Go]. p21WAF1 has also been identified as an anti-estrogen-regulated factor capable of inhibiting the Cdk4 activity present in extracts of estrogen-treated human BC cells [168Go].

Prognostic role
The prognostic role of p21WAF1 has been retrospectively analyzed in several adjuvant studies [169Go–178Go], with a number of patients ranging from 104 to 798, and a wide range of median follow-up times (Table 8). IHC was the evaluation method for p21WAF1 in all trials, and only tumor cells with detectable nuclear staining were considered positive. However, different monoclonal antibodies and scoring systems were adopted. When p21WAF1 was correlated with standard prognostic factors and other biological markers, contradictory results were obtained. Since p21WAF1 is a downstream effector in the p53-specific pathway of growth control, p53 was analyzed in all trials, although in the majority of them no association was found. Low expression of p21WAF1 was correlated with high histologic grade in three studies [171Go, 172Go, 178Go], while in one study the opposite was observed [170Go]. A positive association with proliferative activity was reported in three studies in which MIB-1 [175Go], Ki-67 [174Go] or cell nuclear antigen (PCNA) were measured; in contrast [177Go], an inverse association was seen with HR status in two [172Go, 175Go] and with N status in three studies [170Go–172Go]. Low levels of p21WAF1 were an independent prognostic factor for DFS only or for DFS and OS in three studies [171Go, 172Go, 178Go], even if in one of them it depended on the cut-off value chosen [178Go]. Moreover, low expression of p21WAF1 was prognostic only in combination with increased expression of p53 in three studies [170Go, 174Go, 178Go]. The combination of low levels of p21WAF1 and low SPF or low levels of cyclin A were associated with good prognosis on multivariate analysis in a study conducted in a relatively small number of patients [169Go]. The heterogeneity of the results reported in these studies calls for the necessity to standardize the evaluation method and scoring system for p21WAF1 determination.


View this table:
[in this window]
[in a new window]
 
Table 8. Studies evaluating p21 as prognostic marker

 
Predictive role
Only one paper responding to our selection criteria could be found that retrospectively evaluated the predictive role of p21WAF1 [179Go]. In this study, 107 N-negative and N-positive BC patients were enrolled, but only 94 received adjuvant CT with CMF. A worse DFS was observed in patients treated with CMF if the tumor expressed high levels of p21 (either cytoplasmic or nuclear), and a positive association was found between high levels of p21 and HER2. These results seem to suggest that p21WAF1 may play a role in HER2-mediated CMF resistance.


    Topoisomerase II{alpha}
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
Description
Topoisomerases II (topo II) are essential nuclear DNA-binding enzymes that control and modify the topological states of DNA by combining nuclease, helicase and ligase activities. In human cells, two distinct isoforms of topo II exist, termed {alpha} and ß. These share considerable homology (72%), but are products of different genes located on chromosomes 17q21 and 3p, respectively. They differ in molecular weight, pattern of expression, function, and their apparent sensitivity to antineoplastic drugs. The ß isoforms show no dependency on cell cycle phase, and its function is still largely unknown; in contrast, topo II{alpha} expression is cell cycle-dependent (G2/M) and reduces DNA supercoiling and twisting by creating a double-strand nick that enables the passage of a second DNA double-strand through the break and subsequent religation of the cleaved one. Topo II{alpha} is also essential for carrying out functions in the segregation and condensation of newly replicated chromosome pairs in dividing cells. Topo II{alpha} protein levels are markedly higher in exponentially growing than in quiescent cell lines in tissue culture, and can be down-regulated by growth of cells at high density or in serum-free conditions [180Go]. Reduced levels of topo II{alpha} have been found in cells induced to differentiate [181Go, 182Go] so that it can be regarded as a marker of cell proliferation. Topo II{alpha} is the target enzyme for topo II{alpha} inhibitors such as anthracyclines, epipodophyllotoxins, actinomycin, mitoxantrone and other drugs, and in vitro data have demonstrated a direct correlation between the intranuclear topo II{alpha} levels and the degree of sensitivity to these agents. Moreover, the topo II{alpha} gene (TOP 2A) is located close to the Her2 oncogene on the 17th chromosome. This could explain the very high incidence of TOP 2A aberrations in Her2 amplified tumors, with about 40% co-amplification and approximately 40% of TOP 2A deletions [183Go]. Only about 8%–10% of TOP 2A aberrations occur in Her2 negative patients [184Go, 185Go]. The exact mechanism by which Her2 amplification leads to TOP 2A deletion is not clear. There is some evidence that deletion of TOP 2A is associated with low levels of topoII{alpha} protein or ‘haploinsufficiency’ [183Go, 186Go]. Interestingly, Her2 amplified, TOP 2A amplified cells were shown to be highly sensitive to anthracyclines, while HER2 amplified, TOP 2A deleted cells displayed a poor degree of sensitivity to these drugs [183Go]. Determination of topo II{alpha} by IHC utilizing two different monoclonal antibodies was compared with other detection methods such as polymerase chain reaction-aided transcript titration assay, enzyme activity assay and western blotting and was competitive and easier to perform [187Go]. Topo II{alpha} expression was also analyzed by mRNA in situ hybridization using synthetic oligonucleotide probes, and a close correlation with IHC was obtained. Discordance between topo II{alpha} amplification and overexpression by IHC has been reported [188Go–190Go], suggesting that the protein expression can be regulated at different levels, including post-transcription and post-translation steps. While in one of these studies a correlation between TOP2A amplification and Her2 amplification was observed [188Go], in the other this correlation was not found [190Go]. TOP2A amplification and Her2 amplification both assayed by chromogenic in situ hybridisation (CISH) were significantly correlated in a small study in locally advanced BC patients [191Go]. In two studies, increased expression of topo II{alpha} was associated with Her2 amplification [192Go] or with HER2 overexpression [193Go], while no association between the overexpression of the two proteins was reported by others [188Go, 194Go, 195Go].

Prognostic role
Only three retrospective adjuvant studies evaluating overexpression of topoII{alpha} by IHC were selected through the criteria chosen (Table 9) [192Go, 193Go, 196Go]. A positive association with HER2 overexpression or amplification was observed in all of them, as well as with tumor size or stage, and in two studies also with high Ki-67 or MIB 1 [192Go, 193Go]. Overexpression of topo II{alpha} predicted a shorter DFS, specific survival (SS) and/or OS in multivariate analysis only in two studies [193Go, 196Go]. Large prospective studies using standardized methods are needed to better define the potential clinical relevance of topo II{alpha} as a prognostic factor.


View this table:
[in this window]
[in a new window]
 
Table 9. Studies evaluating topoisomerase II{alpha} as prognostic marker

 
Predictive role
Some retrospective studies have investigated the role of either the TOP 2A gene or the topo II{alpha} protein as predictive factors of response to antracycline-based therapy in all stages of disease, but for this review we could find only one ‘eligible’ study. Response to primary CT utilizing an anthracycline-based regimen was retrospectively evaluated in patients with operable metastasis-free BC larger than 3 cm. A high percentage of topo II{alpha} measured by IHC was correlated to high Ki-67 and to HR-negativity, but not to HER2 overexpression, and it was predictive of response to CT, in multivariate analysis, with tumor size <40 mm and ER-negativity [194Go]. In this paper, HER2 did not seem to be directly involved in the prediction of anthracycline efficacy, in contrast to what has been reported by other authors [188Go, 191Go, 197Go]. The predictive value of TOP 2A amplification when it does not translate into protein overexpression remains to be determined, but it could be hypothesized that both the TOP 2A gene and the topo II{alpha} protein might be useful tools to define the tumor profile with regard to sensitivity to anthracyclines. An ongoing meta-analysis of four studies comparing CMF to anthracycline-based regimens in the adjuvant setting and involving 4600 patients will likely clarify the predictive value of topo II{alpha} and HER2 status for response to anthracyclines (Di Leo A, personal communication).


    Conclusions
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
One hundred and thirty-five papers evaluating the prognostic and predictive roles of both some ‘old’ and some ‘new’ proliferative markers have been reviewed. A total of 159 516 BC patients have been examined in these studies, exemplifying the tremendous efforts made in an attempt to classify these patients better according to risk and to select their optimal treatment. While as many as 102 studies were ‘positive’, not one of these markers has been included among the standard ones. The results obtained for some markers related to cell cycle control are summarized in Figure 1. The criteria required to define a clinically useful prognostic/predictive factor are quite complex, and all must be fulfilled [198Go]. For some markers, such as SPF, Ki 67 and p27, we selected several studies that reported a significant correlation of the examined marker with clinical outcome on multivariate analysis, but the majority had a level of evidence III or IV and, therefore, their prognostic or predictive value still remains undefined. For other markers such as TLI, the predictive value shown in prospective trials reached level 1 evidence, but the practical difficulties linked to the determination method precluded its adoption in clinical practice.



View larger version (34K):
[in this window]
[in a new window]
 
Figure 1. The prognostic and predictive value of the proliferative markers related to cell cycle control in breast cancer: summary of results; IHC, immunohistochemistry; Tam, tamoxifen; chemo, chemotherapy; CMF, cyclophosphamide, methotrexate; 5-fluorouracil.

 
Among the reasons for these ambiguous results are: (1) the retrospective nature of the great majority of studies; (2) the utilization of archived materials handled and preserved in different conditions; (3) the relatively small numbers of patients analyzed and the fact that the group of patients included in the translational research study is always a subset of the clinical trial population, hence inducing bias; (4) the heterogeneity of the population of patients evaluated; (5) the different methods used to assess the expression of biological and molecular markers, as well as the threshold values to define their expression; (6) the lack of standardization and quality control evaluation of different methods; and, sometimes (7) the relatively short duration of the median follow-up. Nevertheless, important messages can be derived from these studies, even if a publication selection bias cannot be ruled out. First, proliferative activity seems to have an important role in defining the behavior of BC, with theoretically possible implications for the selection of treatments, such as dose dense CT. Secondly, a standardization of the evaluation methods with intra- and interlaboratory quality controls is the first requisite to evaluate a marker. Thirdly, it is crucial to properly design large prospective trials trying to include more than one proliferative marker in order to select the best ones or the best combinations with prognostic/predictive value. International collaboration and close interaction between the basic and clinical research worlds (through translational research) are essential to avoid the duplication of efforts and fragmentation of research, and to lead to the needed level 1 evidence. In the era of high throughput technologies, such as microarray gene profiling and proteomics, there is a greater chance to develop multimarker models that might turn out to be powerful prognostic or predictive tools. Microarray studies focusing on re-classification of BC have consistently shown the existence of a special subtype, called basal-like, which is characterized among other things, by a high level of proliferative markers such as topoisomerase II{alpha}.

Received for publication January 18, 2005. Revision received April 11, 2005. Accepted for publication April 12, 2005.


    References
 Top
 Abstract
 Introduction
 Materials and methods
 Measurement of cells in...
 3H-thymidine labeling index
 Flow cytometry
 Tymidine kinase
 Proliferation associated...
 Cyclins E
 Cyclin D1
 Cdk inhibitors
 p21WAF1/CIP1
 Topoisomerase II{alpha}
 Conclusions
 References
 
1. Goldhirsch A, Wood WC, Gelber RD et al. Meeting highlights: updated international expert consensus on the primary therapy of early breast cancer. J Clin Oncol 2003; 21: 3357–3365.[Abstract/Free Full Text]

2. Bast RC, Jr, Ravdin P, Hayes DF et al. 2000 update of recommendations for the use of tumor markers in breast and colorectal cancer: clinical practice guidelines of the American Society of Clinical Oncology. J Clin Oncol 2001; 19: 1865–1878.[Abstract/Free Full Text]

3. van Diest PJ, van der Wall E, Baak JP. Prognostic value of proliferation in invasive breast cancer: a review. J Clin Pathol 2004; 57: 675–681.[Abstract/Free Full Text]

4. Hayes DF, Bast RC, Desch CE et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst 1996; 88: 1456–1466.[Abstract/Free Full Text]

5. Meyer JS, Connor RE. In vitro labeling of solid tissues with tritiated thymidine for autoradiographic detection of S-phase nuclei. Stain Technol 1977; 52: 185–195.[ISI][Medline]

6. Meyer JS, McDivitt RW. Reliability and stability of the thymidine labeling index of breast carcinoma. Lab Invest 1986; 54: 160–164.[ISI][Medline]

7. Sasaki K, Matsumura K, Tsuji T et al. Relationship between labeling indices of Ki-67 and BrdUrd in human malignant tumors. Cancer 1988; 62: 989–993.[ISI][Medline]

8. Moriki T, Takahashi T, Kataoka H et al. Proliferation marker MIB-1 correlates well with proliferative activity evaluated by BrdU in breast cancer: an immunohistochemical study including correlation with PCNA, p53, c-erbB-2 and estrogen receptor status. Pathol Int 1996; 46: 953–961.[ISI][Medline]

9. Meyer JS, Friedman E, McCrate MM, Bauer WC. Prediction of early course of breast carcinoma by thymidine labeling. Cancer 1983; 51: 1879–1886.[ISI][Medline]

10. Meyer JS, Province M. Proliferative index of breast carcinoma by thymidine labeling: prognostic power independent of stage, estrogen and progesterone receptors. Breast Cancer Res Treat 1988; 12: 191–204.[CrossRef][ISI][Medline]

11. Tubiana M, Pejovic MH, Koscielny S et al. Growth rate, kinetics of tumor cell proliferation and long-term outcome in human breast cancer. Int J Cancer 1989; 44: 17–22.[ISI][Medline]

12. Silvestrini R, Daidone MG, Valagussa P et al. Cell kinetics as a prognostic indicator in node-negative breast cancer. Eur J Cancer Clin Oncol 1989; 25: 1165–1171.[CrossRef][ISI][Medline]

13. Silvestrini R, Daidone MG, Luisi A et al. Cell proliferation in 3,800 node-negative breast cancers: consistency over time of biological and clinical information provided by 3H-thymidine labelling index. Int J Cancer 1997; 74: 122–127.[CrossRef][ISI][Medline]

14. Silvestrini R, Daidone MG, Del Bino G et al. Prognostic significance of proliferative activity and ploidy in node-negative breast cancers. Ann Oncol 1993; 4: 213–219.[Abstract]

15. Volpi A, De Paola F, Nanni O et al. Prognostic significance of biologic markers in node-negative breast cancer patients: a prospective study. Breast Cancer Res Treat 2000; 63: 181–192.[CrossRef][ISI][Medline]

16. Medri L, Volpi A, Nanni O et al. Prognostic relevance of mitotic activity in patients with node-negative breast cancer. Mod Pathol 2003; 16: 1067–1075.[CrossRef][ISI][Medline]

17. Medri L, Nanni O, Volpi A et al. Tumor microvessel density and prognosis in node-negative breast cancer. Int J Cancer 2000; 89: 74–80.[CrossRef][ISI][Medline]

18. Lloveras B, Edgerton S, Thor AD. Evaluation of in vitro bromodeoxyuridine labeling of breast carcinomas with the use of a commercial kit. Am J Clin Pathol 1991; 95: 41–47.[ISI][Medline]

19. Waldman FM, Chew K, Ljung BM et al. A comparison between bromodeoxyuridine and 3H thymidine labeling in human breast tumors. Mod Pathol 1991; 4: 718–722.[ISI][Medline]

20. Silvestrini R, Luisi A, Zambetti M et al. Cell proliferation and outcome following doxorubicin plus CMF regimens in node-positive breast cancer. Int J Cancer 2000; 87: 405–411.[CrossRef][ISI][Medline]

21. Amadori D, Nanni O, Marangolo M et al. Disease-free survival advantage of adjuvant cyclophosphamide, methotrexate, and fluorouracil in patients with node-negative, rapidly proliferating breast cancer: a randomized multicenter study. J Clin Oncol 2000; 18: 3125–3134.[Abstract/Free Full Text]

22. Paradiso A, Schittulli F, Cellamare G et al. Randomized clinical trial of adjuvant fluorouracil, epirubicin, and cyclophosphamide chemotherapy for patients with fast-proliferating, node-negative breast cancer. J Clin Oncol 2001; 19: 3929–3937.[Abstract/Free Full Text]

23. Duigou F, Herlin P, Marnay J, Michels JJ. Variation of flow cytometric DNA measurement in 1,485 primary breast carcinomas according to guidelines for DNA histogram interpretation. Cytometry 2000; 42: 35–42.[CrossRef][ISI][Medline]

24. Hedley DW, Rugg CA, Gelber RD. Association of DNA index and S-phase fraction with prognosis of nodes positive early breast cancer. Cancer Res 1987; 47: 4729–4735.[Abstract]

25. Hedley DW, Clark GM, Cornelisse CJ et al. Consensus review of the clinical utility of DNA cytometry in carcinoma of the breast. Report of the DNA Cytometry Consensus Conference. Cytometry 1993; 14: 482–485.[ISI][Medline]

26. Baldetorp B, Bendahl PO, Ferno M et al. Reproducibility in DNA flow cytometric analysis of breast cancer: comparison of 12 laboratories' results for 67 sample homogenates. Cytometry 1995; 22: 115–127.[CrossRef][ISI][Medline]

27. Tirindelli Danesi D, Spano M, Altavista P et al. Quality control study of the Italian Group of Cytometry on flow cytometry DNA content measurements: II. Factors affecting inter- and intralaboratory variability. Cytometry 1997; 30: 85–97.[CrossRef][ISI][Medline]

28. Ormerod MG, Tribukait B, Giaretti W. Consensus report of the task force on standardisation of DNA flow cytometry in clinical pathology. DNA Flow Cytometry Task Force of the European Society for Analytical Cellular Pathology. Anal Cell Pathol 1998; 17: 103–110.[ISI][Medline]

29. Chassevent A, Jourdan ML, Ferrero-Pous M et al. Standardization and quality control in the evaluation of proliferation parameters in T1T2, N0N1, M0 breast cancer: multicentric retrospective study II. DNA-ploidy and S-phase fraction. Bull Cancer 1999; 86: 685–691.[ISI][Medline]

30. D'Hautcourt JL, Spyratos F, Chassevent A. Quality control study by the French Cytometry Association on flow cytometric DNA content and S-phase fraction (S%). The Association Francaise de Cytometrie. Cytometry 1996; 26: 32–39.[CrossRef][ISI][Medline]

31. Baldetorp B, Bendahl PO, Ferno M, Stal O. Improved DNA flow cytometric, DNA ploidy, and S-phase reproducibility between 15 laboratories in analysis of breast cancer using generalized guidelines. Cytometry 2003; 56A: 1–7.

32. Clark GM, Wenger CR, Beardslee S et al. How to integrate steroid hormone receptor, flow cytometric, and other prognostic information in regard to primary breast cancer. Cancer 1993; 71 (Suppl 6): 2157–2162.[ISI][Medline]

33. Clark GM, Dressler LG, Owens MA et al. Prediction of relapse or survival in patients with node-negative breast cancer by DNA flow cytometry. N Engl J Med 1989; 320: 627–633.[Abstract]

34. Clark GM, Mathieu MC, Owens MA et al. Prognostic significance of S-phase fraction in good-risk, node-negative breast cancer patients. J Clin Oncol 1992; 10: 428–432.[Abstract/Free Full Text]

35. Sigurdsson H, Baldetorp B, Borg A et al. Flow cytometry in primary breast cancer: improving the prognostic value of the fraction of cells in the S-phase by optimal categorisation of cut-off levels. Br J Cancer 1990; 62: 786–90.[ISI][Medline]

36. Sigurdsson H, Baldetorp B, Borg A et al. Indicators of prognosis in node-negative breast cancer. N Engl J Med 1990; 322: 1045–1053.[Abstract]

37. Stal O, Wingren S, Carstensen J et al. Prognostic value of DNA ploidy and S-phase fraction in relation to estrogen receptor content and clinicopathological variables in primary breast cancer. Eur J Cancer Clin Oncol 1989; 25: 301–309.[CrossRef][ISI][Medline]

38. Stal O, Carstensen J, Hatschek T, Nordenskjold B. Significance of S-phase fraction and hormone receptor content in the management of young breast cancer patients. Br J Cancer 1992; 66: 706–711.[ISI][Medline]

39. Stal O, Skoog L, Rutqvist LE et al. S-phase fraction and survival benefit from adjuvant chemotherapy or radiotherapy of breast cancer. Br J Cancer 1994; 70: 1258–1262.[ISI][Medline]

40. Stal O, Dufmats M, Hatschek T et al. S-phase fraction is a prognostic factor in stage I breast carcinoma. J Clin Oncol 1993; 11: 1717–1722.[Abstract/Free Full Text]

41. Ferno M, Baldetorp B, Borg A et al. Flow cytometric DNA index and S-phase fraction in breast cancer in relation to other prognostic variables and to clinical outcome. Acta Oncol 1992; 31: 157–165.[ISI][Medline]

42. Lawry J, Rogers K, Duncan JL, Potter CW. The identification of informative parameters in the flow cytometric analysis of breast carcinoma. Eur J Cancer 1993; 29A: 719–723.

43. Wingren S, Stal O, Sullivan S et al. S-phase fraction after gating on epithelial cells predicts recurrence in node-negative breast cancer. Int J Cancer 1994; 59: 7–10.[ISI][Medline]

44. Michels JJ, Duigou F, Marnay J. Flow cytometry in primary breast carcinomas. Prognostic impact of proliferative activity. Breast Cancer Res Treat 2000; 62: 117–126.[CrossRef][ISI][Medline]

45. Michels JJ, Marnay J, Delozier T et al. Proliferative activity in primary breast carcinomas is a salient prognostic factor. Cancer 2004; 100: 455–464.[CrossRef][ISI][Medline]

46. Simpson JF, Gray R, Dressler LG et al. Prognostic value of histologic grade and proliferative activity in axillary node-positive breast cancer: results from the Eastern Cooperative Oncology Group Companion Study, EST 4189. J Clin Oncol 2000; 18: 2059–2069.[Abstract/Free Full Text]

47. Bagwell CB, Clark GM, Spyratos F et al. Optimizing flow cytometric DNA ploidy and S-phase fraction as independent prognostic markers for node-negative breast cancer specimens. Cytometry 2001; 46: 121–135.[CrossRef][ISI][Medline]

48. Pinto AE, Andre S, Soares J. Short-term significance of DNA ploidy and cell proliferation in breast carcinoma: a multivariate analysis of prognostic markers in a series of 308 patients. J Clin Pathol 1999; 52: 604–611.[Abstract]

49. Malmstrom P, Bendahl PO, Boiesen P et al. S-phase fraction and urokinase plasminogen activator are better markers for distant recurrences than Nottingham Prognostic Index and histologic grade in a prospective study of premenopausal lymph node-negative breast cancer. J Clin Oncol 2001; 19: 2010–2019.[Abstract/Free Full Text]

50. Chassevent A, Jourdan ML, Romain S et al. S-phase fraction and DNA ploidy in 633 T1T2 breast cancers: a standardized flow cytometric study. Clin Cancer Res 2001; 7: 909–917.[Abstract/Free Full Text]

51. Kute TE, Russell GB, Zbieranski N et al. Prognostic markers in node-negative breast cancer: a prospective study. Cytometry 2004; 59B: 24–31.[CrossRef][Medline]

52. Kute TE, Muss HB, Cooper MR et al. The use of flow cytometry for the prognosis of stage II adjuvant treated breast cancer patients. Cancer 1990; 66: 1810–1816.[ISI][Medline]

53. Fisher B, Gunduz N, Costantino J et al. DNA flow cytometric analysis of primary operable breast cancer. Relation of ploidy and S-phase fraction to outcome of patients in NSABP B-04. Cancer 1991; 68: 1465–1475.[ISI][Medline]

54. Mandard AM, Denoux Y, Herlin P et al. Prognostic value of DNA cytometry in 281 premenopausal patients with lymph node negative breast carcinoma randomized in a control trial: multivariate analysis with Ki-67 index, mitotic count, and microvessel density. Cancer 2000; 89: 1748–1757.[CrossRef][ISI][Medline]

55. O'Reilly SM, Camplejohn RS, Barnes DM, Millis RR, Rubens RD, Richards MA. Node-negative breast cancer: prognostic subgroups defined by tumor size and flow cytometry. J Clin Oncol 1990; 8: 2040–2046.[Abstract/Free Full Text]

56. Joensuu H, Toikkanen S, Klemi PJ. DNA index and S-phase fraction and their combination as prognostic factors in operable ductal breast carcinoma. Cancer 1990; 66: 331–340.[ISI][Medline]

57. Harbeck N, Dettmar P, Thomssen C et al. Prognostic impact of tumor biological factors on survival in node-negative breast cancer. Anticancer Res 1998; 18: 2187–2197.[ISI][Medline]

58. Bergers E, Baak JP, van Diest PJ et al. Prognostic implications of different cell cycle analysis models of flow cytometric DNA histograms of 1,301 breast cancer patients: results from the Multicenter Morphometric Mammary Carcinoma Project (MMMCP). Int J Cancer 1997; 74: 260–269.[CrossRef][ISI][Medline]

59. Gasparini G, Boracchi P, Verderio P, Bevilacqua P. Cell kinetics in human breast cancer: comparison between the prognostic value of the cytofluorimetric S-phase fraction and that of the antibodies to Ki-67 and PCNA antigens detected by immunocytochemistry. Int J Cancer 1994; 57: 822–829.[ISI][Medline]

60. Stanton PD, Cooke TG, Oakes SJ et al. Lack of prognostic significance of DNA ploidy and S phase fraction in breast cancer. Br J Cancer 1992; 66: 925–929.[ISI][Medline]

61. Camplejohn RS, Ash CM, Gillett CE et al. The prognostic significance of DNA flow cytometry in breast cancer: results from 881 patients treated in a single centre. Br J Cancer 1995; 71: 140–145.[ISI][Medline]

62. Peiro G, Lerma E, Climent MA et al. Prognostic value of S-phase fraction in lymph-node-negative breast cancer by image and flow cytometric analysis. Mod Pathol 1997; 10: 216–222.[ISI][Medline]

63. Balslev I, Christensen IJ, Rasmussen BB et al. Flow cytometric DNA ploidy defines patients with poor prognosis in node-negative breast cancer. Int J Cancer 1994; 56: 16–25.[ISI][Medline]

64. Meyer JS, Province MA. S-phase fraction and nuclear size in long term prognosis of patients with breast cancer. Cancer 1994; 74: 2287–2299.[ISI][Medline]

65. Merkel DE, Winchester DJ, Goldschmidt RA et al. DNA flow cytometry and pathologic grading as prognostic guides in axillary lymph node-negative breast cancer. Cancer 1993; 72: 1926–1932.[ISI][Medline]

66. Kallioniemi OP, Blanco G, Alavaikko M et al. Improving the prognostic value of DNA flow cytometry in breast cancer by combining DNA index and S-phase fraction. A proposed classification of DNA histograms in breast cancer. Cancer 1988; 62: 2183–2190.[ISI][Medline]

67. Brooks SA, Leathem AJ, Camplejohn RS, Gregory W. Markers of prognosis in breast cancer—the relationship between binding of the lectin HPA and histological grade, SPF, and ploidy. Breast Cancer Res Treat 1993; 25: 247–256.[CrossRef][ISI][Medline]

68. Toikkanen S, Joensuu H, Klemi P. The prognostic significance of nuclear DNA content in invasive breast cancer—a study with long-term follow-up. Br J Cancer 1989; 60: 693–700.[ISI][Medline]

69. Romero H, Schneider J, Burgos J, Bilbao J, Rodriguez-Escudero FJ. S-phase fraction identifies high-risk subgroups among DNA-diploid breast cancers. Breast Cancer Res Treat 1996; 38: 265–275.[CrossRef][ISI][Medline]

70. Wenger CR, Beardslee S, Owens MA et al. DNA ploidy, S-phase, and steroid receptors in more than 127,000 breast cancer patients. Breast Cancer Res Treat 1993; 28: 9–20.[CrossRef][ISI][Medline]

71. Winchester DJ, Duda RB, August CZ et al. The importance of DNA flow cytometry in node-negative breast cancer. Arch Surg 1990; 125: 886–889.[Abstract]

72. Witzig TE, Gonchoroff NJ, Therneau T et al. DNA content flow cytometry as a prognostic factor for node-positive breast cancer. The role of multiparameter ploidy analysis and specimen sonication. Cancer 1991; 68: 1781–1788.[ISI][Medline]

73. Witzig TE, Ingle JN, Cha SS et al. DNA ploidy and the percentage of cells in S-phase as prognostic factors for women with lymph node negative breast cancer. Cancer 1994; 74: 1752–1761.[ISI][Medline]

74. Johnson H Jr., Masood S, Belluco C et al. Prognostic factors in node-negative breast cancer. Arch Surg 1992; 127: 1386–1391.[Abstract]

75. Bosari S, Lee AK, Tahan SR et al. DNA flow cytometric analysis and prognosis of axillary lymph node-negative breast carcinoma. Cancer 1992; 70: 1943–1950.[ISI][Medline]

76. Ewers SB, Attewell R, Baldetorp B et al. Prognostic potential of flow cytometric S-phase and ploidy prospectively determined in primary breast carcinomas. Breast Cancer Res Treat 1992; 20: 93–108.[CrossRef][ISI][Medline]

77. Remvikos Y, Mosseri V, Asselain B et al. S-phase fractions of breast cancer predict overall and post-relapse survival. Eur J Cancer 1997; 33: 581–586.[CrossRef][Medline]

78. Ferno M, Baldetorp B, Bendahl PO et al. Recurrence-free survival in breast cancer improved by adjuvant tamoxifen–especially for progesterone receptor positive tumors with a high proliferation. Breast Cancer Res Treat 1995; 36: 23–34.[CrossRef][ISI][Medline]

79. O'Neill KL, Hoper M, Odling-Smee GW. Can thymidine kinase levels in breast tumors predict disease recurrence? J Natl Cancer Inst 1992; 84: 1825–1828.[Abstract]

80. Spyratos F, Martin PM, Hacene K et al. Multiparametric prognostic evaluation of biological factors in primary breast cancer. J Natl Cancer Inst 1992; 84: 1266–1272.[Abstract]

81. Romain S, Martin PM, Klijn JG et al. DNA-synthesis enzyme activity: a biological tool useful for predicting anti-metabolic drug sensitivity in breast cancer? Int J Cancer 1997; 74: 156–161.[CrossRef][ISI][Medline]

82. Romain S, Christensen IJ, Chinot O et al. Prognostic value of cytosolic thymidine kinase activity as a marker of proliferation in breast cancer. Int J Cancer 1995; 61: 7–12.[ISI][Medline]

83. Arner ES, Eriksson S. Mammalian deoxyribonucleoside kinases. Pharmacol Ther 1995; 67: 155–186.[CrossRef][ISI][Medline]

84. Broet P, Romain S, Daver A et al. Thymidine kinase as a proliferative marker: clinical relevance in 1,692 primary breast cancer patients. J Clin Oncol 2001; 19: 2778–2787.[Abstract/Free Full Text]

85. Scholzen T, Gerdes J. The Ki- 67 protein: from the known and the unknown. J Cell Physiol 2000; 182: 311–322.[CrossRef][ISI][Medline]

86. Brown DC, Gatter KC. Ki67 protein: the immaculate deception? Histopathology 2002; 40: 2–11.[CrossRef][ISI][Medline]

87. Cattoretti G, Becker MH, Key G et al. Monoclonal antibodies against recombinant parts of the Ki-67 antigen (MIB 1 and MIB 3) detect proliferating cells in microwave-processed formalin-fixed paraffin sections. J Pathol 1992; 168: 357–363.[ISI][Medline]

88. Veronese SM, Maisano C, Scibilia J. Comparative prognostic value of Ki-67 and MIB-1 proliferation indices in breast cancer. Anticancer Res 1995; 15 (6B): 2717–2722.[ISI][Medline]

89. Querzoli P, Albonico G, Ferretti S et al. MIB-1 proliferative activity in invasive breast cancer measured by image analysis. J Clin Pathol 1996; 49: 926–930.[Abstract]

90. Railo M, Nordling S, von Boguslawsky K et al. Prognostic value of Ki-67 immunolabelling in primary operable breast cancer. Br J Cancer 1993; 68: 579–583.[ISI][Medline]

91. Railo M, Lundin J, Haglund C et al. Ki-67, p53, Er-receptors, ploidy and S-phase as prognostic factors in T1 node negative breast cancer. Acta Oncol 1997; 36: 369–374.[ISI][Medline]

92. Gaglia P, Bernardi A, Venesio T et al. Cell proliferation of breast cancer evaluated by anti-BrdU and anti-Ki-67 antibodies: its prognostic value on short-term recurrences. Eur J Cancer 1993; 29A: 1509–1513.[CrossRef]

93. Keshgegian AA, Cnaan A. Proliferation markers in breast carcinoma. Mitotic figure count, S-phase fraction, proliferating cell nuclear antigen, Ki-67 and MIB-1. Am J Clin Pathol 1995; 104: 42–49.[ISI][Medline]

94. Pinder SE, Wencyk P, Sibbering DM et al. Assessment of the new proliferation marker MIB1 in breast carcinoma using image analysis: associations with other prognostic factors and survival. Br J Cancer 1995; 71: 146–149.[ISI][Medline]

95. Jansen RL, Hupperets PS, Arends JW et al. MIB-1 labelling index is an independent prognostic marker in primary breast cancer. Br J Cancer 1998; 78: 460–465.[ISI][Medline]

96. Brown RW, Allred CD, Clark GM et al. Prognostic value of Ki-67 compared to S-phase fraction in axillary node-negative breast cancer. Clin Cancer Res 1996; 2: 585–592.[Abstract/Free Full Text]

97. Pierga JY, Leroyer A, Viehl P et al. Long term prognostic value of growth fraction determination by Ki-67 immunostaining in primary operable breast cancer. Breast Cancer Res Treat 1996; 37: 57–64.[CrossRef][ISI][Medline]

98. Domagala W, Markiewski M, Harezga B, Dukowicz A, Osborn M. Prognostic significance of tumor cell proliferation rate as determined by the MIB-1 antibody in breast carcinoma: its relationship with vimentin and p53 protein. Clin Cancer Res 1996; 2: 147–154.[Abstract]

99. Seshadri R, Leong AS, McCaul K et al. Relationship between p53 gene abnormalities and other tumour characteristics in breast-cancer prognosis. Int J Cancer 1996; 69: 135–141.[CrossRef][ISI][Medline]

100. Molino A, Micciolo R, Turazza M et al. Ki-67 immunostaining in 322 primary breast cancers: associations with clinical and pathological variables and prognosis. Int J Cancer 1997; 74: 433–437.[CrossRef][ISI][Medline]

101. Thor AD, Liu S, Moore DH, Edgerton SM. Comparison of mitotic index, in vitro bromodeoxyuridine labeling, and MIB-1 assays to quantitate proliferation in breast cancer. J Clin Oncol 1999; 17: 470–477.[Abstract/Free Full Text]

102. Clahsen PC, van de Velde CJ, Duval C et al. The utility of mitotic index, oestrogen receptor and Ki-67 measurements in the creation of novel prognostic indices for node-negative breast cancer. Eur J Surg Oncol 1999; 25: 356–363.[CrossRef][ISI][Medline]

103. Trihia H, Murray S, Price K et al. Ki-67 expression in breast carcinoma: its association with grading systems, clinical parameters, and other prognostic factors–a surrogate marker? Cancer 2003; 97: 1321–1331.[CrossRef][ISI][Medline]

104. Goodson WH, Moore DH, Ljung BM et al. The prognostic value of proliferation indices: a study with in vivo bromodeoxyuridine and Ki-67. Breast Cancer Res Treat 2000; 59: 113–123.[CrossRef][ISI][Medline]

105. Eifel P, Axelson JA, Costa J et al. National Institutes of Health Consensus Development Conference Statement: adjuvant therapy for breast cancer, November 1–3, 2000. J Natl Cancer Inst 2001; 93: 979–989.[Abstract/Free Full Text]

106. MacGrogan G, Mauriac L, Durand M et al. Primary chemotherapy in breast invasive carcinoma: predictive value of the immunohistochemical detection of hormonal receptors, p53, c-erbB-2, MiB1, pS2 and GST pi. Br J Cancer 1996; 74: 1458–1465.[ISI][Medline]

107. Bottini A, Berruti A, Bersiga A et al. Relationship between tumour shrinkage and reduction in Ki67 expression after primary chemotherapy in human breast cancer. Br J Cancer 2001; 85: 1106–1112.[CrossRef][ISI][Medline]

108. Colleoni M, Zahrieh D, Gelber RD et al. Preoperative systemic treatment: prediction of responsiveness. Breast 2003; 12: 538–542.[CrossRef][ISI][Medline]

109. Assersohn L, Salter J, Powles TJ et al. Studies of the potential utility of Ki67 as a predictive molecular marker of clinical response in primary breast cancer. Breast Cancer Res Treat 2003; 82: 113–123.[CrossRef][ISI][Medline]

110. Chang J, Powles TJ, Allred DC et al. Biologic markers as predictors of clinical outcome from systemic therapy for primary operable breast cancer. J Clin Oncol 1999; 17: 3058–3063.[Abstract/Free Full Text]

111. Iqbal S, Anderson TJ, Marson LP et al. MIB-1 assessments in breast cancers. Breast 2002; 11: 252–256.[CrossRef][ISI][Medline]

112. Moroy T, Geisen C. Cyclin E. Int J Biochem Cell Biol 2004; 36: 1424–1439.[CrossRef][ISI][Medline]

113. Geisen C, Moroy T. The oncogenic activity of cyclin E is not confined to Cdk2 activation alone but relies on several other, distinct functions of the protein. J Biol Chem 2002; 277: 39909–39918.[Abstract/Free Full Text]

114. Gudas JM, Payton M, Thukral S et al. Cyclin E2, a novel G1 cyclin that binds Cdk2 and is aberrantly expressed in human cancers. Mol Cell Biol 1999; 19: 612–622.[Abstract/Free Full Text]

115. Lauper N, Beck AR, Cariou S et al. Cyclin E2: a novel CDK2 partner in the late G1 and S phases of the mammalian cell cycle. Oncogene 1998; 17: 2637–2643.[CrossRef][ISI][Medline]

116. Zariwala M, Liu J, Xiong Y. Cyclin E2, a novel human G1 cyclin and activating partner of CDK2 and CDK3, is induced by viral oncoproteins. Oncogene 1998; 17: 2787–2798.[CrossRef][ISI][Medline]

117. Payton M, Coats S. Cyclin E2, the cycle continues. Int J Biochem Cell Biol 2002; 34: 315–320.[CrossRef][ISI][Medline]

118. Wingate H, Bedrosian I, Akli S, Keyomarsi K. The low molecular weight (LMW) isoforms of cyclin E deregulate the cell cycle of mammary epithelial cells. Cell Cycle 2003; 2: 461–466.[Medline]

119. Akli S, Zheng PJ, Multani AS et al. Tumor-specific low molecular weight forms of cyclin E induce genomic instability and resistance to p21, p27, and antiestrogens in breast cancer. Cancer Res 2004; 64: 3198–3208.[Abstract/Free Full Text]

120. Keyomarsi K, Tucker SL, Buchholz TA et al. Cyclin E and survival in patients with breast cancer. N Engl J Med 2002; 347: 1566–1575.[Abstract/Free Full Text]

121. Loden M, Stighall M, Nielsen NH et al. The cyclin D1 high and cyclin E high subgroups of breast cancer: separate pathways in tumorogenesis based on pattern of genetic aberrations and inactivation of the pRb node. Oncogene 2002; 21: 4680–4690.[CrossRef][ISI][Medline]

122. Payton M, Scully S, Chung G, Coats S. Deregulation of cyclin E2 expression and associated kinase activity in primary breast tumors. Oncogene 2002; 21: 8529–8534.[CrossRef][ISI][Medline]

123. Sutherland RL, Musgrove EA. Cyclins and breast cancer. J Mammary Gland Biol Neoplasia 2004; 9: 95–104.[CrossRef][ISI][Medline]

124. Nielsen NH, Arnerlov C, Emdin SO, Landberg G. Cyclin E overexpression, a negative prognostic factor in breast cancer with strong correlation to oestrogen receptor status. Br J Cancer 1996; 74: 874–880.[ISI][Medline]

125. Kim HK, Park IA, Heo DS et al. Cyclin E overexpression as an independent risk factor of visceral relapse in breast cancer. Eur J Surg Oncol 2001; 27: 464–471.[CrossRef][ISI][Medline]

126. Kuhling H, Alm P, Olsson H et al. Expression of cyclins E, A, and B, and prognosis in lymph node-negative breast cancer. J Pathol 2003; 199: 424–431.[CrossRef][ISI][Medline]

127. Rudolph P, Kuhling H, Alm P et al. Differential prognostic impact of the cyclins E and B in premenopausal and postmenopausal women with lymph node-negative breast cancer. Int J Cancer 2003; 105: 674–680.[CrossRef][ISI][Medline]

128. Han S, Park K, Bae BN et al. Prognostic implication of cyclin E expression and its relationship with cyclin D1 and p27Kip1 expression on tissue microarrays of node negative breast cancer. J Surg Oncol 2003; 83: 241–247.[CrossRef][ISI][Medline]

129. Lindahl T, Landberg G, Ahlgren J et al. Overexpression of cyclin E protein is associated with specific mutation types in the p53 gene and poor survival in human breast cancer. Carcinogenesis 2004; 25: 375–380.[Abstract/Free Full Text]

130. Foulkes WD, Brunet JS, Stefansson IM et al. The prognostic implication of the basal-like (cyclin E high/p27 low/p53+/glomeruloid-microvascular-proliferation+) phenotype of BRCA1-related breast cancer. Cancer Res 2004; 64: 830–835.[Abstract/Free Full Text]

131. Donnellan R, Kleinschmidt I, Chetty R. Cyclin E immunoexpression in breast ductal carcinoma: pathologic correlations and prognostic implications. Hum Pathol 2001; 32: 89–94.[CrossRef][ISI][Medline]

132. Span PN, Tjan-Heijnen VC, Manders P, Beex LV, Sweep CG. Cyclin-E is a strong predictor of endocrine therapy failure in human breast cancer. Oncogene 2003; 22: 4898–4904.[CrossRef][ISI][Medline]

133. Porter PL, Malone KE, Heagerty PJ et al. Expression of cell-cycle regulators p27Kip1 and cyclin E, alone and in combination, correlate with survival in young breast cancer patients. Nat Med 1997; 3: 222–225.[CrossRef][ISI][Medline]

134. Stendahl M, Kronblad A, Ryden L, Emdin S, Bengtsson NO, Landberg G. Cyclin D1 overexpression is a negative predictive factor for tamoxifen response in postmenopausal breast cancer patients. Br J Cancer 2004; 90: 1942–1948.[CrossRef][ISI][Medline]

135. Barnes DM, Gillett CE. Cyclin D1 in breast cancer. Breast Cancer Res Treat 1998; 52: 1–15.[CrossRef][ISI][Medline]

136. Sherr CJ. G1 phase progression: cycling on cue. Cell 1994; 79: 551–555.[CrossRef][ISI][Medline]

137. Gillett C, Smith P, Gregory W et al. Cyclin D1 and prognosis in human breast cancer. Int J Cancer 1996; 69: 92–99.[CrossRef][ISI][Medline]

138. Hwang TS, Han HS, Hong YC, Lee HJ, Paik NS. Prognostic value of combined analysis of cyclin D1 and estrogen receptor status in breast cancer patients. Pathol Int 2003; 53: 74–80.[CrossRef][ISI][Medline]

139. Michalides R, Hageman P, van Tinteren H et al. A clinicopathological study on overexpression of cyclin D1 and of p53 in a series of 248 patients with operable breast cancer. Br J Cancer 1996; 73: 728–734.[ISI][Medline]

140. Pelosio P, Barbareschi M, Bonoldi E et al. Clinical significance of cyclin D1 expression in patients with node-positive breast carcinoma treated with adjuvant therapy. Ann Oncol 1996; 7: 695–703.[Abstract]

141. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev 1995; 9: 1149–1163.[CrossRef][ISI][Medline]

142. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 1999; 13: 1501–1512.[Free Full Text]

143. Slingerland J, Pagano M. Regulation of the cdk inhibitor p27 and its deregulation in cancer. J Cell Physiol 2000; 183: 10–17.[CrossRef][ISI][Medline]

144. Massague J, Blain SW, Lo RS. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 2000; 103: 295–309.[CrossRef][ISI][Medline]

145. Alkarain A, Jordan R, Slingerland J. p27 deregulation in breast cancer: prognostic significance and implications for therapy. J Mammary Gland Biol Neoplasia 2004; 9: 67–80.[CrossRef][ISI][Medline]

146. Tan P, Cady B, Wanner M et al. The cell cycle inhibitor p27 is an independent prognostic marker in small (T1a,b) invasive breast carcinomas. Cancer Res 1997; 57: 1259–1263.[Abstract]

147. Wu J, Shen ZZ, Lu JS et al. Prognostic role of p27Kip1 and apoptosis in human breast cancer. Br J Cancer 1999; 79: 1572–1578.[CrossRef][ISI][Medline]

148. Tsuchiya A, Zhang GJ, Kanno M. Prognostic impact of cyclin-dependent kinase inhibitor p27kip1 in node-positive breast cancer. J Surg Oncol 1999; 70: 230–234.[CrossRef][ISI][Medline]

149. Chappuis PO, Kapusta L, Begin LR et al. Germline BRCA1/2 mutations and p27(Kip1) protein levels independently predict outcome after breast cancer. J Clin Oncol 2000; 18: 4045–4052.[Abstract/Free Full Text]

150. Catzavelos C, Bhattacharya N, Ung YC et al. Decreased levels of the cell-cycle inhibitor p27Kip1 protein: prognostic implications in primary breast cancer. Nat Med 1997; 3: 227–230.[CrossRef][ISI][Medline]

151. Gillett CE, Smith P, Peters G, Lu X, Barnes DM. Cyclin-dependent kinase inhibitor p27Kip1 expression and interaction with other cell cycle-associated proteins in mammary carcinoma. J Pathol 1999; 187: 200–206.[CrossRef][ISI][Medline]

152. Barbareschi M, van Tinteren H, Mauri FA et al. p27(kip1) expression in breast carcinomas: an immunohistochemical study on 512 patients with long-term follow-up. Int J Cancer 2000; 89: 236–241.[CrossRef][ISI][Medline]

153. Leivonen M, Nordling S, Lundin J et al. p27 expression correlates with short-term, but not with long-term prognosis in breast cancer. Breast Cancer Res Treat 2001; 67: 15–22.[CrossRef][ISI][Medline]

154. Nohara T, Ryo T, Iwamoto S, Gon G et al. Expression of cell-cycle regulator p27 is correlated to the prognosis and ER expression in breast carcinoma patients. Oncology 2001; 60: 94–100.[CrossRef][ISI][Medline]

155. Barnes A, Pinder SE, Bell JA et al. Expression of p27kip1 in breast cancer and its prognostic significance. J Pathol 2003; 201: 451–459.[CrossRef][ISI][Medline]

156. Lau R, Grimson R, Sansome C et al. Low levels of cell cycle inhibitor p27kip1 combined with high levels of Ki-67 predict shortened disease-free survival in T1 and T2 invasive breast carcinomas. Int J Oncol 2001; 18: 17–23.[ISI][Medline]

157. Fredersdorf S, Burns J, Milne AM et al. High level expression of p27(kip1) and cyclin D1 in some human breast cancer cells: inverse correlation between the expression of p27(kip1) and degree of malignancy in human breast and colorectal cancers. Proc Natl Acad Sci USA 1997; 94: 6380–6385.[Abstract/Free Full Text]

158. Newman L, Xia W, Yang HY et al. Correlation of p27 protein expression with HER-2/neu expression in breast cancer. Mol Carcinog 2001; 30: 169–175.[CrossRef][ISI][Medline]

159. Spataro VJ, Litman H, Viale G et al. Decreased immunoreactivity for p27 protein in patients with early-stage breast carcinoma is correlated with HER-2/neu overexpression and with benefit from one course of perioperative chemotherapy in patients with negative lymph node status: results from International Breast Cancer Study Group Trial V. Cancer 2003; 97: 1591–1600.[CrossRef][ISI][Medline]

160. Loden M, Perris F, Nielsen NH et al. C-erbB2, p27 and G1/S aberrations in human primary breast cancer. Anticancer Res 2003; 23 (3A): 2053–2061.[ISI][Medline]

161. Ye D, Mendelsohn J, Fan Z. Augmentation of a humanized anti-HER2 mAb 4D5 induced growth inhibition by a human-mouse chimeric anti-EGF receptor mAb C225. Oncogene 1999; 18: 731–738.[CrossRef][ISI][Medline]

162. Nahta R, Takahashi T, Ueno NT et al. p27(kip1) down-regulation is associated with trastuzumab resistance in breast cancer cells. Cancer Res 2004; 64: 3981–3986.[Abstract/Free Full Text]

163. Reed W, Florems VA, Holm R et al. Elevated levels of p27, p21 and cyclin D1 correlate with positive oestrogen and progesterone receptor status in node-negative breast carcinoma patients. Virchows Arch 1999; 435: 116–124.[CrossRef][ISI][Medline]

164. Pohl G, Rudas M, Dietze O et al. High p27Kip1 expression predicts superior relapse-free and overall survival for premenopausal women with early-stage breast cancer receiving adjuvant treatment with tamoxifen plus goserelin. J Clin Oncol 2003; 21: 3594–3600.[Abstract/Free Full Text]

165. Dupont J, Karas M, LeRoith D. The cyclin-dependent kinase inhibitor p21CIP/WAF is a positive regulator of insulin-like growth factor I-induced cell proliferation in MCF-7 human breast cancer cells. J Biol Chem 2003; 278: 37256–37264.[Abstract/Free Full Text]

166. Giles KM, Daly JM, Beveridge DJ et al. The 3'-untranslated region of p21WAF1 mRNA is a composite cis-acting sequence bound by RNA-binding proteins from breast cancer cells, including HuR and poly(C)-binding protein. J Biol Chem 2003; 278: 2937–2946.[Abstract/Free Full Text]

167. Zhou BP, Liao Y, Xia W, Spohn B et al. Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells. Nat Cell Biol 2001; 3: 245–252.[CrossRef][ISI][Medline]

168. Skildum AJ, Mukherjee S, Conrad SE. The cyclin-dependent kinase inhibitor p21WAF1/Cip1 is an antiestrogen-regulated inhibitor of Cdk4 in human breast cancer cells. J Biol Chem 2002; 277: 5145–5152.[Abstract/Free Full Text]

169. Michels JJ, Duigou F, Marnay J et al. Flow cytometry and quantitative immunohistochemical study of cell cycle regulation proteins in invasive breast carcinoma: prognostic significance. Cancer 2003; 97: 1376–1386.[CrossRef][ISI][Medline]

170. Caffo O, Doglioni C, Veronese S et al. Prognostic value of p21(WAF1) and p53 expression in breast carcinoma: an immunohistochemical study in 261 patients with long-term follow-up. Clin Cancer Res 1996; 2: 1591–1599.[Abstract/Free Full Text]

171. Jiang M, Shao ZM, Wu J et al. p21/waf1/cip1 and mdm-2 expression in breast carcinoma patients as related to prognosis. Int J Cancer 1997; 74: 529–534.[CrossRef][ISI][Medline]

172. Wakasugi E, Kobayashi T, Tamaki Y et al. p21(Waf1/Cip1) and p53 protein expression in breast cancer. Am J Clin Pathol 1997; 107: 684–691.[ISI][Medline]

173. Diab SG, Yu YY, Hilsenbeck SG et al. WAF1/CIP1 protein expression in human breast tumors. Breast Cancer Res Treat 1997; 43: 99–103.[CrossRef][ISI][Medline]

174. Mathoulin-Portier MP, Viens P, Cowen D et al. Prognostic value of simultaneous expression of p21 and mdm2 in breast carcinomas treated by adjuvant chemotherapy with antracyclin. Oncol Rep 2000; 7: 675–680.[ISI][Medline]

175. Thor AD, Liu S, Moore DH et al. p(21WAF1/CIP1) expression in breast cancers: associations with p53 and outcome. Breast Cancer Res Treat 2000; 61: 33–43.[CrossRef][ISI][Medline]

176. Ceccarelli C, Santini D, Chieco P et al. Quantitative p21(waf-1)/p53 immunohistochemical analysis defines groups of primary invasive breast carcinomas with different prognostic indicators. Int J Cancer 2001; 95: 128–134.[CrossRef][ISI][Medline]

177. Gohring UJ, Bersch A, Becker M et al. p21(waf) correlates with DNA replication but not with prognosis in invasive breast cancer. J Clin Pathol 2001; 54: 866–870.[Abstract/Free Full Text]

178. Domagala W, Welcker M, Chosia M et al. p21/WAF1/Cip1 expression in invasive ductal breast carcinoma: relationship to p53, proliferation rate, and survival at 5 years. Virchows Arch 2001; 439: 132–140.[CrossRef][Medline]

179. Yang W, Klos KS, Zhou X et al. ErbB2 overexpression in human breast carcinoma is correlated with p21Cip1 up-regulation and tyrosine-15 hyperphosphorylation of p34Cdc2: poor responsiveness to chemotherapy with cyclophoshamide methotrexate, and 5-fluorouracil is associated with Erb2 overexpression and with p21Cip1 overexpression. Cancer 2003; 98: 1123–1130.[CrossRef][ISI][Medline]

180. Hsiang YH, Wu HY, Liu LF. Proliferation-dependent regulation of DNA topoisomerase II in cultured human cells. Cancer Res 1988; 48: 3230–3235.[Abstract]

181. Constantinou A, Henning-Chubb C, Huberman E. Novobiocin- and phorbol-12-myristate-13-acetate-induced differentiation of human leukemia cells associated with a reduction in topoisomerase II activity. Cancer Res 1989; 49: 1110–1117.[Abstract]

182. Zwelling LA, Hinds M, Chan D et al. Phorbol ester effects on topoisomerase II activity and gene expression in HL-60 human leukemia cells with different proclivities toward monocytoid differentiation. Cancer Res 1990; 50: 7116–7122.[Abstract]

183. Jarvinen TA, Tanner M, Rantanen V et al. Amplification and deletion of topoisomerase IIalpha associate with ErbB-2 amplification and affect sensitivity to topoisomerase II inhibitor doxorubicin in breast cancer. Am J Pathol 2000; 156: 839–847.[Abstract/Free Full Text]

184. Knoop A KH, Balslev E et al. Topoisomerase II alpha (Top2A) alteration as a predictive marker for epirubicin sensitivity in 805 high risk breast cancer patients. A randomised DBCG trial (DBCG89D). Eur J Cancer 2003; Suppl 1: S202 (Abstr 674).

185. Bofin AM, Ytterhus B, Hagmar BM. TOP2A and HER-2 gene amplification in fine needle aspirates from breast carcinomas. Cytopathology 2003; 14: 314–319.[CrossRef][ISI][Medline]

186. Jarvinen TA, Tanner M, Barlund M et al. Characterization of topoisomerase II alpha gene amplification and deletion in breast cancer. Genes Chromosomes Cancer 1999; 26: 142–150.[CrossRef][ISI][Medline]

187. Jarvinen TA, Kononen J, Pelto-Huikko M, Isola J. Expression of topoisomerase II alpha is associated with rapid cell proliferation, aneuploidy, and c-erbB2 overexpression in breast cancer. Am J Pathol 1996; 148: 2073–2082.[Abstract]

188. Coon JS, Marcus E, Gupta-Burt S et al. Amplification and overexpression of topoisomerase II alpha predict response to anthracycline-based therapy in locally advanced breast cancer. Clin Cancer Res 2002; 8: 1061–1067.[Abstract/Free Full Text]

189. Mueller RE, Parkes RK, Andrulis I, O'Malley FP. Amplification of the TOP2A gene does not predict high levels of topoisomerase II alpha protein in human breast tumor samples. Genes Chromosomes Cancer 2004; 39: 288–297.[CrossRef][ISI][Medline]

190. Durbecq V, Desmed C, Paesmans M et al. Correlation between topoisomerase-II alpha gene amplification and protein expression in HER-2 amplified breast cancer. Int J Oncol 2004; 25: 1473–1479.[ISI][Medline]

191. Park K, Kim J, Lim S, Han S. Topoisomerase II-alpha (topoII) and HER2 amplification in breast cancers and response to preoperative doxorubicin chemotherapy. Eur J Cancer 2003; 39: 631–634.[CrossRef][ISI][Medline]

192. Depowski PL, Rosenthal SI, Brien TP et al. Topoisomerase II alpha expression in breast cancer: correlation with outcome variables. Mod Pathol 2000; 13: 542–547.[CrossRef][ISI][Medline]

193. Rudolph P, Olsson H, Bonatz G et al. Correlation between p53, c-erbB-2, and topoisomerase II alpha expression, DNA ploidy, hormonal receptor status and proliferation in 356 node-negative breast carcinomas: prognostic implications. J Pathol 1999; 187: 207–216.[CrossRef][ISI][Medline]

194. MacGrogan G, Rudolph P, Mascarel Id I et al. DNA topoisomerase II alpha expression and the response to primary chemotherapy in breast cancer. Br J Cancer 2003; 89: 666–671.[CrossRef][ISI][Medline]

195. Campiglio M, Somenzi G, Olgiati C et al. Role of proliferation in HER2 status predicted response to doxorubicin. Int J Cancer 2003; 105: 568–573.[CrossRef][ISI][Medline]

196. Rudolph P, MacGrogan G, Bonichon F et al. Prognostic significance of Ki-67 and topoisomerase II alpha expression in infiltrating ductal carcinoma of the breast. A multivariate analysis of 863 cases. Breast Cancer Res Treat 1999; 55: 61–71.[CrossRef][ISI][Medline]

197. Di Leo A, Gancberg D, Larsimont D et al. HER-2 amplification and topoisomerase II alpha gene aberrations as predictive markers in node-positive breast cancer patients randomly treated either with an anthracycline-based therapy or with cyclophosphamide, methotrexate, and 5-fluorouracil. Clin Cancer Res 2002; 8: 1107–1116.[Abstract/Free Full Text]

198. Henderson IC, Patek AJ. The relationship between prognostic and predictive factors in the management of breast cancer. Breast Cancer Res Treat 1998; 52: 261–288.[CrossRef][ISI][Medline]

199. van Diest PJ, Michalides RJ, Jannink L et al. Cyclin D1 expression in invasive breast cancer. Correlations and prognostic value. Am J Pathol 1997; 150: 705–11.[Abstract]

200. Kenny FS, Hui R, Musgrove EA et al. Overexpression of cyclin D1 messenger RNA predicts for poor prognosis in estrogen receptor-positive breast cancer. Clin Cancer Res 1999; 5: 2069–2076.[Abstract/Free Full Text]

201. Umekita Y, Ohi Y, Sagara Y, Yoshida H. Overexpression of cyclinD1 predicts for poor prognosis in estrogen receptor-negative breast cancer patients. Int J Cancer 2002; 98: 415–418.[CrossRef][ISI][Medline]

202. Pellikainen MJ, Pekola TT, Ropponen KM et al. p21WAF1 expression in invasive breast cancer and its association with p53, AP-2, cell proliferation, and prognosis. J Clin Pathol 2003; 56: 214–220.[Abstract/Free Full Text]





This Article
Abstract
Full Text (PDF)
All Versions of this Article:
16/11/1723    most recent
mdi352v1
E-letters: Submit a response
Alert me when this article is cited
Alert me when E-letters are posted
Alert me if a correction is posted
Services
Email this article to a friend
Similar articles in this journal
Similar articles in ISI Web of Science
Similar articles in PubMed
Alert me to new issues of the journal
Add to My Personal Archive
Download to citation manager
Search for citing articles in:
ISI Web of Science (1)
Disclaimer
Request Permissions
Google Scholar
Articles by Colozza, M.
Articles by Piccart, M. J.
PubMed
PubMed Citation
Articles by Colozza, M.
Articles by Piccart, M. J.