当前位置: 首页 > 期刊 > 《临床肿瘤学》 > 2005年第10期 > 正文
编号:11332210
Proliferation Marker Ki-67 in Early Breast Cancer
http://www.100md.com 《临床肿瘤学》
     the Academic Department of Biochemistry and Breast Unit, Royal Marsden Hospital, London, UK

    ABSTRACT

    Molecular markers have been extensively investigated with a view to providing early and accurate information on long-term outcome and prediction of response to treatment of early breast cancer. Proliferation is a key feature of the progression of tumors and is now widely estimated by the immunohistochemical assessment of the nuclear antigen Ki-67. The expression of Ki-67 correlates with other measurements of proliferation, including S-phase and bromodeoxyuridine uptake. High Ki-67 is a sign of poor prognosis associated with a good chance of clinical response to chemotherapy, but its independent significance is modest and does not merit measurements in most routine clinical scenarios. However, its application as a pharmacodynamic intermediate marker of the effectiveness of medical therapy holds great promise for rapid evaluation of new drugs.

    INTRODUCTION

    Early breast cancer (EBC) has a highly variable prognosis and benefit from available therapies is unpredictable for the individual patient. Key factors such as tumor size, histological grade, vascular invasion, and nodal status are helpful, but increasing attention is being paid to the molecular features of the tumor. Estrogen receptor (ER) and Her-2–neu are now well established as predictive factors for treatment response and prognosis. Although not considered an obligatory marker, Ki-67 is also frequently measured both as a static marker of proliferative activity and, by making multiple measurements during treatment, as a possible dynamic intermediate or surrogate marker of treatment efficacy. We provide here an overview of the basic molecular biology of Ki-67 and its prognostic and predictive value at presentation of disease. In particular, we have focused on the evidence supporting the use of change in Ki-67 as an early predictor of treatment efficacy and as a prognostic factor of long-term outcome. The review is based on published evidence, identifying articles with searches of PubMed (key words: "Ki-67," "anti-Ki-67," "breast cancer," "prognostic," "proliferation") as well as references from relevant articles and from the authors’ personal experience. Other than when stated, we refer to Ki-67 as the presence of the Ki-67 antigen determined by immunohistochemistry.

    BIOLOGY OF Ki-67

    Nature, Sequence, and Function

    Ki-67 was identified by Gerdes et al in 19911 as a nuclear nonhistone protein, shortly after the corresponding antibody was described by the same group2 in the city of Kiel (hence "Ki") after immunization of mice with the Hodgkin’s lymphoma cell line L428 (67 refers to the clone number on the 96-well plate in which it was found). The absence of Ki-67 in quiescent cells and its universal expression in proliferating tissues created great interest on its potential role as a marker of cell proliferation. A large number of studies (see references below for correlation with other proliferation markers) have confirmed this feature, and the expression of Ki-67 in resting cells has rarely been reported.3

    The Ki-67 gene is on the long arm of human chromosome 10 (10q25).4 In 1993, Schluter et al published the complete sequence of the cDNA encoding for the protein.5 Two alternative mRNA species resulting from alternative splicing encode two isoforms of the protein. The "large" Ki-67 protein isoform has a calculated molecular mass of 359 kD, and the "small" isoform, a mass of 320 kD. The presence or absence of the sequence encoded by exon 7 of the gene differentiates one isoform from the other. The most outstanding feature when the sequence is analyzed is the presence of 16 repeated elements, the Ki-67 repeats," in the large exon 13 that makes up 70% and 79% of the open reading frame of the large and small species, respectively. These are concatenated sequences depicting an important similarity with 43% to 62% identical amino acids. Within these repeats, a sequence of 22 amino acids, the so-called Ki-67 motif, can be found. This motif is highly conserved between species and includes the epitope (FKEL) targeted by the original Ki-67 antibody. There is some evidence derived from murine and human cell lines that more postsplicing variants may exist with a consequent larger range of protein isoforms.6

    The expression of Ki-67 varies in intensity throughout the cell cycle, and this has raised concern that it could lead to a misclassification of cycling cells as resting ones.3 Overall, evidence indicates that levels of Ki-67 are low during G1- and early S-phase and progressively increase to reach a maximum during mitosis. A rapid decrease in expression starts during anaphase and telophase.7 Some authors have found that the degree of expression in G1 can be minimal8-10 and may represent a handicap to Ki-67’s accuracy in identifying cells in this phase. This irregular presence of Ki-67 in G1 has been explained as the result of the differences between G1-phase in a previously dormant cell and G1 immediately following a previous cycle8 or even as a marker of different conditions of growth.9

    The half-life of Ki-67 protein has been estimated at around 60 to 90 minutes.10,11 Differences of expression during the cell cycle do not seem to be due to the accumulation of nondegraded protein; rather, they seem largely to reflect variable de-novo synthesis.

    The cellular appearance and location of the Ki-67 protein throughout the cell cycle is not homogeneous. During early G1, it is found as generally weakly staining discrete foci throughout karyoplasm.12 It progressively condenses during late G1 in larger perinucleolar granules.13,14 During S and G2 phases, it is mainly found associated with the nucleolar region in larger foci as well as with some heterochromatin regions. When the nuclear membrane disrupts during early mitosis, Ki-67 shows an intense expression associated with the surface of condensed chromosomes in the cytoplasm. This intensity rapidly disappears in anaphase-telophase.

    Despite all this information about the nature, location, and sequence of Ki-67 protein, there is little known of its function beyond its being a protein phosphorylated via serine and threonine11 with a critical role in cell division. This has been concluded from the arrest of cell proliferation when Ki-67 is blocked either by microinjection of blocking antibodies13 or by inhibition of dephosphorylation.11

    Determination of Ki-67

    The original Ki-67 monoclonal antibody, when used for immunostaining was initially reported to stain proliferating cells in unfixed tissues but not formalin-fixed paraffin-embedded samples. In 1992, Cattoretti et al reported better success in staining Ki-67 in paraffin-embedded samples after development of the new antibodies MIB-1 and MIB-3. These antibodies seem to produce results equivalent to the original Ki-67 antibody targeting the same epitope (contained in the Ki-67 motif), as demonstrated by identical bands in Western immunoblots.15 Staining by MIB-1 and -3 of formalin-fixed paraffin-embedded samples is greatly enhanced by antigen retrieval (most frequently by microwave heating).16 Although several antibodies are now commercially available to stain Ki-67 in fresh and paraffin-embedded tissue, MIB-1 is the most widely used in recent studies (Fig 1). Ki-67 expression is usually estimated as the percentage of tumor cells positively stained by the antibody, with nuclear staining being the most common criterion of positivity. Currently, there are few data on the between-batch or between-laboratory variability in these measurements and whether the application of image analysis techniques (which have difficulties in tissues with heterogeneous cell populations such as breast cancer) will enhance the precision of assessment.

    Ki-67 and Other Markers of Proliferation

    Many studies have investigated the correlation between Ki-67 and other well-known markers of proliferation. A strong correlation has been found in most.

    S-phase fraction as measured by flow cytometry,17-25 mitotic index,19,20,26-30 tyrosine kinase,20 and in vivo uptake of bromodeoxyuridine (a laborious but highly reliable procedure considered by many to be the gold standard of cell proliferation measurements)26,29,31-33 have each been regularly reported to correlate well with Ki-67.

    The correlation of Ki-67 with proliferating cell nuclear antigen has been reported as positive,34 but weak,31 and the multiplicity of functions developed by proliferating cell nuclear antigen may account for this discrepancy.35 Studies of the correlation with DNA ploidy are also conflicting,20,21,33 and no correlation has been found with thymidilate synthase.28 A recently described marker of cell proliferation, the minichromosome maintenance protein 2 (Mcm-2), showed a strong correlation with Ki-67 although it seemed to be more sensitive in detecting cycling cells.36

    Ki-67 in the Normal Breast

    Few studies have reported results for Ki-67 on normal breast tissue. In samples from normal breast37,38 or in normal epithelium adjacent to fibroadenomas39 it is expressed at a very low level (< 3% of cells). One of the most interesting features found in these studies is the absence of Ki-67 positivity in the ER-positive population38 (ie, only ER-negative cells are proliferative in normal breast). This feature is lost in breast cancer where both markers are present in a higher percentage of cells and these populations overlap. There seems to be a progressively changing pattern from normal to cancerous tissue, with an intermediate degree of coincidence of both markers in noninvasive precancerous lesions.40

    Ki-67 and Other Biologic Markers in Breast Cancer

    Apoptosis, or programmed cell death, provides the major counterbalancing determinant of tumor growth to proliferation and has been consistently reported to be positively correlated with Ki-67,41 as shown in Figure 2. An index based on the Ki-67–apoptosis ratio (the cell turnover or growth index) has been developed to approximate the contribution that these two factors may make to tumor growth. While it is clear that this cannot accurately reflect growth dynamics, it may have utility as an early marker of response to treatment in primary therapy of breast cancer,42-44 though this remains to be rigorously demonstrated.

    Bcl-2, an antiapoptotic protein, has been reported as being inversely correlated with baseline Ki-67.45 The oncogene p53 is frequently mutated or overexpressed in breast cancer, and those tumors presenting these changes show higher rates of proliferation as measured by Ki-67.19,31,45-48 Several studies have addressed the relation between Ki-67 and Her-2 expression, and although there seems to be a positive correlation,28,45,49 it is not always well-defined.22,50 The relationship with epidermal growth factor is similarly conflicting.22,45,50,51 ER status has been widely reported as being inversely correlated with Ki-67, with the higher rates of ER positivity shown in the least proliferating tumors17,19,20,31,34,45,46,52,53 and only a few studies have contradicted this.28,29,48

    Of the commonly used pathological features of breast cancer probably the most robustly related to Ki-67 is histological grade with virtually no studies refuting this positive correlation.19-21,28,30,31,33,46,48,53-55 This is to be expected given that mitotic index is one of the three components of grade. Tumor size is not consistently linked to Ki-67 scores with some authors finding a positive relationship31,34,46,52,55 but others not.17,19,22,54

    The strongest prognostic factor in EBC, lymph node status, has been intensively studied with regard to its correlation with Ki-67 in an attempt to find an easy marker of nodal involvement that would avoid unnecessary axillary surgery. In studies with more than 200 patients, there seems to be more evidence in favor46,52,56 than against54 a positive correlation (see below). The number of smaller studies favoring a lack of correlation is large.19,20,29,45,48 This may reflect the size of studies and/or the relative weakness in the relationship.

    Ki-67 BASELINE MEASUREMENT: PROGNOSTIC VALUE

    The search for useful tools to discriminate prognostic subgroups in patients affected by early breast cancer, and in particular, as noted above, in those without lymph node involvement, has led to an extensive study of Ki-67 in this setting. Data from 40 studies involving more than 11,000 patients have been analyzed. Those studies with a sample size greater than 200 patients have been summarized in Table 1.17,18,26-28,34,46,52,54,57-65

    In this Table, the studies are presented as statistically significant (P, yellow box) or nonsignificant (N, pink box) for the relation between Ki-67 and clinical outcome. Results are considered in terms of disease-free survival (including metastasis free and local relapse free survival) and overall survival (including disease-specific survival) and the data are shown in three groups made according to the lymph node status (negative, positive, and mixed population). The statistical analysis is specified as univariate or multivariate.

    There is strong evidence from these large studies, of the ability of Ki-67 to discriminate between good and bad prognostic groups in the node-negative population when considered as a single variable. While this significance is lost in the multivariate analyses in two of the studies,26,57 in most, it is maintained. In this node-negative group of patients, the large majority of studies failing to report a significant prognostic value of Ki-67 involved patient populations smaller than 200 (not depicted in the Table). In the lymph node–positive and mixed population, the results are less clear, but positive studies predominate.

    There are difficulties in assessing the possible prognostic uses of Ki-67 since virtually all of these studies are retrospective and involve a highly heterogeneous population. The use of different antibodies, techniques and scoring protocols without a standard minimum number of cells to be counted may account for some of the differences between studies. The lack of an optimal cutoff point for the definition of prognostic subgroups is probably the main obstacle to a synthesis of the results for extrapolation to clinical practice. Overall it seems that Ki-67 does carry independent prognostic significance from routinely measured pathological features but this is modest and does not merit routine inclusion in the work-up of primary breast cancer.

    Ki-67 AND PREDICTION OF TREATMENT RESPONSE

    The association of Ki-67 and response to medical treatment is clearer. Five out of six studies reporting the value of Ki-67 to predict response (clinical and/or pathological) to chemotherapy in early or locally advanced breast cancer found that higher Ki-67 was associated with better response66-70 but one found no association.71 In contrast no significant relationship between Ki-67 score and response to treatment has been reported for neoadjuvant endocrine treatment.44,72,73 This may explain why inconsistent results have been reported when mixed chemotherapy and hormonal therapy have been used, with both negative24,45 and positive74 results reported.

    It is important to note that while high scores of Ki-67 are associated with higher chance of response to chemotherapy, high Ki-67 is a marker of poor prognosis overall. However, good clinical response, particularly pathological complete response, is associated with good long-term prognosis. This suggests that stratification according to Ki-67 levels might improve the prognostic significance of clinical response in neoadjuvant chemotherapy. To our knowledge this approach has not been undertaken.

    CHANGES IN Ki-67 THROUGH TREATMENT: CLINICAL SIGNIFICANCE

    The availability of minimally invasive tumor sampling techniques in EBC (ie, fine-needle aspirate, core-cut biopsy) has prompted investigators to study whether changes in Ki-67 score occur after a variable time of treatment and can provide a better predictive or prognostic value than baseline measurements. If so, these early changes might provide valuable intermediate markers of treatment benefit, particularly in relation to drug development.

    Studies with MCF-7 xenograft tumors in nude mice with estrogen deprivation75 or antiestrogen hormonal treatments76 provide encouraging proof of principle. Each of these treatments results in a rapid decrease of Ki-67 within 1 week of treatment. The study by Johnston et al demonstrated a complex relation between Ki-67 decrease, changes in apoptosis, and tumor regression following estrogen withdrawal compared with tamoxifen (Fig 3). Estrogen withdrawal induced a profound decrease in Ki-67 (approximately five-fold), accompanied by an increase in apoptosis (approximately four-fold), and this resulted in tumor regression. With tamoxifen, lesser reductions in Ki-67 (approximately two-fold) and increases in apoptosis (approximately three-fold) were seen, and these were associated with no more than stabilization of tumor size. These findings suggest that yet smaller changes in Ki-67 and apoptosis could lead to slowing of tumor growth but still be associated with progressive disease. The implications of these findings for clinical studies are discussed below.

    Before assuming that change in any biomarker is due to an intervention, the intrinsic variability of the marker without the mediating intervention should be assessed. Few reports have addressed this issue. Our studies on the reproducibility of Ki-67 measurements in core-cut biopsies suggest that a change in Ki-67 score of at least 32% to 50% between two determinations is required to consider the difference statistically different for an individual patient, and attributable to treatment effect.77,78 These individual requirements do not apply in the same way to populations but are important to take into consideration in the statistical powering of these studies. Further data on the variability in the measurements of Ki-67 are provided in the placebo arms of several short-term presurgical studies,79-82 and these largely support our estimates of variability. However, different biopsy sites, staining, and counting procedures will affect this variability.

    In the clinic, effective hormonal treatments suppress Ki-67 levels in both short-term (< 6 weeks)42,79-87 and long-term (eg, 12 weeks) studies.44,72,73,88-90 This has also been shown with withdrawal of hormone replacement therapy.91 As would be expected, these changes are only seen in hormone receptor–positive tumors.80-82,91,92 Reduced Ki-67 levels with hormonal therapy are illustrated in Figure 4, which shows the substantial falls in Ki-67 at 2 weeks and 12 weeks in 56 patients treated with anastrozole before surgery.93 At 2 weeks, only four patients did not show a reduction in Ki-67 levels. The geometric means of reduction were 76% and 82% at 2 and 12 weeks, respectively. There were some patients who showed increases in Ki-67 between 2 and 12 weeks, and it is possible that this is an early indication of resistance to the therapy. It is, however, difficult to distinguish these relatively modest increases in individual patients from the between-sample variability that is discussed above.

    The reduction in Ki-67 with endocrine therapy probably reflects the well-described cytostatic effect of the drugs.94 The changes in Ki-67 occur early and well before significant tumor regression is seen. This suggests that treatment selection of a less proliferative cell population probably plays little part in the changes.

    Ki-67 decreases also occur with chemotherapy and mixed chemohormonal treatments.24,45,66,67,70,74,77,78,95,96 We have found only one study failing to show this,69 and our own work suggests this may be related to the timing of the second biopsy.97 The reduced proliferation with chemotherapy may be at least partly due to increased apoptosis in actively proliferating cells98 such that the residual population would be enriched for Ki-67–negative cells. The relationship of decrease in Ki-67 during cytotoxic chemotherapy over 2 to 3 months with long-term outcome has been addressed in two studies,45,95 which found in a multivariate analysis that a decrease in Ki-67 of more than 25% and a residual score less than 10% each predicted a longer disease-free survival.

    Early changes in Ki-67 have also been found to correlate positively with clinical and/or pathologic response in EBC with both hormone therapy and chemotherapy.24,45,66,70,72,77,88-90,95 In these studies, the timing of the second biopsy varied. The relationship to clinical and/or pathologic response within 2 weeks is particularly useful since these changes preceded clinical response, while the changes at 12 to 16 weeks are usually concordant with clinical response rather than predictive.

    This issue has been recently supported by a report on the results of the IMPACT trial.93,99,100 This trial compared 12 weeks of neoadjuvant treatment with anastrozole or tamoxifen or the combination of both drugs in postmenopausal women with ER-positive EBC, followed after surgery by the same regimen as adjuvant treatment. Ki-67 decrease as determined in a biopsy taken 2 weeks after initiation of treatment predicted the better long-term outcome seen in the parallel adjuvant trial with anastrozole.101

    The possibility that changes in Ki-67 might predict long-term efficacy (after 10-14 days of treatment) has opened a new scenario for clinical research in EBC. This so-called "short-term preoperative" setting refers to the time between diagnosis of primary operable EBC and the surgery itself, an interval when treatment is not usually administered. Thus, drugs with established safety and low toxicity may be tested and compared against placebo using a decrease in Ki-67 as the primary end point. This model has been exploited in the assessment of several treatments; these have been mainly hormonal,42,79-84 but have more recently included inhibitors of signal transduction pathways.102,103 Most of these showed a significant drop in Ki-67 in the treatment arms compared with placebo or no treatment.

    Different treatments have also been compared in this short-term setting to obtain rapid information on potential differences in efficacy.42,81,82,87 Comparative studies between drugs require their having similar pharmacokinetics or the use of schedules to compensate for different pharmacokinetics; they should also have a mechanism of action that similarly affects proliferation (or another end point such as apoptosis). These approaches could also allow the differential effectiveness of drugs on subgroups of patients to be assessed leading to the identification of targets to select optimal therapy. For example, a recent report by Ellis et al found differences in the Ki-67 response to letrozole and tamoxifen depending on whether tumors overexpressed Her-1 and/or Her-2,89 and these results paralleled differential clinical response in the same subgroups.104 These differences had been previously suggested in a pooled analyses of several smaller studies in the short-term preoperative setting.105 The effect on Ki-67 of Her-2–targetted therapy combined with endocrine therapy would seem to be a rational approach to evaluating efficacy.

    While the above rationale and examples of the use of early changes in Ki-67 suggest that there will be a growing interest in its use as an intermediate marker of benefit, there remain challenges in the handling of the derived data that need to be met to allow its widespread use. One complication is the non-normal distribution of the data, which logarithmic transformation may also not normalize.89 It is also not clear that the proportional reduction in Ki-67 is the most relevant measure for predicting outcome. The arguments made above suggest that proportional reduction may be an appropriate parameter for predicting benefit in the adjuvant setting, but that the residual (on-treatment) level of Ki-67 may be a better predictor of response and/or absolute long-term outcome since this is more likely to relate to the growth rate of the persistent disease. Currently available and future data sets should examine these possibilities.

    SUMMARY AND CONCLUSIONS

    The Ki-67 protein, measured with immunohistochemical techniques, is a useful marker of cell proliferation. The suggested lack of accuracy to identify some cells entering the cell cycle in early G0 has not substantially affected the ability of Ki-67 to identify the proliferating population of cells in comparison with other markers of proliferation. Compared with these other markers, Ki-67 staining is easy to perform, economical, and more reproducible. Correlation between Ki-67 mRNA levels and the presence of the protein identified by immunohistochemistry has not been yet fully studied106; this will be required to allow appropriate interpretation of Ki-67 expression as determined with new diagnostic tools, including expression microarrays.

    Definitions of cut-offs for Ki-67 vary and hamper comparisons between studies: it may be more helpful to consider Ki-67 as a continuous variable. In multivariate analyses, Ki-67 retains prognostic significance in lymph node–negative patients, but its clinical utility seems slight. Instead, a key role for Ki-67 in EBC may relate to the measurable changes in its expression during treatment. The lack of a close agreement between decreases in Ki-67 and tumor response in some clinical trials possibly results from the need for major reductions in proliferation and/or increases in apoptosis to lead to the regression of a fast-growing tumor. These decreases in proliferation may nonetheless be indicative of benefit from the treatment. There is now a strong rationale for the future design of trials using short-term changes in Ki-67 as a marker of treatment efficacy. However, for analysis of Ki-67 to reach its potential either as a baseline or as a pharmacodynamic measurement, greater attention will need to be paid to standardized methodologies, approaches to scoring, statistical analysis, and between-laboratory quality assessment.

    Authors' Disclosures of Potential Conflicts of Interest

    The authors indicated no potential conflicts of interest.

    Acknowledgment

    We thank our numerous clinical and laboratory colleagues who have contributed to the studies we have conducted leading to the development of this review.

    NOTES

    Authors' disclosures of potential conflicts of interest are found at the end of this article.

    REFERENCES

    Gerdes J, Li L, Schlueter C, et al: Immunobiochemical and molecular biologic characterization of the cell proliferation-associated nuclear antigen that is defined by monoclonal antibody Ki-67. Am J Pathol 138:867-873, 1991

    Gerdes J, Schwab U, Lemke H, et al: Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13-20, 1983

    van Dierendonck JH, Keijzer R, van de Velde CJ, et al: Nuclear distribution of the Ki-67 antigen during the cell cycle: Comparison with growth fraction in human breast cancer cells. Cancer Res 49:2999-3006, 1989

    Fonatsch C, Duchrow M, Rieder H, et al: Assignment of the human Ki-67 gene (MK167) to 10q25-qter. Genomics 11:476-477, 1991

    Schluter C, Duchrow M, Wohlenberg C, et al: The cell proliferation-associated antigen of antibody Ki-67: A very large, ubiquitous nuclear protein with numerous repeated elements, representing a new kind of cell cycle-maintaining proteins. J Cell Biol 123:513-522, 1993

    Scholzen T, Dimmler C, Wohlenberg C, et al: Novel splice forms of the Ki-67 mRNA. Eur J Cell Biol 72:40-44, 1997

    Lopez F, Belloc F, Lacombe F, et al: Modalities of synthesis of Ki67 antigen during the stimulation of lymphocytes. Cytometry 12:42-49, 1991

    Gerdes J, Lemke H, Baisch H, et al: Cell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67. J Immunol 133:1710-1715, 1984

    du Manoir S, Guillaud P, Camus E, et al: Ki-67 labeling in postmitotic cells defines different Ki-67 pathways within the 2c compartment. Cytometry 12:455-463, 1991

    Bruno S, Darzynkiewicz Z: Cell cycle dependent expression and stability of the nuclear protein detected by Ki-67 antibody in HL-60 cells. Cell Prolif 25:31-40, 1992

    Heidebrecht HJ, Buck F, Haas K, et al: Monoclonal antibodies Ki-S3 and Ki-S5 yield new data on the 'Ki-67' proteins. Cell Prolif 29:413-425, 1996

    Kill IR: Localisation of the Ki-67 antigen within the nucleolus: Evidence for a fibrillarin-deficient region of the dense fibrillar component. J Cell Sci 109:1253-1263, 1996 (part 6)

    Starborg M, Gell K, Brundell E, et al: The murine Ki-67 cell proliferation antigen accumulates in the nucleolar and heterochromatic regions of interphase cells and at the periphery of the mitotic chromosomes in a process essential for cell cycle progression. J Cell Sci 109:143-153, 1996 (part 1)

    Braun N, Papadopoulos T, Muller-Hermelink HK: Cell cycle dependent distribution of the proliferation-associated Ki-67 antigen in human embryonic lung cells. Virchows Arch B Cell Pathol Incl Mol Pathol 56:25-33, 1988

    Key G, Becker MH, Baron B, et al: New Ki-67-equivalent murine monoclonal antibodies (MIB 1-3) generated against bacterially expressed parts of the Ki-67 cDNA containing three 62 base pair repetitive elements encoding for the Ki-67 epitope. Lab Invest 68:629-636, 1993

    Shi SR, Key ME, Kalra KL: Antigen retrieval in formalin-fixed, paraffin-embedded tissues: An enhancement method for immunohistochemical staining based on microwave oven heating of tissue sections. J Histochem Cytochem 39:741-748, 1991

    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 2:585-592, 1996

    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 36:369-374, 1997

    Pietilainen T, Lipponen P, Aaltomaa S, et al: The important prognostic value of Ki-67 expression as determined by image analysis in breast cancer. J Cancer Res Clin Oncol 122:687-692, 1996

    Spyratos F, Ferrero-Pous M, Trassard M, et al: Correlation between MIB-1 and other proliferation markers: Clinical implications of the MIB-1 cutoff value. Cancer 94:2151-2159, 2002

    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 37:57-64, 1996

    Gasparini G, Pozza F, Meli S, et al: Breast cancer cell kinetics: Immunocytochemical determination of growth fractions by monoclonal antibody Ki-67 and correlation with flow cytometric S-phase and with some features of tumor aggressiveness. Anticancer Res 11:2015-2021, 1991

    Ellis PA, Makris A, Burton SA, et al: Comparison of MIB-1 proliferation index with S-phase fraction in human breast carcinomas. Br J Cancer 73:640-643, 1996

    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 17:3058-3063, 1999

    Makris A, Powles TJ, Dowsett M, et al: Prediction of response to neoadjuvant chemoendocrine therapy in primary breast carcinomas. Clin Cancer Res 3:593-600, 1997

    Thor AD, Liu S, Moore DH, et al: Comparison of mitotic index, in vitro bromodeoxyuridine labeling, and MIB-1 assays to quantitate proliferation in breast cancer. J Clin Oncol 17:470-477, 1999

    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 25:356-363, 1999

    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 97:1321-1331, 2003

    Bouzubar N, Walker KJ, Griffiths K, et al: Ki67 immunostaining in primary breast cancer: Pathological and clinical associations. Br J Cancer 59:943-947, 1989

    Sahin AA, Ro J, Ro JY, et al: Ki-67 immunostaining in node-negative stage I/II breast carcinoma: Significant correlation with prognosis. Cancer 68:549-557, 1991

    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 46:953-961, 1996

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

    Sasaki K, Matsumura K, Murakami T, et al: Measurement of bromodeoxyuridine labeling index, Ki-67 score and Ag-NOR count in breast carcinomas. Comparison with DNA ploidy. Oncology 49:147-153, 1992

    Haerslev T, Jacobsen GK, Zedeler K: Correlation of growth fraction by Ki-67 and proliferating cell nuclear antigen (PCNA) immunohistochemistry with histopathological parameters and prognosis in primary breast carcinomas. Breast Cancer Res Treat 37:101-113, 1996

    Prosperi E: Multiple roles of the proliferating cell nuclear antigen: DNA replication, repair and cell cycle control. Prog Cell Cycle Res 3:193-210, 1997

    Gonzalez MA, Pinder SE, Callagy G, et al: Minichromosome maintenance protein 2 is a strong independent prognostic marker in breast cancer. J Clin Oncol 21:4306-4313, 2003

    Harper-Wynne C, Ross G, Sacks N, et al: Effects of the aromatase inhibitor letrozole on normal breast epithelial cell proliferation and metabolic indices in postmenopausal women: A pilot study for breast cancer prevention. Cancer Epidemiol Biomarkers Prev 11:614-621, 2002

    Clarke RB, Howell A, Potten CS, et al: Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Res 57:4987-4991, 1997

    de Lima GR, Facina G, Shida JY, et al: Effects of low dose tamoxifen on normal breast tissue from premenopausal women. Eur J Cancer 39:891-898, 2003

    Shoker BS, Jarvis C, Clarke RB, et al: Estrogen receptor-positive proliferating cells in the normal and precancerous breast. Am J Pathol 155:1811-1815, 1999

    Lipponen P: Apoptosis in breast cancer: Relationship with other pathological parameters. Endocr Relat Cancer 6:13-16, 1999

    Bundred NJ, Anderson E, Nicholson RI, et al: Fulvestrant, an estrogen receptor downregulator, reduces cell turnover index more effectively than tamoxifen. Anticancer Res 22:2317-2319, 2002

    Cleator S, Parton M, Dowsett M: The biology of neoadjuvant chemotherapy for breast cancer. Endocr Relat Cancer 9:183-195, 2002

    Harper-Wynne CL, Sacks NP, Shenton K, et al: Comparison of the systemic and intratumoral effects of tamoxifen and the aromatase inhibitor vorozole in postmenopausal patients with primary breast cancer. J Clin Oncol 20:1026-1035, 2002

    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 85:1106-1112, 2001

    Liu S, Edgerton SM, Moore DH 2nd, et al: Measures of cell turnover (proliferation and apoptosis) and their association with survival in breast cancer. Clin Cancer Res 7:1716-1723, 2001

    Beck T, Weller EE, Weikel W, et al: Usefulness of immunohistochemical staining for p53 in the prognosis of breast carcinomas: Correlations with established prognosis parameters and with the proliferation marker, MIB-1. Gynecol Oncol 57:96-104, 1995

    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 71:146-149, 1995

    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 187:207-216, 1999

    Nicholson RI, McClelland RA, Finlay P, et al: Relationship between EGF-R, c-erbB-2 protein expression and Ki67 immunostaining in breast cancer and hormone sensitivity. Eur J Cancer 29A:1018-1023, 1993

    Gasparini G, Bevilacqua P, Pozza F, et al: Value of epidermal growth factor receptor status compared with growth fraction and other factors for prognosis in early breast cancer. Br J Cancer 66:970-976, 1992

    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 74:433-437, 1997

    Rudas M, Gnant MF, Mittlbock M, et al: Thymidine labeling index and Ki-67 growth fraction in breast cancer: Comparison and correlation with prognosis. Breast Cancer Res Treat 32:165-175, 1994

    Weikel W, Beck T, Mitze M, et al: Immunohistochemical evaluation of growth fractions in human breast cancers using monoclonal antibody Ki-67. Breast Cancer Res Treat 18:149-154, 1991

    Wintzer HO, Zipfel I, Schulte-Monting J, et al: Ki-67 immunostaining in human breast tumors and its relationship to prognosis. Cancer 67:421-428, 1991

    Bader AA, Tio J, Petru E, et al: T1 breast cancer: Identification of patients at low risk of axillary lymph node metastases. Breast Cancer Res Treat 76:11-17, 2002

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

    Billgren AM, Tani E, Liedberg A, et al: Prognostic significance of tumor cell proliferation analyzed in fine needle aspirates from primary breast cancer. Breast Cancer Res Treat 71:161-170, 2002

    Joensuu H, Isola J, Lundin M, et al: Amplification of erbB2 and erbB2 expression are superior to estrogen receptor status as risk factors for distant recurrence in pT1N0M0 breast cancer: A nationwide population-based study. Clin Cancer Res 9:923-930, 2003

    Iacopetta B, Grieu F, Powell B, et al: Analysis of p53 gene mutation by polymerase chain reaction-single strand conformation polymorphism provides independent prognostic information in node-negative breast cancer. Clin Cancer Res 4:1597-1602, 1998

    Rudolph P, Alm P, Heidebrecht HJ, et al: Immunologic proliferation marker Ki-S2 as prognostic indicator for lymph node-negative breast cancer. J Natl Cancer Inst 91:271-278, 1999

    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 105:674-680, 2003

    Rudolph P, Alm P, Olsson H, et al: Concurrent overexpression of p53 and c-erbB-2 correlates with accelerated cycling and concomitant poor prognosis in node-negative breast cancer. Hum Pathol 32:311-319, 2001

    Weikel W, Brumm C, Wilkens C, et al: Growth fractions (Ki-67) in primary breast cancers, with particular reference to node-negative tumors. Cancer Detect Prev 19:446-450, 1995

    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 29A:1509-1513, 1993

    Euler U, Tulusan AH: An attempt to identify biological markers predicting response to primary chemotherapy with epirubicin/cyclophosphamid in locally advanced breast cancer. Proc Am Soc Clin Oncol 21:888, 2003 (abstr 3569)

    Faneyte IF, Schrama JG, Peterse JL, et al: Breast cancer response to neoadjuvant chemotherapy: Predictive markers and relation with outcome. Br J Cancer 88:406-412, 2003

    Petit T, Wilt M, Velten M, et al: Comparative value of tumour grade, hormonal receptors, Ki-67, HER-2 and topoisomerase II alpha status as predictive markers in breast cancer patients treated with neoadjuvant anthracycline-based chemotherapy. Eur J Cancer 40:205-211, 2004

    Pohl G, Rudas M, Taucher S, et al: Expression of cell cycle regulatory proteins in breast carcinomas before and after preoperative chemotherapy. Breast Cancer Res Treat 78:97-103, 2003

    Chang J, Ormerod M, Powles TJ, et al: Apoptosis and proliferation as predictors of chemotherapy response in patients with breast carcinoma. Cancer 89:2145-2152, 2000

    Estevez LG, Cuevas JM, Anton A, et al: Weekly Docetaxel as neoadjuvant chemotherapy for stage II, and III breast cancer: Efficacy and correlation with biological markers in a phase II, multicenter study. Clin Cancer Res 9:686-692, 2003

    Chang J, Powles TJ, Allred DC, et al: Prediction of clinical outcome from primary tamoxifen by expression of biologic markers in breast cancer patients. Clin Cancer Res 6:616-621, 2000

    Makris A, Powles TJ, Allred DC, et al: Changes in hormone receptors and proliferation markers in tamoxifen treated breast cancer patients and the relationship with response. Breast Cancer Res Treat 48:11-20, 1998

    Parton M, Krajewski S, Smith I, et al: Coordinate expression of apoptosis-associated proteins in human breast cancer before and during chemotherapy. Clin Cancer Res 8:2100-2108, 2002

    Detre S, Salter J, Barnes DM, et al: Time-related effects of estrogen withdrawal on proliferation- and cell death-related events in MCF-7 xenografts. Int J Cancer 81:309-313, 1999

    Johnston SR, Boeddinghaus IM, Riddler S, et al: Idoxifene antagonizes estradiol-dependent MCF-7 breast cancer xenograft growth through sustained induction of apoptosis. Cancer Res 59:3646-3651, 1999

    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 82:113-123, 2003

    Ellis PA, Smith IE, Detre S, et al: Reduced apoptosis and proliferation and increased Bcl-2 in residual breast cancer following preoperative chemotherapy. Breast Cancer Res Treat 48:107-116, 1998

    Clarke RB, Laidlaw IJ, Jones LJ, et al: Effect of tamoxifen on Ki67 labelling index in human breast tumours and its relationship to oestrogen and progesterone receptor status. Br J Cancer 67:606-611, 1993

    Dowsett M, Dixon JM, Horgan K, et al: Antiproliferative effects of idoxifene in a placebo-controlled trial in primary human breast cancer. Clin Cancer Res 6:2260-2267, 2000

    Dowsett M, Bundred NJ, Decensi A, et al: Effect of raloxifene on breast cancer cell Ki67 and apoptosis: A double-blind, placebo-controlled, randomized clinical trial in postmenopausal patients. Cancer Epidemiol Biomarkers Prev 10:961-966, 2001

    DeFriend DJ, Howell A, Nicholson RI, et al: Investigation of a new pure antiestrogen (ICI 182780) in women with primary breast cancer. Cancer Res 54:408-414, 1994

    Robertson JF, Nicholson RI, Bundred NJ, et al: Comparison of the short-term biological effects of 7alpha-[9-(4,4,5,5,5-pentafluoropentylsulfinyl)-nonyl]estra-1,3,5, (10)-triene-3,17beta-diol (Faslodex) versus tamoxifen in postmenopausal women with primary breast cancer. Cancer Res 61:6739-6746, 2001

    Johnston SR, MacLennan KA, Sacks NP, et al: Modulation of Bcl-2 and Ki-67 expression in oestrogen receptor-positive human breast cancer by tamoxifen. Eur J Cancer 30A:1663-1669, 1994

    Bajetta E, Celio L, Di Leo A, et al: Effects of short-term pre-operative tamoxifen on steroid receptor and Ki-67 expression in primary breast cancer: An immunocytochemical study. Int J Oncol 12:853-858, 1998

    Dardes RD, Horiguchi J, Jordan VC: A pilot study of the effects of short-term tamoxifen therapy on Ki-67 labelling index in women with primary breast cancer. Int J Oncol 16:25-30, 2000

    Decensi A, Robertson C, Viale G, et al: A randomized trial of low-dose tamoxifen on breast cancer proliferation and blood estrogenic biomarkers. J Natl Cancer Inst 95:779-790, 2003

    Geisler J, Detre S, Berntsen H, et al: Influence of neoadjuvant anastrozole (Arimidex) on intratumoral estrogen levels and proliferation markers in patients with locally advanced breast cancer. Clin Cancer Res 7:1230-1236, 2001

    Ellis MJ, Coop A, Singh B, et al: Letrozole inhibits tumor proliferation more effectively than tamoxifen independent of HER1/2 expression status. Cancer Res 63:6523-6531, 2003

    Kenny FS, Willsher PC, Gee JM, et al: Change in expression of ER, bcl-2 and MIB1 on primary tamoxifen and relation to response in ER positive breast cancer. Breast Cancer Res Treat 65:135-144, 2001

    Prasad R, Boland GP, Cramer A, et al: Short-term biologic response to withdrawal of hormone replacement therapy in patients with invasive breast carcinoma. Cancer 98:2539-2546, 2003

    Singletary SE, Atkinson EN, Hoque A, et al: Phase II clinical trial of N-(4-Hydroxyphenyl)retinamide and tamoxifen administration before definitive surgery for breast neoplasia. Clin Cancer Res 8:2835-2842, 2002

    Dowsett M, Smith I, Ebbs SR, et al: Short-term changes in Ki67 during neoadjuvant treatment of primary breast cancer with anastrozole or tamoxifen alone or combined correlate with recurrence free survival. Clin Cancer Res 11:951S-958S, 2005

    Bardon S, Vignon F, Derocq D, et al: The antiproliferative effect of tamoxifen in breast cancer cells: Mediation by the estrogen receptor. Mol Cell Endocrinol 35:89-96, 1984

    Billgren AM, Rutqvist LE, Tani E, et al: Proliferating fraction during neoadjuvant chemotherapy of primary breast cancer in relation to objective local response and relapse-free survival. Acta Oncol 38:597-601, 1999

    Linn SC, Pinedo HM, van Ark-Otte J, et al: Expression of drug resistance proteins in breast cancer, in relation to chemotherapy. Int J Cancer 71:787-795, 1997

    Ellis PA, Smith IE, McCarthy K, et al: Preoperative chemotherapy induces apoptosis in early breast cancer. Lancet 349:849, 1997

    Daidone MG, Silvestrini R, Luisi A, et al: Changes in biological markers after primary chemotherapy for breast cancers. Int J Cancer 61:301-305, 1995

    Dowsett M, Smith I: Greater Ki67 response after 2 weeks neoadjuvant treatment with anastrozole than with tamoxifen or anastrozole plus tamoxifen in the IMPACT trial: A potential predictor of relapse-free survival. San Antonio Breast Cancer Conference, Breast Cancer Research and Treatment, San Antonio, TX, December 3-6, 2003

    Smith I, Dowsett M, Ebbs S, et al: Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen or both in combination: The IMPACT multicentre double-blind randomised trial. J Clin Oncol, 23:5108-5116, 2005

    Baum M, Buzdar A, Cuzick J, et al: Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: Results of the ATAC (Arimidex, Tamoxifen Alone or in Combination) trial efficacy and safety update analyses. Cancer 98:1802-1810, 2003

    Malik SN, Siu LL, Rowinsky EK, et al: Pharmacodynamic evaluation of the epidermal growth factor receptor inhibitor OSI-774 in human epidermis of cancer patients. Clin Cancer Res 9:2478-2486, 2003

    Baselga J, Rischin D, Ranson M, et al: Phase I safety, pharmacokinetic, and pharmacodynamic trial of ZD1839, a selective oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with five selected solid tumor types. J Clin Oncol 20:4292-4302, 2002

    Ellis MJ, Coop A, Singh B, et al: Letrozole is more effective neoadjuvant endocrine therapy than tamoxifen for ErbB-1- and/or ErbB-2-positive, estrogen receptor-positive primary breast cancer: Evidence from a phase III randomized trial. J Clin Oncol 19:3808-3816, 2001

    Dowsett M, Harper-Wynne C, Boeddinghaus I, et al: HER-2 amplification impedes the antiproliferative effects of hormone therapy in estrogen receptor-positive primary breast cancer. Cancer Res 61:8452-8458, 2001

    Duchrow M, Ziemann T, Windhovel U, et al: Colorectal carcinomas with high MIB-1 labelling indices but low pKi67 mRNA levels correlate with better prognostic outcome. Histopathology 42:566-574, 2003(Ander Urruticoechea, Ian )