Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

CD133 Is a Useful Surrogate Marker for Predicting Chemosensitivity to Neoadjuvant Chemotherapy in Breast Cancer

  • Naoki Aomatsu,

    Affiliation Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan

  • Masakazu Yashiro ,

    m9312510@med.osaka-cu.ac.jp

    Affiliations Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan, Oncology Institute of Geriatrics and Medical Science, Osaka City University Graduate School of Medicine, Osaka, Japan

  • Shinichiro Kashiwagi,

    Affiliation Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan

  • Tsutomu Takashima,

    Affiliation Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan

  • Tetsuro Ishikawa,

    Affiliation Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan

  • Masahiko Ohsawa,

    Affiliation Department of Diagnostic Pathology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan

  • Kenichi Wakasa,

    Affiliation Department of Diagnostic Pathology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan

  • Kosei Hirakawa

    Affiliation Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan

Abstract

Background

Neoadjuvant chemotherapy (NAC) is a standard care regimen for patients with breast cancer. However, the pathologic complete response (pCR) rate remains at 30%. We hypothesized that a cancer stem cell marker may identify NAC-resistant patients, and evaluated CD133 and ALDH1 as a potential surrogate marker for breast cancer. The aim of this study was to find a surrogate maker to predict chemosensitivity of NAC for breast cancer.

Methodology/Findings

A total of 102 patients with breast cancer were treated with NAC consisting of epirubicin followed by paclitaxel. Core needle biopsy (CNB) specimens and resected tumors were obtained from all patients before and after NAC, respectively. Chemosensitivity and prognostic potential of CD133 or ALDH1 expression was evaluated by immunohistochemistry. Clinical CR (cCR) and pCR rates were 18% (18/102) and 29% (30/102), respectively. Forty-seven (46%) patients had CD133-positive tumors before NAC, and CD133 expression was significantly associated with a low pCR rate (p = 0.035) and clinical non-responders. Multivariate analysis revealed that CD133 expression was significantly (p = 0.03) related to pCR. Recurrence was more frequent in patients with CD133-positive tumors (21/47, 45%) than that in patients with CD133-negative tumors (7/55, 13%). The number of patients with CD133-positive tumors (62%) after NAC was higher than that (46%) before NAC. Furthermore, most patients with CD133-positive tumors before NAC maintained the same status after NAC.

Conclusion/Significance

CD133 before NAC might be a useful marker for predicting the effectiveness of NAC and recurrence of breast cancer after NAC.

Introduction

Neoadjuvant chemotherapy (NAC) increases the resectability of tumors and decreases the risk of postoperative recurrence; thus resulting in superior long-term survival [1], [2]. For this reason, NAC is a standard care regimen for patients with various types of carcinomas, including breast cancer [3]. The optimal regimen for NAC in breast cancer involves a combination of 5-fluorouracil, epirubicin, and cyclophosphamide (FEC), followed by paclitaxel (PTX). [4], [5] The main aim of NAC is to reduce the size of the primary tumor, increase the likelihood of breast conservation [6], and allow evaluation of the therapeutic effects that facilitate establishment of therapeutic strategies based on the evaluation results [7]. Recent studies have demonstrated that pathologic complete response (pCR) in primary breast tumors after NAC correlates with improved disease-free survival (DFS) and overall survival (OS) [5], [8]. NAC for breast cancer has a pCR rate of approximately 30% [6], [9], [10] and a clinical CR (cCR) rate of approximately 60% [10]. In contrast, NAC is ineffective in approximately half of all patients, and many experience toxicity. Therefore, it would be advantageous to identify patients with chemosensitive tumors before initiating NAC, to avoid potential therapy-related complications and inappropriate delay of surgical treatment.

NAC has numerous advantages, including the use of pathological response data as a surrogate marker for long-term clinical outcome [11], [12] and assessment of responsiveness to NAC that allows the evaluation of potential predictive molecular markers for chemosensitivity. Several biological markers, including the estrogen receptor (ER), progesterone receptor (PgR), HER2, Ki-67, p21, p53, Bcl, multi-drug-resistant P-glycoprotein, and topoisomerase 2A, have recently been investigated; however, there is no clear correlation between marker expression and chemosensitivity after sequential taxane- and anthracycline-based chemotherapies [13][17], and more useful predictive markers for chemosensitivity need to be clinically identified.

thumbnail
Figure 1. Association between pCR and survival.

DFS in pathologic non-responders was significantly (p = 0.01) shorter than that for responders, while OS was not significantly different (p = 0.12).

https://doi.org/10.1371/journal.pone.0045865.g001

The recent discovery of the hierarchical organization among cancer stem cells (CSCs) and the finding that cancers emerge from their own progenitor stem cells has had important implications in cancer therapy [18]. In addition to being considered the source of tumor initiation and metastasis [19], [20], CSCs have been demonstrated to be resistant to chemotherapy, indicating that they are also responsible for tumor recurrence [21], [22]. In fact, several in vitro studies have shown that CSCs are resistant to PTX, doxorubicin, 5-fluorouracil, and platinum. Recently, Prominin-1 (CD133) has been considered to be a CSC marker in many types of cancers, such as breast [23][25], colorectal [19], [20], brain [26], [27], prostate [28], pancreatic [29], and gastric cancers [30]. In addition, recently, aldehyde dehydrogenase (ALDH) 1 has been identified as a reliable marker for breast CSC marker [31][33].

thumbnail
Table 1. Clinical and pathological response in 102 primary breast cancers.

https://doi.org/10.1371/journal.pone.0045865.t001

In this retrospective study, to evaluate the potential of CD133 or ALDH1 as a surrogate marker for NAC resistance, we examined the correlation between chemosensitivity to NAC and CD133 or ALDH1 as well as prognosis of patients with breast cancer after NAC treatment.

Patients and Methods

Patients

A total of 102 patients with breast cancer that was considered to be stage IIA, IIB, and IIIA, was treated with NAC from 2004 to 2009. Tumors were confirmed histopathologically by core needle biopsy (CNB) and were staged by ultrasonography or computed tomography. The clinical stage was based on the TNM Classification of Malignant Tumors, 6th Edition [34]. The tumor size and axillary lymph node metastasis were examined by ultrasonography. No patients had evidence of distant metastasis at the time of surgery. The median age of the patients was 55.0 years old (range 26–78 years old). All of the cases were received neoadjuvant chemotherapy with 4 cycle of 5FU 500 mg/m2, epirubicin 75 or 100 mg/m2, and cyclophosphamide 500 mg/m2 (FEC) followed by 12 cycles of weekly paclitaxel 80 mg/m2 (wPTX). Sixteen of 102 patients showed HER2-positive breast cancer, and were administered weekly trastuzumab with wPTX. Patients were underwent mastectomy or breast-conserving surgery after NAC. Patients who underwent breast-conserving surgery were administered postoperative radiotherapy. Overall survival time was set in days as the period from the NAC starting day. This study was conducted with the consent of the ethical committee of Osaka City University, and informed consent was obtained from all subjects.

Assesment of Clinical and Pathological Response to NAC

Clinical response of primary tumor was assessed by ultrasonography and physical examination after NAC. Clinical responses were classified according to WHO criteria [35]. After NAC, patients underwent appropriate surgery. The clinical response to preoperative chemotherapy was decided from the two diameters measurable in two dimensions determined by multiplying the longest diameter by the greatest perpendicular diameter: clinical complete response (cCR); a disappearance of all known disease determined by two observations not less than four weeks apart, clinical partial response (cPR); a 50% or more decrease in total tumor lesions, clinical no change (cNC); a 50% decrease in total tumor size, nor a 25% increase in the tumor size, clinical progressive disease (cPD); a 25% or more increase in the tumor size, or the appearance of new lesions. cCR and cPR were judged as effective. Pathological response of tumor and dissected lymph nodes were classified according to the evaluation criteria of the Japanese Breast Cancer Society (JBCS) [36], using a 5-grade scale (Grade 0, 1a, 1b, 2, and 3) as follows: Grade 0, no response or almost no change in cancer cells after treatment; Grade 1, slight response; Grade 1a, mild response, mild change in cancer cells regardless of the area, or marked changes in cancer cells in less than one-third of total cancer cells; Grade 1b, moderate response, marked changes in one-third or more but less than two-thirds of tumor cells; Grade 2,marked response or marked changes in two-third or more of tumor cells; and Grade 3,no residual tumor cells, necrosis or disappearance of all tumor cells, or replacement of all cancer cells by granuloma-like and/or fibrous tissue. pCR (pathological complete response) were defined as the complete disappearance of infiltrates, including lymph node infiltrates, with or without intraductal components. Tumors with residual ductal carcinoma in situ were included in the pCR group. Marked changes approaching a complete response with only a few remaining cancer cells were classified as near pCR [37], [38]. The others were grouped in the non-pCR.

thumbnail
Figure 2. Immunohistochemical staining of CD133 or ALDH1.

(A), Immunohistochemical determination of CD133 expression. The CD133 antibody stained intensely at the membrane and in the cytoplasm of cancer cells. Scores were applied as follows: 0, negative staining in all cells; 1+, weakly positive or focally positive staining in <10% of cells; 2+, moderately positive-staining in 10%-50% of cells; and score 3+, strongly positive-staining, involving 50% or more of the cells. (B), Immunohistochemical determination of ALDH1 expression. The ALDH1 antibody stained intensely at the membrane and in the cytoplasm of cancer cells. Scores were applied as follows: 0, negative staining in all cells; 1+, weakly positive or focally positive staining in <10% of cells; 2+, moderately positive-staining in 10%-50% of cells; and score 3+, strongly positive-staining, involving 50% or more of the cells.

https://doi.org/10.1371/journal.pone.0045865.g002

Immunohistochemical Examinations

All patients were underwent a core needle biopsy before NAC, and had received a curative operation of a mastectomy or a conservative surgery with axillary lymph node dissection after NAC in Osaka City University. Tissues from each patient were fixed in buffered formalin and embedded in paraffin. Serial tissue sections with 4 µm were stained with hematoxylin-eosin and used for immunohistochemical staning. Expressions of CD133, estrogen receptor (ER), progesterone receptor (PgR), and HER2 were assessed by immunohistochemistry. After the paraffin sections were deparaffinized, they were heated for 20 min at 105°C by autoclave in Target Retrieval Solution (Dako, Carpinteria, CA). After blocking with 10% goat serum, the slides were incubated with each primary monoclonal antibody against, ER (clone 1D5, dilution 1∶80; Dako, Cambridge, UK), PgR (clone PgR636, dilution 1∶100; Dako), HER2 (Hercep Test, Dako), CD133 (clone NCH-38, dilution 1∶200; Dako), and ALDH1 (dilution 1∶100; BD Bioscience, San Jose, CA) overnight at 4°C. Peroxidase was introduced using a streptavidin conjugate and then peroxidase reactivity was visualized using a DAB solution, followed by counterstaining with haematoxylin.

thumbnail
Table 2. Correlations between CD133 or ALDH1 expression and clinicopathological parameters in CNB of 102 primary breast cancers.

https://doi.org/10.1371/journal.pone.0045865.t002

Immunohistochemical Assessment

Immunohistochemical scoring was performed in a blind manner. The cut-off for ER positivity and PgR positivity was ≥1% positive tumor cells with nuclear staining. HER2 was graded according to the accepted grading scheme as 0, 1+, 2+, 3+. the following criteria were used for scoring: 0, no reactivity or membranous reactivity in less than 10% of cells; 1+, faint/barely perceptible membranous reactivity in 10% of cells or higher or reactivity in only part of the cell membrane; 2+, weak to moderate complete or basolateral membranous reactivity in 10% of tumor cells or higher; 3+, strong complete or basolateral membranous reactivity in 10% of tumor cells or higher. HER-2 was considered to be positive if immunostaining was 3+ or if a 2+ result showed gene amplication by fluorescent in situ hybridization (FISH). In FISH analyses, each copy of the HER2 gene and its centromere 17 (CEP17) reference were counted. The interpretation followed the criteria of the ASCO/CAP guidelines for HER2 IHC interpretation for breast cancer [39]: positive if the HER2/CEP17 ratio was higher than 2.2. CD133 and ALDH1 antibody stained intensely the membrane and cytoplasm of cancer cells. Scores were applied as follows: score 0, negative staining in all cells; score 1+, weekly positive or focally positive staining in <10% of the cells; score 2+, moderately positive staining covering 10% to 50% of the cells; and score 3+, stringly positive staining, including >50% of the cells. CD133 and ALDH1 expression was considered positive when scores were ≥2.

Statistical Analysis

Statistical analysis was performed using SPSS 13.0 statistical software (SPSS Inc, Chicago, USA). The association between the expression of CD133 or ALDH1 and clinicopathological parameters was analyzed with the chi-square test. The Kaplan-Meier method was used to estimate the values of DFS. DFS was compared using a Log-rank test. Events for the calculation of DFS included all local, regional, or distant recurrence. The Cox regression model was used for multivariate analysis of prognostic factors. In all of the tests, a p value less than 0.05 was considered to be statistically significant.

Results

Clinical and Pathological Response of Primary Breast Cancers to NAC

The cCR rate was 18% (18/102), cPR was 61% (62/102), cNC was 20% (20/102), and cPD was 2% (2/102). Clinical responders (cCR + cPR) included 78% (80/102) of the patients. Of the tumors investigated, 12% (12/102) were grade 1a, 33% (34/102) were grade 1b, 20% (20/102) were grade 2a, 16% (16/102) were grade 2b, and 20% (20/102) were grade 3. Patients were was classified according to the grade of tumor into pathologic responders, who had grade 2 and 3 tumors and equaled 55% of all patients, and non-responders, who had grade 1 tumors and equaled 45%. Overall, the pCR rate was 29% (30/102) (Table 1). DFS in pathologic non-responders was significantly poorer (p = 0.01) than that in pathologic responders (Fig. 1).

Association between Clinicopathological Parameters and CD133 or ALDH1 Expression in Before NAC

Immunohistochemical patterns of CD133 or ALDH1 expression were analyzed in core needle biopsy (CNB) specimens from 102 patients. Forty-seven patients had CD133-positive primary breast tumors (46%) (Fig. 2A), while 55 (54%) had CD133-negative tumors before NAC. Sixteen patients (16%) had ALDH1-positive primary breast tumors, while 86 patients (84%) had ALDH1-negative tumors before NAC (Fig. 2B). Table 2 shows the characteristics of patients included in the study. CD133 expression in CNB specimens significantly correlated with lymph node metastasis (43%, p = 0.042) and lymphatic invasion (45%, p<0.001). In contrast, no significant association was found between ALDH1 expression and clinicopathological factors. The pCR rate of CD133-positive tumors (19%, 9/47) was significantly lower (p = 0.035) than that of CD133-negative tumors (38%, 21/55). The pathological non-responder (grade 1) tumors were more frequently (p = 0.002) CD133 positive (69%, 38/55) than the pathological responder (grade 2 and 3) tumors (38%, 18/47). CD133 positivity was associated with the clinical response (cCR + cPR) (p = 0.062). Recurrence was observed in 28 patients and was more frequently found in patients with CD133-positive tumors (21/47, 45%) than those with CD133-negative tumors (7/55, 13%). There was no significant association between CD133 expression and other clinicopathological factors.

Association between pCR and CD133 or ALDH1 Expression in CNB Specimens

Univariate and multivariate analyses of pCR of tumors before NAC are shown in Table 3. Univariate analysis revealed that CD133, ER, and PgR expression was significantly associated with pCR. However, multivariate analysis revealed that only CD133 expression was significantly associated with pCR.

thumbnail
Table 3. Univariate and multivariate analysis with pathological complete response in 102 breast cancers.

https://doi.org/10.1371/journal.pone.0045865.t003

Association between Survival and CD133 or ALDH1 Expression

DFS and OS in patients with CD133-positive tumors were significantly shorter (p = 0.002 and p = 0.030, respectively) than those in patients with CD133-negative tumors before NAC. In contrast, ALDH1 expression did not correlate with response to DFS and OS. Since 20 of the 102 breast tumors were of pathological grade 3 before NAC, CD133 expression was examined in only 82 tumors after NAC. DFS in patients with CD133-positive tumors was significantly shorter (p = 0.028) than that for patients with CD133-negative tumors after NAC. Univariate and multivariate regression analyses indicated that CD133 expression before NAC was an independent prognostic factor for DFS (Table 4). Although OS in patients with CD133-positive tumors was shorter than that in patients with CD133-negative tumors, this difference was not significantly different (p = 0.109, Figure 3).

thumbnail
Table 4. Univariate and multivariate analysis with respect to disease free survival in primary breast cancers.

https://doi.org/10.1371/journal.pone.0045865.t004

thumbnail
Figure 3. Association between CD133 expression and survival.

In CNB specimens before NAC, DFS and OS in patients with CD133-positive tumors were significantly shorter than those in patients with CD133-negative tumors. In resected tumors after NAC, DFS for patients with CD133-positive tumors were significantly shorter than for patients with CD133-negative tumors, while OS was not significantly different. Since 20 of the 102 breast tumors were of pathological grade 3 before NAC, CD133 expression was examined in only 82 tumors after NAC.

https://doi.org/10.1371/journal.pone.0045865.g003

Association between CD133 Expression Before and After NAC

The association of CD133 expression between CD133 expression before and after NAC is shown in Table 5. The number of patients with CD133-positive tumors was higher after NAC (51/82; 62%) than that before NAC (47/102; 46%). Changes in CD133 expression were compared before and after NAC in the 82 patients who did not achieve pCR. Of these 82 patients, 42 patients had CD133-positive tumors and 40 had CD133-negative tumors before NAC. Of the 40 patients with CD133-negative tumors before NAC, 20 (50%) remained CD133 negative, whereas 20 (50%) changed to CD133 positive after NAC. Significantly more patients who were CD133-negative before and after NAC (18/20) were pathological responders of grade 2 (p<0.001). On the other hand, most patients who were CD133-positive before NAC showed the same status after NAC (31/42; 74%). Patients with CD133-negative tumors (7/11) were significantly more frequently pathological responders (p = 0.019) than patients with CD133-positive tumors. In contrast, tumor recurrence was more frequent in patients with CD133-positive (19/31) tumors before and after NAC (Figure 4).

thumbnail
Table 5. CD133 Status between pre-NAC CNB specimens and post-NAC resected tumors.

https://doi.org/10.1371/journal.pone.0045865.t005

thumbnail
Figure 4. Association between CD133 expression and survival before and after NAC.

DFS in patients CD133-positive tumors after NAC was significantly shorter than that for patients with CD133-neagtive tumors, while OS was not significant.

https://doi.org/10.1371/journal.pone.0045865.g004

Discussion

In this study, NAC included FEC followed by PTX, which is a standard treatment regimen for patients with breast cancer [4], [5]. Despite the various definitions of pathological response in neoadjuvant trials, no significant differences in survival among the various classification systems, including cTMN, Fisher’s, Chevailler’s, and JBCS, has been reported. This study used the histological response criteria of WHO and JBCS. In this study, the pCR rate was 29% (30/102) and the cCR rate was 78% (80/102). These response rates were very similar to those previously reported [6], [9], [10]. NAC has numerous advantages, including the use of pathological response data as a surrogate marker for long-term clinical outcome [11], [12]. Forty-seven (46%) of patients had CD133-positive primary breast tumors before NAC, which was similar to findings from a previous study [40]. Before NAC, CD133 expression significantly associated with lymph node status and lymph-vascular invasion, as reported previously [23], [40], [41]. These findings may suggest that the function of CD133 is associated with lymph node metastasis.

The cCR rate of CD133-positive tumors was significantly lower than that of CD133-negative tumors. Multivariate analysis revealed that CD133 expression before NAC was an independent predictive factor for pCR. These results may indicate that CD133-positive tumors play a significant role in resistance to chemotherapy.

Recent progress in CSC research has led to a better understanding of the mechanism of resistance to chemotherapy and development of more effective chemotherapeutic regimens and new antitumor agents [42]. An association between CSCs and drug resistance in breast cancer cell lines has been shown in vitro [43][45]. In breast cancer, CD44+CD24−/low [46], [47], aldehyde dehydrogenase (ALDH1) [48], [49], and CD133 [23][25] have been considered as markers of CSCs. However, recent studies have shown that CD44+CD24−/low tumor cell numbers are not associated with pCR rates after NAC [50], [51]. In our study, ALDH1 expression did not correlate with pCR rates and response to DFS and OS. As previous report, ALDH1 expression in tumor cells did not correlate with response to neoadjuvant therapy, DFS, or OS after NAC [18]. CD133 may indicate CSC properties in a more restricted manner than other CSC markers such as CD44/CD24 or ALDH1 [52]. Although CD133 has been considered as a CSC marker in breast cancer [23][25], there has been no report regarding CD133 expression in breast cancer treated with NAC until date. In the present study, we found that CD133 expression before NAC may be a useful marker for predicting the effectiveness of NAC in breast cancer. To the best of our knowledge, this is the first study to report the potential clinical benefits of evaluating CD133 expression.

Correlative studies of tumor samples before and after treatment may provide further information on markers that could predict response or resistance to NAC; however, until date, few reports have examined pathological responses in tumors before and after chemotherapy [50]. Changes in CD133 expression before and after NAC were studied in 82 patients who did not achieve pathological grade 3. CD133 expression after NAC (62%) was higher than that before NAC (46%); thus, enrichment for CD133-positive cells was observed in post-NAC tumor specimens than in pre-NAC specimens. Most patients with CD133-positive tumors before NAC remained positive after NAC. These findings suggest that NAC was effective in reducing CD133-negative cells, and resulted in an increase in CD133-positive CSCs.

Distant recurrence was observed in 28 of 102 patients, and was significantly (p<0.001) frequent in patients with CD133-positive tumors (21/47, 45%). However, 7 of 28 patients who developed distant recurrence were CD133-negative. CD133 might not detect all of CSCs. Another CSC marker besides CD133 might be necessary to predict a distant recurrence of breast cancer patients after curative operation. In contrast, 26 of 47 patients with CD133-positive tumors did not develop distant recurrence within 7 years after operation. However, the 26 patients might have a possibility of recurrence in the future, because some breast cancer recurrent after the long disease-free interval. It might be necessary to continue the follow-up of distant recurrence for the 26 patients with CD133-positive tumors.

CD133 expression in after NAC was not correlated with prognosis, while before NAC it appeared to be associated with poor prognosis. Thus, CD133 expression after NAC may not be clinically informative for patients treated with NAC.

ER- or PR-negative patients were associated with higher pCR rates after NAC than in ER- or PR-positive patients, as previously reported [5], [17], while multivariate analysis showed that neither of these factors were independent surrogate markers for pCR. The response to NAC could be predicted more accurately by adding CD133 expression to that of ER and PR.

In conclusion, CD133 is a useful surrogate maker for predicting chemosensitivity and recurrence to FEC followed by PTX in breast cancer.

Author Contributions

Conceived and designed the experiments:. Performed the experiments: NA. Analyzed the data: NA SK. Contributed reagents/materials/analysis tools: TT MO TI KH. Wrote the paper: NA. Took part in the pathological diagnosis: MO KW. Participated in study concepts, study design: NA MY. Reviewed the manuscript: TI KH.

References

  1. 1. Mayer EL, Carey LA, Burstein HJ (2007) Clinical trial update: implications and management of residual disease after neoadjuvant therapy for breast cancer. Breast Cancer Res 9: 110.
  2. 2. Sachelarie I, Grossbard ML, Chadha M, Feldman S, Ghesani M, et al. (2006) Primary systemic therapy of breast cancer. Oncologist 11: 574–589.
  3. 3. Fisher B, Brown A, Mamounas E, Wieand S, Robidoux A, et al. (1997) Effect of preoperative chemotherapy on local-regional disease in women with operable breast cancer: findings from National Surgical Adjuvant Breast and Bowel Project B-18. J Clin Oncol 15: 2483–2493.
  4. 4. Wolmark N, Wang J, Mamounas E, Bryant J, Fisher B (2001) Preoperative chemotherapy in patients with operable breast cancer: nine-year results from National Surgical Adjuvant Breast and Bowel Project B-18. J Natl Cancer Inst Monogr: 96–102.
  5. 5. Bear HD, Anderson S, Brown A, Smith R, Mamounas EP, et al. (2003) The effect on tumor response of adding sequential preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: preliminary results from National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol 21: 4165–4174.
  6. 6. Smith IC, Heys SD, Hutcheon AW, Miller ID, Payne S, et al. (2002) Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel. J Clin Oncol 20: 1456–1466.
  7. 7. Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, et al. (2007) Progress and promise: highlights of the international expert consensus on the primary therapy of early breast cancer 2007. Ann Oncol 18: 1133–1144.
  8. 8. Fisher CS, Ma CX, Gillanders WE, Aft RL, Eberlein TJ, et al. (2012) Neoadjuvant chemotherapy is associated with improved survival compared with adjuvant chemotherapy in patients with triple-negative breast cancer only after complete pathologic response. Ann Surg Oncol 19: 253–258.
  9. 9. Chollet P, Amat S, Cure H, de Latour M, Le Bouedec G, et al. (2002) Prognostic significance of a complete pathological response after induction chemotherapy in operable breast cancer. Br J Cancer 86: 1041–1046.
  10. 10. Jones RL, Smith IE (2006) Neoadjuvant treatment for early-stage breast cancer: opportunities to assess tumour response. Lancet Oncol 7: 869–874.
  11. 11. Evans TR, Yellowlees A, Foster E, Earl H, Cameron DA, et al. (2005) Phase III randomized trial of doxorubicin and docetaxel versus doxorubicin and cyclophosphamide as primary medical therapy in women with breast cancer: an anglo-celtic cooperative oncology group study. J Clin Oncol 23: 2988–2995.
  12. 12. Bear HD, Anderson S, Smith RE, Geyer CE Jr, Mamounas EP, et al. (2006) Sequential preoperative or postoperative docetaxel added to preoperative doxorubicin plus cyclophosphamide for operable breast cancer:National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol 24: 2019–2027.
  13. 13. Estevez LG, Cuevas JM, Anton A, Florian J, Lopez-Vega JM, et al. (2003) 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.
  14. 14. Chuthapisith S, Eremin JM, El-Sheemy M, Eremin O (2006) Neoadjuvant chemotherapy in women with large and locally advanced breast cancer: chemoresistance and prediction of response to drug therapy. Surgeon 4: 211–219.
  15. 15. Ross JS, Symmans WF, Pusztai L, Hortobagyi GN (2005) Breast cancer biomarkers. Adv Clin Chem 40: 99–125.
  16. 16. Colleoni M, Viale G, Zahrieh D, Pruneri G, Gentilini O, et al. (2004) Chemotherapy is more effective in patients with breast cancer not expressing steroid hormone receptors: a study of preoperative treatment. Clin Cancer Res 10: 6622–6628.
  17. 17. Rody A, Karn T, Gatje R, Ahr A, Solbach C, et al. (2007) Gene expression profiling of breast cancer patients treated with docetaxel, doxorubicin, and cyclophosphamide within the GEPARTRIO trial: HER-2, but not topoisomerase II alpha and microtubule-associated protein tau, is highly predictive of tumor response. Breast 16: 86–93.
  18. 18. Resetkova E, Reis-Filho JS, Jain RK, Mehta R, Thorat MA, et al. (2010) Prognostic impact of ALDH1 in breast cancer: a story of stem cells and tumor microenvironment. Breast Cancer Res Treat 123: 97–108.
  19. 19. O’Brien CA, Pollett A, Gallinger S, Dick JE (2007) A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445: 106–110.
  20. 20. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, et al. (2007) Identification and expansion of human colon-cancer-initiating cells. Nature 445: 111–115.
  21. 21. Neuzil J, Stantic M, Zobalova R, Chladova J, Wang X, et al. (2007) Tumour-initiating cells vs. cancer ‘stem’ cells and CD133: what’s in the name? Biochem Biophys Res Commun 355: 855–859.
  22. 22. Tang C, Ang BT, Pervaiz S (2007) Cancer stem cell: target for anti-cancer therapy. Faseb J 21: 3777–3785.
  23. 23. Xiao Y, Ye Y, Yearsley K, Jones S, Barsky SH (2008) The lymphovascular embolus of inflammatory breast cancer expresses a stem cell-like phenotype. Am J Pathol 173: 561–574.
  24. 24. Wright MH, Calcagno AM, Salcido CD, Carlson MD, Ambudkar SV, et al. (2008) Brca1 breast tumors contain distinct CD44+/CD24− and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res 10: R10.
  25. 25. Storci G, Sansone P, Trere D, Tavolari S, Taffurelli M, et al. (2008) The basal-like breast carcinoma phenotype is regulated by SLUG gene expression. J Pathol 214: 25–37.
  26. 26. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, et al. (2004) Identification of human brain tumour initiating cells. Nature 432: 396–401.
  27. 27. Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, et al. (2003) Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A 100: 15178–15183.
  28. 28. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ (2005) Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65: 10946–10951.
  29. 29. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, et al. (2007) Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1: 313–323.
  30. 30. Smith LM, Nesterova A, Ryan MC, Duniho S, Jonas M, et al. (2008) CD133/prominin-1 is a potential therapeutic target for antibody-drug conjugates in hepatocellular and gastric cancers. Br J Cancer 99: 100–109.
  31. 31. Deng S, Yang X, Lassus H, Liang S, Kaur S, et al. (2010) Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS One 5: e10277.
  32. 32. Charafe-Jauffret E, Ginestier C, Iovino F, Tarpin C, Diebel M, et al. (2010) Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res 16: 45–55.
  33. 33. Kai K, Arima Y, Kamiya T, Saya H (2010) Breast cancer stem cells. Breast Cancer 17: 80–85.
  34. 34. Singletary SE, Greene FL (2003) Revision of breast cancer staging: the 6th edition of the TNM Classification. Semin Surg Oncol 21: 53–59.
  35. 35. Miller AB, Hoogstraten B, Staquet M, Winkler A (1981) Reporting results of cancer treatment. Cancer 47: 207–214.
  36. 36. Kurosumi M, Akashi-Tanaka S, Akiyama F, Komoike Y, Mukai H, et al. (2008) Histopathological criteria for assessment of therapeutic response in breast cancer (2007 version). Breast Cancer 15: 5–7.
  37. 37. Toi M, Nakamura S, Kuroi K, Iwata H, Ohno S, et al. (2008) Phase II study of preoperative sequential FEC and docetaxel predicts of pathological response and disease free survival. Breast Cancer Res Treat 110: 531–539.
  38. 38. Nishimura R, Osako T, Okumura Y, Hayashi M, Arima N (2010) Clinical significance of Ki-67 in neoadjuvant chemotherapy for primary breast cancer as a predictor for chemosensitivity and for prognosis. Breast Cancer 17: 269–275.
  39. 39. Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, et al. (2007) American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25: 118–145.
  40. 40. Zhao P, Lu Y, Jiang X, Li X (2011) Clinicopathological significance and prognostic value of CD133 expression in triple-negative breast carcinoma. Cancer Sci 102: 1107–1111.
  41. 41. Liu Q, Li JG, Zheng XY, Jin F, Dong HT (2009) Expression of CD133, PAX2, ESA, and GPR30 in invasive ductal breast carcinomas. Chin Med J (Engl) 122: 2763–2769.
  42. 42. Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea–a paradigm shift. Cancer Res 66: 1883–1890; discussion 1895–1886.
  43. 43. Liu G, Yuan X, Zeng Z, Tunici P, Ng H, et al. (2006) Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer 5: 67.
  44. 44. Ghods AJ, Irvin D, Liu G, Yuan X, Abdulkadir IR, et al. (2007) Spheres isolated from 9L gliosarcoma rat cell line possess chemoresistant and aggressive cancer stem-like cells. Stem Cells 25: 1645–1653.
  45. 45. Fillmore CM, Kuperwasser C (2008) Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res 10: R25.
  46. 46. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100: 3983–3988.
  47. 47. Honeth G, Bendahl PO, Ringner M, Saal LH, Gruvberger-Saal SK, et al. (2008) The CD44+/CD24- phenotype is enriched in basal-like breast tumors. Breast Cancer Res 10: R53.
  48. 48. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, et al. (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1: 555–567.
  49. 49. Charafe-Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, et al. (2009) Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res 69: 1302–1313.
  50. 50. Tanei T, Morimoto K, Shimazu K, Kim SJ, Tanji Y, et al. (2009) Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin Cancer Res 15: 4234–4241.
  51. 51. Horiguchi K, Toi M, Horiguchi S, Sugimoto M, Naito Y, et al. (2012) Predictive value of CD24 and CD44 for neoadjuvant chemotherapy response and prognosis in primary breast cancer patients. J Med Dent Sci 57: 165–175.
  52. 52. Lorico A, Rappa G (2011) Phenotypic heterogeneity of breast cancer stem cells. J Oncol 2011: 135039.