FDG-PET/CT在评估可手术乳腺癌手术后转归中的价值——可用于三阴性乳腺癌恶性度分级

Role of FDG-PET/CT in evaluating surgical outcomes of operable breast cancer – Usefulness for malignant grade of triple-negative breast cancer
作者:Masahiro Ohara,  Hideo Shigematsu,  Yasuhiro Tsu
期刊: The Breast2013年6月期卷

 

 

Role of FDG-PET/CT in evaluating surgical outcomes of operable breast cancer – Usefulness for malignant grade of triple-negative breast cancer

  • Masahiro Ohara, 
  • Hideo Shigematsu, 
  • Yasuhiro Tsutani, 
  • Akiko Emi, 
  • Norio Masumoto, 
  • Shinji Ozaki,
  • Takayuki Kadoya, 
  • Morihito OkadaCorresponding author contact information
  • Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3-Kasumi, Minami-Ku, Hiroshima City, Hiroshima 734-0037, Japan
  • http://dx.doi.org/10.1016/j.breast.2013.05.003, How to Cite or Link Using DOI

Abstract

Background

The aim of this study was to evaluate the significance of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) for speculating the malignant level and prognostic value of operable breast cancers.

Methods

Of 578 consecutive patients with primary invasive breast cancer who underwent curative surgery between 2005 and 2010, 311 patients (53.8%) who received FDG-PET/CT before initial therapy were examined.

Results

Receiver operating characteristics (ROC) curve analysis showed the cutoff value of the maximum standardized uptake value (SUVmax) to predict cancer recurrence was 3.8 in all patients and 8.6 in patients with the triple-negative subtype, respectively. In all patients, 3-year DFS rates were 98.8% for patients with a tumor of SUVmax ≤ 3.8 and 91.6% for patients with a tumor of SUVmax > 3.8 (p < 0.001). High value of SUVmax was significantly associated with large tumor size (p < 0.001), lymph node metastasis (p = 0.040), high nuclear grade (p < 0.001), lymphovascular invasion (p = 0.032), negative hormone receptor status (p < 0.001), and positive HER2 status (p = 0.014). Based on the results of multivariate Cox analysis in all patients, high SUVmax (p = 0.001) and negative hormone receptor status (p = 0.005) were significantly associated with poor prognosis. In patients with triple-negative subtype, 3-year DFS rates were 90.9% for patients with a tumor of SUVmax ≤ 8.6 and 42.9% for patients with a tumor of SUVmax > 8.6 (p = 0.002), and high SUVmax was the only significant independent prognostic factor (p = 0.047).

Conclusion

FDG-PET/CT is useful for predicting malignant behavior and prognosis in patients with operable breast cancer, especially the triple-negative subtype.

Keywords

  • Breast cancer
  • PET/CT
  • SUVmax
  • Prognosis
  • Triple-negative breast cancer

Abbreviations

  • AUCarea under the curve
  • CTcomputed tomography
  • DFSdisease-free survival
  • ERestrogen receptor
  • FDG-PET[18F]-fluoro-2-deoxyglucose-positron emission tomography
  • HER2human epidermal growth factor receptor type 2
  • IDCinvasive ductal carcinoma
  • PgRprogesterone receptor
  • ROCreceiver operating characteristic
  • SUVmaxmaximum standardized uptake value

Introduction

Glucose metabolism in cancer cells can be visualized by FDG-PET and thus primary cancer can be staged and distant metastasis can be detected.1, 2 and 3 The glycolysis level in cancer cells is higher than that in normal tissue and various types of malignant tumors, such as glioma, malignant lymphoma,4 lung cancer,5liver tumors6 and breast cancer7 and 8 can be visualized by PET using the tracer FDG.

Accurate initial staging of patients with breast cancer is essential for precise prognostication and optimal choice of therapy. For initial and recurrent breast cancer staging, PET/CT has been demonstrated to be more accurate than conventional imaging methods.9, 10, 11, 12, 13 and 14

This technique has also been used not only to detect cancer but also to evaluate the proliferative activity and/or malignancy grades of specific types of tumors.15 and 16 High levels are considered to indicate more aggressive potential than low levels of 18F-FDG uptake by primary breast cancers.17 and 18 Several pioneering studies have identified associations between the intensity of 18F-FDG uptake and histological and biological characteristics such as tumor type, grade, hormonal receptor status and HER2 status.19, 20,21, 22, 23 and 24 However, the prognostic value of SUVmax has not been fully assessed.

We therefore investigated the prognostic value of primary site SUVmax for disease-free survival (DFS) in patients with operable primary breast cancer.

Patients and methods

Patients

Between September 2005 and October 2010, 578 consecutive patients with primary breast cancer underwent curative surgery at our institution. Of these, 311 (53.8%) had been evaluated by FDG-PET/CT before initial therapy and a SUVmax had been established. Patients who had noninvasive primary breast cancer and refused to undergo PET/CT were excluded. The data of all patients were retrospectively analyzed in the present study. The mean age of the patients was 58.4 (range, 28–91) years and those who had received preoperative chemotherapy were included. A total of 104 patients underwent mastectomy, 207 underwent breast-conserving therapy, and clinicopathological information was complete for all of them. The follow-up period ended on 30 June 2011. The median follow-up period was 29.8 months. This study was approved by our institutional review board (No. 337), which waived the requirement for informed consent from individual patients.

FDG-PET/CT

The patients fasted for at least four hours before being intravenously injected with 3.7 MBq/kg of FDG, and then they rested for about one hour before being scanned. Blood glucose was measured to ensure a level of <150 mg/dL before tracer injection. Patients with blood glucose values of ≥150 mg/dL during PET/CT image acquisition were excluded. All patients were assessed using a Discovery ST16 integrated PET/CT scanner (GE Healthcare). An unenhanced CT image of a 2–4-mm-thick section that matched the PET images was obtained from the head to the pelvic floor of each patient using a standard protocol. Immediately thereafter, PET covered the identical axial field of view for 2–4 min per table position depending on the condition of the patient and scanner performance. Both PET and CT studies proceeded under normal tidal breathing. All PET images were reconstructed using an iterative algorithm with CT-derived attenuation correction. The SUVmaxfor each patient was established by drawing regions of interest around the primary tumor on attenuation-corrected FDG-PET images and calculated using the software within the PET/CT scanner based on the following formula: SUVmax = [C(μCi/mL)/ID(μCi)]/w, where C represents activity at a pixel within the tissue identified by regions of interest and ID represents the injected dose/kg of body weight (w). We used SUVmaxin the present analysis because it is less variable than mean SUV in terms of measurements.25

Clinicopathological factors

The optimal SUVmax cut-off was compared with clinicopathological factors such as age, tumor size, lymph nodes metastasis, nuclear grade, estrogen receptor (ER) positivity, and progesterone receptor (PgR) positivity, which are established prognostic factors for breast carcinoma. Nuclear grade was determined according to General Rules for Clinical and Pathological Recording of Breast Cancer, 16th edition.26 Positive ER and PgR were assessed by immunohistochemistry (IHC) and scored according to the Allred system. HER-2 positivity was defined as 3+ by IHC or 2+ by gene amplification using fluorescent in situ hybridization (FISH) >2.2.

 

Follow-up evaluation

All patients were followed up from the day of surgery. Recurrence was defined as any unequivocal occurrence of new cancer foci in a disease-free patient. The site of the first cancer recurrence and the interval between the surgery and recurrence were determined. The duration of DFS was calculated as the interval between the date of surgery and that of the first confirmation of cancer recurrence, death without evidence of recurrence, or the last clinical contact attesting to disease-free status.

Statistical analysis

Data are presented as numbers (%) or means unless otherwise stated. Frequencies were compared using the χ2 test for categorical variables, and Fisher's exact test was applied to small samples. Continuous variables were compared using the Mann–Whitney U test. Receiver operating characteristic (ROC) curves of SUVmax for the prediction of recurrence were generated to determine the cut-off value that yielded optimal sensitivity and specificity from Youden Index. The patient population was subdivided on the basis of a cut-off value for SUVmax derived from ROC curves, and the duration of DFS was analyzed using the Kaplan–Meier method. Differences in DFS were assessed using the log-rank test. The potential independent effects of SUVmax on DFS were assessed by multivariate analyses using the Cox proportional hazards model (backward stepwise selection) and p < 0.05 was considered statistically significant. Data were statistically analyzed using SPSS software (version 10.5, SPSS Inc., Chicago, IL, USA).

Results

Patient and tumor characteristics

Table 1 shows the clinicopathological characteristics of the 311 patients. A total of 261 (83.9%) tumors were ER-positive and 236 (75.9%) were PgR-positive according to IHC, and 50 (16.1%) were HER-2-positive according to IHC or FISH. Patients in whom enhanced computed tomography or FDG-PET/CT uncovered no evidence of distant metastatic spread were eligible for primary breast cancer surgery. Sixty (19.3%) patients had received neoadjuvant chemotherapy and 111 (35.7%) patients had received adjuvant chemotherapy. 213 (68.5%) patients had received radiotherapy to the breast or chest wall. All patients with hormone receptor-positive breast cancer had received adjuvant hormonal therapy.

Table 1. Patient's characteristics.

Variables n = 311 %
Mean age (y, range) 58.4 (28–91)  
Mean SUVmax (range) 3.75 (0.6–22.1)  
Histology
 IDC 267 85.9%
Special type 44 14.1%
T stage
 T1 223 71.7%
 T2 88 24.8%
 T3 3 1.0%
 T4 7 2.3%
Nodal status
 Negative 212 68.2%
 Positive 99 31.8%
Nuclear grade
 I 29 9.3%
 II 131 42.1%
 III 147 47.3%
 Missing 4 1.2%
Lymphovascular invasion
 Negative 198 63.7%
 Positive 110 35.4%
 Missing 3 0.9%
Estrogen receptor status
 Negative 50 16.1%
 Positive 261 83.9%
Progesterone receptor status
 Negative 75 24.1%
 Positive 236 75.9%
HER2 status
 Negative 261 83.9%
 Positive 50 16.1%
Triple-negative status
 No 281 90.4%
 Yes 30 9.6%
Adjuvant treatment
 Neoadjuvant chemotherapy (CT) 60 19.3%
 Adjuvant CT 111 35.7%
 Radiotherapy (RT) 213 68.5%
 CT + RT 117 37.6%

HER2, human epidermal growth factor receptor 2; IDC, invasive ductal carcinoma.

The ROC curve showed an optimal SUVmax cut-off value of 3.8 for predicting recurrence (n = 311, area under the curve [AUC], 0.789; sensitivity, 83.3%; specificity, 68.2%; Fig. 1).

Full-size image (17 K)

Fig. 1. ROC curves of SUVmax for predicting cancer recurrence in all patients. Optimal cut-off of SUVmax was 3.8 (n = 311, AUC = 0.789; 95% CI, 0.6584–0.920; p = 0.01).

At a cut-off value of 3.8, a high SUVmax significantly associated with large tumor, lymph node positivity, nuclear grade III, lymphovascular invasion positivity, hormone receptor negativity and HER2 positivity (Table 2).

Table 2. Comparison of clinicopathological parameters between SUVmax ≤3.8 and >3.8 in all patients.

Variable SUVmax ≤ 3.8 (n = 205) SUVmax > 3.8 (n = 106) p
Mean age 59.8 (30–91) 55.9 (28–79) 0.008
T stage
 T1 180 (87.7%) 43 (40.6%) <0.001
 T2,T3,T4 25 (12.2%) 63 (59.4%)
Nodal status
 Negative 148 (72.2%) 64 (60.4%) 0.040
 Positive 57 (27.8%) 42 (39.6%)
Nuclear grade
 I, II 127 (62.3%) 33 (32.0%) <0.001
 III 77 (37.7%) 70 (68.0%)
Lymphovascular invasion
 Negative 140 (68.6%) 58 (55.8%) 0.032
 Positive 64 (31.4%) 46 (44.2%)
Hormone receptor status
 Negative 20 (9.8%) 30 (28.3%) <0.001
 Positive 185 (90.2%) 76 (71.7%)
HER2 status
 Negative 180 (87.8%) 81 (76.4%) 0.014
 Positive 25 (12.2%) 25 (23.6%)

HER2, human epidermal growth factor receptor 2; IDC, invasive ductal carcinoma.

 

Fig. 2 shows a significant difference in DFS between patients with SUVmax ≤ 3.8 and >3.8 (3-year DFS rates: 98.8% vs. 91.6%; p < 0.001). The variables included in the univariate analysis of DFS in the 311 patients were SUVmax, tumor size, nodal status, nuclear grade, lymphovascular invasion status, hormone receptor status, HER2 status, and adjuvant treatment (Table 3). High SUVmax, large tumors and negative hormone receptor status were significantly associated with a short DFS. Moreover, multivariate analysis that included SUVmax, tumor size, nodal status, nuclear grade, as well as the status of lymphovascular invasion, hormone receptors, HER2, and adjuvant treatment showed that SUVmax (hazard ratio: 1.18, 95% CI: 1.07–1.30; p = 0.001) and negative hormone receptor status (hazard ratio: 6.02, 95% CI: 1.72–21.1; p = 0.005) were independent prognostic factors for DFS (Table 3).

Full-size image (21 K)

Fig. 2. DFS curves according to SUVmax values for all patients. Three-year DFS of 98.8% and 91.6% for SUVmax ≤3.8 and >3.8, respectively (p < 0.001).

Table 3. Univariate and multivariate analyses of disease-free survival in all patients.

Variable   HR (95%CI) p
Univariate analysis
 SUVmax High 1.24 (1.13–1.36) <0.001
 T stage T2,3,4 5.67 (1.70–18.9) 0.005
 Nodal status Positive 2.05 (0.662–6.37) 0.205
 Nuclear grade III 1.55 (0.468–5.13) 0.473
 Lymphovascular invasion Positive 3.07 (0.896–10.5) 0.074
 Hormone receptor status Negative 8.32 (2.63–26.3) <0.001
 HER2 status Positive 1.58 (0.425–5.86) 0.496
 (Neo-) Adjuvant chemotherapy Done 5.24 (1.14–24.0) 0.033
 Radiotherapy Done 0.845 (0.254–2.82) 0.784
 
Multivariate analysis
 SUVmax High 1.18 (1.07–1.30) 0.001
 T stage T2,3,4 2.50 (0.591–10.6) 0.213
 Nodal status Positive 1.88 (0.443–7.97) 0.393
 Nuclear grade III 0.347 (0.076–1.58) 0.172
 Lymphovascular invasion Positive 3.29 (0.952–11.4) 0.060
 Hormone receptor status Negative 6.02 (1.72–21.1) 0.005
 HER2 status Positive 1.69 (0.394–7.21) 0.482
 (Neo-) Adjuvant chemotherapy Done 0.957 (0.142–6.47) 0.964
 Radiotherapy Done 3.14 (0.551–17.9) 0.197

HER2, human epidermal growth factor receptor 2.

The patients were categorized into four subtypes based upon hormone receptor and HER2 status. We identified 12 recurrent events in all patients, of which 6 (50%) occurred in patients with the triple-negative breast cancer subtype (hormone receptor-negative and HER2-negative). Patients with this subtype had early relapse and a poor prognosis. We then evaluated the SUVmax value as a prognostic factor in this subtype. The SUVmax ranged from 0.8 to 22.1, and the mean SUVmax in the triple-negative subtype was 6.32, which was considerably higher than of the value of 3.75 for all patients (Fig. 3). An analysis of ROC curves revealed an optimal SUVmax cut-off value of 8.6 for predicting recurrence of the triple-negative subtype (n = 30; AUC, 0.767; sensitivity, 66.7%; specificity, 80.0%; Fig. 4).

Full-size image (29 K)

Fig. 3. Histograms of SUVmax distribution in primary tumor.

Full-size image (21 K)

Fig. 4. ROC curves of SUVmax for predicting cancer recurrence in triple-negative patients. Optimal cut-off of SUVmax was 8.6 (n = 30, AUC = 0.767; 95% CI, 0.528–0.100; p = 0.046).

At a cut-off value of 8.6, a high SUVmax significantly associated with large tumors (Table 4). Fig. 5 shows a significant difference in DFS between patients who had SUVmax ≤ 8.6 and >8.6 (3-year DFS rates: 90.9% vs. 42.9%; p = 0.002).

 

Table 4. Comparison of clinicopathological parameters between SUVmax ≤8.6 and >8.6 in triple-negative patients.

Variables SUVmax ≤ 8.6 (n = 23) SUVmax > 8.6 (n = 7) p
T stage
 T1 16 (69.6%) 0 (0%) 0.002
 T2,T3,T4 7 (30.4%) 7 (100%)
Nodal status
 Negative 15 (65.2%) 5 (71.4%) 1.0
 Positive 8 (34.8%) 2 (28.6%)
Nuclear grade
 I, II 11 (47.8%) 0 (0%) 0.125
 III 12 (52.2%) 5 (100%)
Lymphovascular invasion
 Negative 16 (69.6%) 3 (50.0%) 0.633
 Positive 7 (30.4%) 3 (50.0%)
Full-size image (19 K)

Fig. 5. DFS curves according to SUVmax values for triple-negative patients. Three-year DFS of 90.9% and 42.9% for SUVmax ≤8.6 and >8.6, respectively (p = 0.002).

Univariate analysis of DFS in the 30 patients with the triple-negative subtype included SUVmax, tumor size, nodal status, nuclear grade, and lymphovascular invasion status as variables (Table 5). A high SUVmax was significantly associated with a short DFS. Moreover, multivariate analysis showed that SUVmax (hazard ratio: 1.27, 95% CI: 1.00–1.27; p = 0.047) was an independent prognostic factor for DFS (Table 5).

Table 5. Univariate and multivariate analyses of disease-free survival in triple-negative patients.

Variable   HR (95%CI) p
Univariate analysis
 SUVmax High 1.14 (1.02–1.27) 0.018
 T stage T2,T3,T4 7.16 (0.819–26.2) 0.073
 Nodal status Positive 2.19 (0.442–10.9) 0.337
 Nuclear grade III 2.96 (0.329–26.4) 0.334
 Lymphovascular invasion Positive 3.14 (0.524–18.8) 0.210
 
Multivariate analysis
 SUVmax High 1.13 (1.00–1.27) 0.047
 T stage T2,T3,T4 2.50 (0.179–35.0) 0.495
 Nodal status Positive 3.84 (0.590–25.0) 0.159
 Nuclear grade III 0.462 (0.016–13.1) 0.651
 Lymphovascular invasion Positive 1.18 (0.140–10.0) 0.877

Discussion

A high SUVmax significantly associated with poor prognostic features in the present study and thus could serve as an independent prognostic factor for patients with operable breast cancer, especially those with the triple-negative subtype.

Some studies have suggested that SUVmax in the primary tumor might be associated with several pathological prognostic factors19, 20, 21, 22, 23 and 24; however, most of these studies did not analyze survival. Oshida and colleagues identified FDG uptake as an independent prognostic factor for the relapse-free survival of patients with breast cancer, along with the number of positive lymph nodes and histological grade.27 They used the differential absorption ratio (DAR) instead of the SUVmax, as an index of FDG uptake. Without using ROC curves, they calculated statistically significant differences at a cut-off point that was set to 1.0 and then increased the value in stepwise increments of 0.5 up to 5.0 between the two groups using the log-rank test. The difference in both overall survival and relapse-free survival rates was the most significant at a cut-off of 3.0. The present findings that poor prognostic features (large tumor size, positive lymph node metastasis, tumor grade III, positive lymphovascular invasion, hormone receptor negativity, and HER2 positivity) were closely associated with our cut-off SUVmax of 3.8 were similar to those of Oshida and colleagues. A high SUVmax was an independent and poor prognostic factor in the multivariate analysis. Further validation studies are required to evaluate the appropriateness of our cut-off values.

We discovered that SUVmax can discriminate two prognoses for patients with the triple-negative subtypes. The mean SUVmax was higher in these patients than in the total number of patients, and a high SUVmax was an independent poor prognostic factor in multivariate analysis. Although triple-negative breast cancer comprises only 15–20% of all breast cancers, the lack of targeted therapy for this subgroup poses therapeutic challenges.28 The ability of platinum chemotherapy alone or in combination with PARP-1 inhibitors to treat triple-negative breast cancer has been investigated in new trials.29 and 30 Triple-negative patients with a high SUVmax were predicted to have early relapse and to require new aggressive treatment strategies. Julia Tchou et al. found an association between SUVmax and Ki67 in patients with triple-negative breast cancer and suggested that PET could be used to monitor the treatment response of such patients.31

The threshold for recommending chemotherapy for patients with hormone receptor-positive and the HER2-negative subtype is particularly difficult to define. The St. Gallen international expert consensus on the primary therapy of early breast cancer 2009 adopted Ki67 as a proliferation index to indirectly support the rationale for augmenting chemotherapy with endocrine therapy in such patients.32 The main disadvantage of Ki67 is the high degree of interobserver variability in assessments.33 Ki67 LI values can vary as a function of several critical factors, including human error, Ki67 selection of the tumor areas to be counted and the specific antibody used.34 SUVmax has similar issues associated with assessment uniformity. However, Nakayama et al. suggested that phantom studies of lung adenocarcinoma can overcome the difficulties in multicenter studies using PET for lung adenocarcinoma.35 SUVmax could substitute for Ki67 as an index of adding chemotherapy. The limitations of our study include a small sample cohort, its retrospective design at a single institution, and the short follow-up period that did not allow adequate assessment of the hormone receptor-positive and HER2 negative-subtype. In spite of the relatively small sample and the small number of events, SUVmax was found to be a strong independent prognostic factor for DFS. Further studies are required to evaluate whether SUVmax has prognostic value in each breast cancer subtype. We are currently investigating in the multi-institutional prospective study.

In conclusion, we demonstrated that high SUVmax in a primary site was an independent prognostic factor for operable breast cancer. PET/CT is a valuable tool to provide information on initial staging and on determining the need for more aggressive therapy to prevent early relapse especially in patients with breast cancer who have triple-negative subtype. SUVmax in a primary site might be used for the new clinical trial about adjuvant chemotherapy such as dose dense chemotherapy or adding new drugs. Conversely, patients with low SUVmax had a good prognosis and might be omitted adjuvant chemotherapy, even with the triple-negative subtype. However, using PET/CT for the purpose stated in our research has not been investigated in cost-effectiveness studies.

Conflict of interest statement

None of the authors have any conflict of interest.

 

References

    • 1
    • J.F. Vansteenkiste, S.G. Stroobants, P.J. Dupont, P.R. De Leyn, E.K. Verbeken, G.J. Deneffe, Leuven Lung Cancer Group et al.
    • Prognostic importance of the standardized uptake value on (18)F-fluoro-2-deoxy-glucose-positron emission tomography scan in non-small-cell lung cancer: an analysis of 125 cases

    • J Clin Oncol, 17 (1999), pp. 3201–3206

    • 2
    • J.F. Santiago, M. Gonen, H. Yeung, H. Macapinlac, S. Larson
    • A retrospective analysis of the impact of 18F-FDG PET scans on clinical management of 133 breast cancer patients

    • Q J Nucl Med Mol Imaging, 50 (2006), pp. 61–67

    • 3
    • E.R. Port, H. Yeung, M. Gonen, L. Liberman, J. Caravelli, P. Borgen et al.
    • 18F-2-fluoro-2-deoxy-D-glucose positron emission tomography scanning affects surgical management in selected patients with high-risk, operable breast carcinoma

    • Ann Surg Oncol, 13 (2006), pp. 677–684

    •  | 
    • 4
    • J. Okada, K. Yoshikawa, M. Itami, K. Imaseki, K. Uno, J. Itami et al.
    • Positron emission tomography using fluorine-18-fluorodeoxyglucose in malignant lymphoma: a comparison with proliferative activity

    • J Nucl Med, 33 (1992), pp. 325–329

    • 5
    • K. Kubota, T. Matsuzawa, T. Fujiwara, M. Ito, J. Hatazawa, K. Ishiwata et al.
    • Differential diagnosis of lung tumor with positron emission tomography: a prospective study

    • J Nucl Med, 31 (1990), pp. 1927–1932

    • 6
    • S. Okazumi, K. Isono, K. Enomoto, T. Kikuchi, M. Ozaki, H. Yamamoto et al.
    • Evaluation of liver tumors using fluorine-18-fluoro-2-deoxy-D-glucose PET

    • J Nucl Med, 33 (1992), pp. 339–344

    •  
    • 7
    • D. Groheux, S. Giacchetti, D. Rubello, A. Al-Nahhas, J.L. Moretti, M. Espié et al.
    • The evolving role of PET/CT in breast cancer

    • Nucl Med Commun, 31 (2010), pp. 271–273

    •  | 
    • 8
    • A. Berriolo-Riedinger, C. Touzery, J.M. Riedinger, M. Toubeau, B. Coudert, L. Arnould et al.
    • [18F]FDG-PET predicts complete pathological response of breast cancer to neoadjuvant chemotherapy

    • Eur J Nucl Med Mol Imaging, 34 (2007), pp. 1915–1924

    •  | 
    • 9
    • H. Schirrmeister, T. Kuhn, A. Guhlmann, C. Santjohanser, T. Hörster, K. Nüssle et al.
    • Fluorine-18 2-deoxy-2-fluoro-Dglucose PET in the preoperative staging of breast cancer: comparison with the standard staging procedures

    • Eur J Nucl Med, 28 (2001), pp. 351–358

    •  | 
    • 10
    • D. Fuster, J. Duch, P. Paredes, M. Velasco, M. Muñoz, G. Santamaría et al.
    • Preoperative staging of large primary breast cancer with [18F] fluorodeoxyglucose positron emission tomography/computed tomography compared with conventional imaging procedures

    • J Clin Oncol, 26 (2008), pp. 4746–4751

    •  | 
    • 11
    • J. Dose, C. Bleckmann, S. Bachmann, K.H. Bohuslavizki, J. Berger, L. Jenicke et al.
    • Comparison of fluorodeoxyglucose positron emission tomography and “conventional diagnostic procedures” for the detection of distant metastases in breast cancer patients

    • Nucl Med Commun, 23 (2002), pp. 857–864

    •  | 
    • 12
    • B. Klaeser, O. Wiederkehr, D. Koeberle, A. Mueller, B. Bubeck, B. Thuerlimann
    • Therapeutic impact of 2-[fluorine-18] fluoro-2-deoxy-D-glucose positron emission tomography in the pre- and postoperative staging of patients with clinically intermediate or high-risk breast cancer

    • Ann Oncol, 18 (2007), pp. 1329–1334

    •  | 
    • 13
    • M.L. Landheer, M.G. Steffens, J.H. Klinkenbijl, A.H. Westenberg, W.J. Oyen
    • Value of fluorodeoxyglucose positron emission tomography in women with breast cancer

    • Br J Surg, 92 (2005), pp. 1363–1367

    •  | 
    • 14
    • A. Dirisamer, B.S. Halpern, D. Flory, F. Wolf, M. Beheshti, M.E. Mayerhoefer et al.
    • Integrated contrast-enhanced diagnostic whole-body PET/CT as a first-line restaging modality in patients with suspected metastatic recurrence of breast cancer

    • Eur J Radiol, 73 (2010), pp. 294–299

    •  |   | 
    • 15
    • M. Okada, H. Nakayama, S. Okumura, H. Daisaki, S. Adachi, M. Yoshimura et al.
    • Multicenter analysis of high-resolution computed tomography and positron emission tomography/computed tomography findings to choose therapeutic strategies for clinical stage IA lung adenocarcinoma

    • J Thorac Cardiovasc Surg, 131 (2011), pp. 1384–1391

    •  |   | 
    • 16
    • Y. Tsutani, Y. Miyata, K. Misumi, T. Ikeda, T. Mimura, J. Hihara et al.
    • Difference in prognostic significance of maximum standardized uptake value on [18F]-fluoro-2-deoxyglucose positron emission tomography between adenocarcinoma and squamous cell carcinoma of the lung

    • Jpn J Clin Oncol, 41 (2011), pp. 890–896

    •  | 
    • 17
    • A. Buck, H. Schirrmeister, T. Kuhn, C. Shen, T. Kalker, J. Kotzerke et al.
    • FDG uptake in breast cancer: correlation with biological and clinical prognostic parameters

    • Eur J Nucl Med Mol Imaging, 29 (2002), pp. 1317–1323

    •  | 
    • 18
    • T.A. Smith, R.I. Sharma, A.M. Thompson, F.E. Paulin
    • Tumor 18F-FDG incorporation is enhanced by attenuation of P53 function in breast cancer cells in vitro

    • J Nucl Med, 47 (2006), pp. 1525–1530

    • 19
    • F. Dehdashti, J.E. Mortimer, B.A. Siegel, L.K. Griffeth, T.J. Bonasera, M.J. Fusselman et al.
    • Positron tomographic assessment of estrogen receptors in breast cancer: comparison with FDG-PET and in vitro receptor assays

    • J Nucl Med, 36 (1995), pp. 1766–1774

    • 20
    • F. Crippa, E. Seregni, R. Agresti, C. Chiesa, C. Pascali, A. Bogni et al.
    • Association between [18F]fluorodeoxyglucose uptake and postoperative histopathology, hormone receptor status, thymidine labelling index and p53 in primary breast cancer: a preliminary observation

    • Eur J Nucl Med, 25 (1998), pp. 1429–1434

    •  | 
 
    • 21
    • N. Avril, M. Menzel, J. Dose, M. Schelling, W. Weber, F. Jänicke et al.
    • Glucose metabolism of breast cancer assessed by 18F-FDG PET: histologic and immunohistochemical tissue analysis

    • J Nucl Med, 42 (2001), pp. 9–16

    • 22
    • R. Bos, J.J. van Der Hoeven, E. van Der Wall, P. van Der Groep, P.J. van Diest, E.F. Comans et al.
    • Biologic correlates of (18)fluorodeoxyglucose uptake in human breast cancer measured by positron emission tomography

    • J Clin Oncol, 20 (2002), pp. 379–387

    •  | 
    • 23
    • R. Kumar, A. Chauhan, H. Zhuang, P. Chandra, M. Schnall, A. Alavi
    • Clinicopathologic factors associated with false negative FDG-PET in primary breast cancer

    • Breast Cancer Res Treat, 98 (2006), pp. 267–274

    •  | 
    • 24
    • D. Groheux, S. Giacchetti, J.L. Moretti, R. Porcher, M. Espié, J. Lehmann-Che et al.
    • Correlation of high 18F-FDG uptake to clinical, pathological and biological prognostic factors in breast cancer

    • Eur J Nucl Med Mol Imaging, 38 (2011), pp. 426–435

    •  | 
    • 25
    • J.R. Lee, M.T. Madsen, D. Bushnel, Y. Menda
    • A threshold method to improve standardized uptake value reproducibility

    • Nucl Med Commun, 21 (2000), pp. 685–690

    •  | 
    • 26
    • The Japanese Breast Cancer Society
    • General rules for clinical and pathological recording of breast cancer

    • (16th ed.)Kanehara, Tokyo (2008)

    •  
    • 27
    • M. Oshida, K. Uno, M. Suzuki, T. Nagashima, H. Hashimoto, H. Yagata et al.
    • Predicting the prognoses of breast carcinoma patients with positron emission tomography using 2-deoxy-2-fluoro[18F]-D-glucose

    • Cancer, 82 (1998), pp. 2227–2234

    •  | 
    • 28
    • W.D. Foulkes, I.M. Stefansson, P.O. Chappuis, L.R. Bégin, J.R. Goffin, N. Wong et al.
    • Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer

    • J Natl Cancer Inst (2003), pp. 1482–1485

    •  | 
    • 29
    • J. O'Shaughnessy, C. Osborne, J.E. Pippen, M. Yoffe, D. Patt, C. Rocha et al.
    • Iniparib plus chemotherapy in metastatic triple-negative breast cancer

    • N Engl J Med, 364 (2011), pp. 205–214

    •  | 
    • 30
    • D.P. Silver, A.L. Richardson, A.C. Eklund, Z.C. Wang, Z. Szallasi, Q. Li et al.
    • Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer

    • J Clin Oncol, 28 (2010), pp. 1145–1153

    •  | 
    • 31
    • J. Tchou, S.S. Sonnad, M.R. Bergey, S. Basu, J. Tomaszewski, A. Alavi et al.
    • Degree of tumor FDG uptake correlates with proliferation index in triple-negative breast cancer

    • Mol Imaging Biol, 12 (2010), pp. 657–662

    •  | 
    • 32
    • A. Goldhirsch, J.N. Ingle, R.D. Gelber, A.S. Coates, B. Thürlimann, H.J. Senn et al.
    • Thresholds for therapies: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer

    • Ann Oncol, 20 (2009), pp. 1319–1329

    •  | 
    • 33
    • B.V. Offersen, F.B. Sorensen, A. Knoop, J. Overgaard, Danish Breast Cancer Cooperative Tumour Biology Committee et al.
    • The prognostic relevance of estimates of proliferative activity in early breast cancer

    • Histopathology, 43 (2003), pp. 573–582

    •  | 
    • 34
    • S. Fasanella, E. Leonardi, C. Cantaloni, C. Eccher, I. Bazzanella, D. Aldovini et al.
    • Proliferative activity in human breast cancer:Ki-67 automated evaluation and the influence of different Ki-67 equivalent antibodies

    • Diagn Pathol, 6 (2011), p. S7

    • 35
    • H. Nakayama, S. Okumura, H. Daisaki, Y. Kato, H. Uehara, S. Adachi et al.
    • Value of integrated positron emission tomography revised using a phantom study to evaluate malignancy grade of lung adenocarcinoma: a multicenter study

    • Cancer, 116 (2010), pp. 3170–3177

学科代码:肿瘤学   关键词:EJC全文 EJC
顶一下(0
您可能感兴趣的文章
    发表评论网友评论(0)
      发表评论
      登录后方可发表评论,点击此处登录