touchONCOLOGY touchONCOLOGY
Breast Cancer, Diagnostics and Screening
Read Time: 2 mins

Positron-emission Tomography Imaging in Breast Cancer

Copy Link
Published Online: Sep 6th 2011 US Oncology & Hematology, 2011;7(2):130-7 DOI: https://doi.org/10.17925/OHR.2011.07.2.130
Authors: William B Eubank, Jean H Lee, David A Mankoff
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

There are four major diagnostic tasks in breast cancer: detection and diagnosis; staging (locoregional and distant metastases); response to treatment (neoadjuvant and metastatic therapy); and characterization of the individual tumor for selection of the most appropriate therapy. The use of fluorodeoxyglucose (FDG) positron-emission tomography (PET) and PET–computed tomography (PET-CT) in all four of these tasks has been investigated. The aim of this article is to discuss the strengths and weaknesses of FDG PET with regard to each of these tasks. The diagnostic areas for which FDG PET appears most suited for clinical decision-making will be highlighted.

Keywords

Fluorodeoxygluxcose positron-emission tomography (FDG PET), positron-emission tomography–computed tomography (PET-CT), recurrence, metastases, breast cancer, restaging

Article:

Detection of Primary Tumor
In breast cancer, the word ‘detection’ is most commonly used to mean breast cancer screening, most commonly using mammography. Breast cancer diagnosis involves the characterization of a suspicious mass or imaging finding, and entails tissue sampling to make a definitive diagnosis of cancer versus benign disease. Most primary cancers are detected by physical examination or mammography during screening.1 Mammography is the primary imaging modality for breast cancer screening, detection, and diagnosis.2 Both ultrasound and magnetic resonance imaging (MRI) are important adjuncts to X-ray mammography for diagnosis, characterization, and determination of the extent of breast cancer (evaluation of multifocal or multicentric disease), and are routinely utilized in this role.3,4 Mammography, ultrasound, and breast MRI can all be used to direct tissue sampling by needle biopsy for breast cancer diagnosis. MRI has also shown utility for screening in highrisk women, and was recently incorporated in American Cancer Society (ACS) recommendations for screening in high-risk patients.5

The level of fluorodeoxyglucose (FDG) uptake in primary breast neoplasms reflects the rate of glucose metabolism and has been correlated with known prognostic factors and biologic characteristics such as tumor size, histologic type (higher uptake in ductal versus lobular), tumor histologic grade, and some indices of cellular proliferation (higher uptake with higher levels of proliferation).6–11 Overall, the sensitivity of FDG positron-emission tomography (PET) in detecting primary breast cancer is 64–96 %, specificity is 73–100 %, positive predictive value is 81–100 %, and negative predictive value is 52–89 %,1 but in one large study12 the sensitivity was 57 % for lesions <1 cm compared with 91 % for tumors >1 cm and 25 % for in situ carcinoma. Another study confirmed the relatively poor sensitivity of whole-body FDG PET with small tumors (<1 cm), lobular histologic type, and well-differentiated in situ breast carcinoma.13 As primary breast cancer detection requires the ability to depict occult, non-palpable, small (<1 cm) invasive, and in situ malignant lesions, whole-body FDG PET is not used in primary breast cancer detection. Dedicated breast positron-emission mammography (PEM) units have been developed.14 The advantages of PEM include higher spatial resolution, shortened imaging time, and reduced attenuation compared with whole-body imaging. Early studies demonstrated the feasibility of PEM for detecting smaller breast tumors (reviewed in reference 14), and a recent multicenter trial also suggested that PEM may aid in the detection and characterization of both invasive and in situ breast carcinoma.15 Current limitations for PEM include imaging posterior lesions, variable FDG uptake in small tumors, and false-positive findings from prior biopsy.15,16 Recently, PEM with biopsy capability has been developed and is being tested.17 The clinical data regarding dedicated positron breast imaging devices are still limited, particularly compared with the large amount of data supporting and validating screening mammography and other adjunctive primary breast imaging modalities such as ultrasound and breast MRI. The utility of PEM compared with existing breast imaging methods awaits validation in larger prospective trials before more general acceptance and more widespread clinical use.

Staging at Presentation
The extent of spread at presentation is one of the most important prognostic factors for breast carcinoma.18 Staging is typically divided into locoregional staging (for regional nodes, especially axillary nodes) and distant or systemic staging (for sites beyond locoregional nodes). The role of imaging in initial staging of breast cancer patients should be carefully considered in terms of performance and cost–benefit analysis. For example, the utility in staging asymptomatic patients with early breast cancer for distant metastases at presentation using conventional imaging (CI) has been shown to be very low in a number of studies, with overall prevalence of distant metastasis of under 2.5 %.19–23 The experience of using whole-body FDG PET and PET–computed tomography (PET-CT) in the initial staging of early breast cancer is small, but has also confirmed these findings.24,25 In this population with a low prevalence of disease at distant sites, the false/true-positive ratio is too high (with any imaging modality) to be cost-effective and could lead to many unnecessary additional tests and biopsies. For these reasons, whole-body FDG PET-CT should be avoided in patients with early breast cancer.

Axillary Staging
For all patients with newly diagnosed invasive disease (all stages), the status of the axillary nodes is important for prognosis and determining adjuvant therapy.26–28 For this reason, and since axillary nodes are easily accessible, axillary node status is confirmed following removal and histologic examination. Sentinel lymph node biopsy (SNLB) has gained widespread acceptance over the last 10 years to minimize the invasiveness of sampling axillary modes in patients with early breast cancer (tumor <2 cm on mammography, ultrasound, or breast MR and clinically negative axillae). This procedure spares many patients the morbidity associated with complete axillary lymph node dissection (ALND). The reported false-negative rate of SNLB of 5–10 % (with no false-positives) has contributed to making this the standard method of staging the axilla in patients with early breast cancer.29–31 Many early studies of FDG PET focused on detecting axillary metastases.32–36 However, these studies included patients with more advanced primary tumors, increasing the pre-test likelihood of positive PET. The specificity has been consistently high across studies, ranging from 80 to 100 %.37 In the largest prospective multicenter trial so far, of 360 patients,38 FDG PET was 61 % sensitive and 80 % specific for axillary metastases. Patients who had false-negative PET results had significantly smaller and fewer tumor-positive lymph nodes than true-positive cases. This limitation of FDG PET to detect small-volume axillary disease (micrometstasis, small volume of infiltration, and solitary or few positive nodes) has been confirmed in more recent series of FDG PET for staging axillae in early breast cancer with SNLB as the reference standard.13,39–48 These studies, which have included a larger proportion of T1 tumors, have demonstrated a much lower sensitivity for axillary metastases—as low as 20–40 %—particularly in smaller (ranging from 1 to 15 mm) and fewer positive nodes. Thus, FDG PET-CT is not sufficiently accurate to replace SNLB in patients with early-stage breast cancer. FDG PET-CT may have a role for patients with a higher risk of axillary metastases such as those with locally advanced breast cancer (LABC; stage IIb or higher) (see Figure 1). Due to its high specificity, pre-operative FDG PET-CT may direct these patients to ALND, bypassing SNLB, in the case of positive FDG uptake in the axilla. Ultrasound, in conjunction with fine-needle aspiration, is widely used to evaluate any suspicious or palpaple nodes in patients prior to undergoing SNLB.49–52 FDG PET-CT may be used to complement the ultrasound evaluation in the case of equivocal or negative findings. This approach has been previously suggested but needs to be validated in larger prospective trials.41,43,48,53–56

Extra-axillary Locoregional Nodal Staging
Detection of disease in nodal regions not addressed with ALND, i.e. level III (apex of axilla) and extra-axial locoregional nodes, has important prognostic and therapeutic implications.57,58 Several recent studies have shown the benefit of staging patients with newly diagnosed LABC or inflammatory breast cancer (IBC) with FDG PET or PET-CT.59–63 In patients with large (but not inflammatory) primary tumors, FDG uptake in extra-axial regional lymph nodes, including subpectoral, infraclavicular, supraclavicular, and internal mammary (IM) sites, was present in 7–8 % of patients,61,62 and distant metastases were detected in 10–28 % of patients.24,61–63 These findings led to a change in staging of 18–42 % of patients and a change in treatment plan in 13 % of patients.62 The benefit of staging patients with IBC may be even stronger.60,64,65 A retrospective study of 41 patients with IBC60 showed FDG uptake in extra-axial regional lymph nodes and distant sites in 44 % (n=18) and 49 % (n=20) of patients, respectively, and 35 % of patients (seven of 20) with distant metastasis were unsuspected by CI. FDG PET is also useful in detecting recurrent disease in extra-axillary locoregional nodes in patients previously treated for their primary tumor (see Figure 2).66

Distant Metastasis Staging
Although staging for distant metastases is most commonly performed in patients with advanced breast cancer having undergone primary treatment (and possibly more), it is also appropriate for patients with newly diagnosed LABC.67,68 FDG PET can be helpful in the evaluation for distant metastases in patients who have equivocal CI (CT, ultrasound, bone scintigraphy) findings and in asymptomatic patients with elevated tumor markers. In terms of diagnostic performance, several studies have shown FDG PET to have a relative advantage over CI in the evaluation of distant metastases in previously treated patients.69–81 In a meta-analysis of FDG PET for the evaluation of breast recurrence and metastases, Isasi et al.82 reported a median sensitivity and specificity of 93 and 82 %, respectively, in a patient-based analysis. A common finding in these studies comparing FDG PET with CI for the detection of recurrent disease is that FDG PET detects a significantly greater number (about two-fold) of extra-axial lymph node metastases,63,69–71,78,79 including mediastinanal and IM nodes.66 Bone is the most frequent site of recurrence after treatment for primary breast cancer: nearly 70 % of patients with advanced disease have skeletal metastases.83 Metastases from breast cancer can produce a varied physiologic response in bone; lesions can be osteolytic, osteoblastic, or a mixture of the two. Cook et al.84 were the first investigators to correlate the diagnostic performance of FDG PET and bone scintigraphy with the morphologic appearance of individual skeletal metastases at plain film radiography or CT. They showed that FDG PET was superior to bone scintigraphy in the detection of osteolytic metastases, and bone scintigraphy detected significantly more osteoblastic metastases. Others have corroborated these findings,85–89 leading to the general conclusion that FDG PET and bone scintigraphy are complementary methods for the detection of skeletal metastases in breast cancer patients. In our center, bone scintigraphy remains one of the routine studies in breast cancer metastatic staging, with FDG PET-CT to help clarify staging in the case of difficult or equivocal conventional staging. Evolving data suggest that [F-18]-fluoride PET and [F-18]-fluoride PET-CT may improve skeletal metastasis detection compared with bone scintigraphy90–92 and may play a role in breast cancer skeletal metastasis staging in the future.

The combined PET and CT system (PET-CT) has emerged as a routine method of restaging many oncologic patients.93 Fusion of anatomic and metabolic information generally leads to an increase in diagnostic confidence, as shown in an early retrospective study.94 Several of the large retrospective studies of patients being restaged after primary treatment for breast cancer comparing the diagnostic performance of PET-CT with contrast-enhanced CT alone, PET alone, or side-by-side evaluation of CT and PET are summarized in Table 1.94–101 Fused PET-CT data consistently detected more malignant foci in these studies; however, on patient-based analysis they showed marginal (no statistical difference) improvement in sensitivity and specificity for the detection of recurrences. Improvement in accuracy occurred in evaluation of mediastinal and cervical lymph nodes and the skeleton (increased sensitivity for osteoblastic metastases).96,99,100 Treatment Response
Breast cancer is one of the more responsive solid tumors, and there is an ever-increasing choice of effective systemic therapies for breast cancer.102 Evaluating the efficacy of systemic treatment is an important diagnostic need for breast cancer. The two settings where imaging plays an important role in response to systemic therapy are patients with LABC (undergoing neoadjuvant therapy) and patients with metastatic, stage IV disease.

Locally Advanced Disease
Although neodajuvant therapy, compared with adjuvant therapy, has not been shown to improve survival, it does improve surgical options and provide prognostic information.103 Studies have demonstrated that the extent of residual breast and axillary disease after treatment is prognostic for both disease-free survival (DFS) and overall survival (OS).104–106 Patients demonstrating complete pathologic response (pCR), defined as no residual invasive tumor on histopathology at post-therapy surgery, have improved long-term outcome compared with patients without pCR.104,106 One of the primary aims of neo-adjuvant therapy is therefore to assess the response of the primary tumor to the treatment regimen.103 Size-based approaches such as physical exam and mammography have trouble distinguishing pCR from other responses.104,107 This is therefore a role where functional imaging, with its ability to serially quantify metabolic changes in the tumor earlier than morphologic changes, may be particularly helpful. Studies evaluating FDG PET for treatment response in LABC (reviewed in reference 108) have compared standardized uptake value (SUV), a semi-quantitative measure of glycolytic activity, at pre-treatment examination with values at varying times during (early and mid-point), and following completion of treatment (see Table 2). The studies in which the mid-point of therapy was evaluated showed that a drop of approximately 50 % or more in SUV from baseline predicted a good response.109–116 Studies evaluating change in FDG uptake early in the course of therapy (after one or two cycles) suggest that early assessment of response is possible and predictive of subsequent pathologic response.112–119 In a large prospective, multicenter trial of 104 patients with LABC being monitored with FDG PET during neoadjuvant therapy, a threshold of 45 % decrease in SUV after the first cycle of therapy correctly identified 11 of 15 histologic responders and the non-responders with a negative predictive value of 90 %, and similar results were found after the second cycle using a threshold of 55 % relative decrease in SUV.119 The clinical role of FDG PET for evaluating treatment response in LABC is not yet clear, but may be most useful in confirming the lack of clinical response; in this way FDG PET would help avoid ineffective treatment and its adverse effects and decide optimal time for surgery. To use FDG PET for treatment stratification, identification of non-responders with high negative predictive value early during therapy is desirable in order to avoid discontinuation of therapy to patients who might still respond to the full course of treatment. Studies performed after the completion of chemotherapy have shown that although residual FDG uptake predicts residual disease, the absence of FDG uptake is not a reliable indicator of pCR.109,120–122 Breast MR is a more sensitive technique for the detection of residual disease but has relatively poor specificity, with sensitivity of 98 % and specificity of 40 % in differentiating minimal residual from gross residual disease in one study.121 In patients with gross residual disease, post-therapy FDG PET has been shown to complement MRI to help define the extent of residual disease.123 The combination of these two imaging modalities may be the most appropriate approach to guide surgical treatment planning, but it is unlikely to replace histopathologic evaluation for the detection of residual disease. Recent studies have shown that the presence of FDG uptake after therapy is highly predictive of relapse.124 Therefore, even though FDG PET may miss small-volume disease after therapy, the presence or absence of uptake may carry prognostic significance that might help direct the intensity of additional therapy and post-surgery surveillance. Metastatic Disease
Metastatic breast cancer (MBC) is often responsive to systemic therapy and, although cure is rarely achieved, with appropriate therapy, patients often have prolonged survival and improved quality of life. As for the assessment of treatment response of LABC, conventional methods used to assess treatment response in metastases, namely whole-body CT or MRI and bone scintigraphy, can be problematic. Morphologic changes detected at CT or MRI and skeletal scintigraphic abnormalities may persist or be slow to decrease despite good response to systemic therapy and some lesions, especially skeletal, are difficult to measure using standard criteria such as Response Evaluation Criteria in Solid Tumors.125,126 Metabolic imaging with FDG PET has shown promise in making predictions of treatment response or non-response earlier and more accurately than CI. Several small studies evaluating FDG PET for measuring treatment response in MBC have shown, similar to neoadjuvant studies, that significant drops in lesion SUV, typically 40–50 % or more, from pre-therapy baseline can predict responders from non-responders more accurately than CI.127–129 The optimal time-point after initiation of therapy to evaluate MBC with FDG has not been fully determined but may be as early as after the first cycle of therapy. The level of FDG uptake after therapy, again similar to neoadjuvant studies, has also shown to be predictive of outcome in patients completing a course of high-dose chemotherapy.130 A particularly vexing clinical problem for breast cancer clinicians is the evaluation of response of skeletal metastases.131 Changes in bone scintigraphy, the standard method of evaluation of patients with bone metastases, may significantly lag response or even ‘flare’ in response to successful treatment.132,133 The differences seen between bone scintigraphy and FDG PET for bone metastases have led to some investigation into the use of serial FDG PET in assessing bone metastasis response (see Figure 3). In a retrospective study, Stafford et al.134 showed that, in patients with FDG-positive bone-dominant metastatic disease, SUV changes of an index lesion on serial FDG PET correlated with clinical assessment of response and change in tumor marker value. Comparing serial changes in SUV of index lesions to patient outcome measures, Specht et al.135 showed that percentage change in SUV is predictive of time to progression. In this retrospective study, a median decline of 41 % or greater was associated with a longer time to progression.FDG PET-CT is ideally suited for evaluating treatment response of skeletal metastases in breast cancer patients since this technique provides accurate registration of metabolic and morphologic information. Tateishi et al.136 showed, in a retrospective study of 102 patients with breast cancer skeletal metastasis undergoing systemic treatment, that a concomitant increase in CT attenuation and decrease in FDG SUV of the index lesion is predictive of a more durable response to therapy. Larger prospective trials are warranted to confirm these initial observations.

Characterization of Disease
As the array of available treatments for breast cancer increases and therapy is increasingly targeted, there is a growing need to help direct therapy. PET tracers beyond FDG can non-invasively provide quantitative information about in vivo breast cancer biology, helping to direct more individualized therapeutic choices. Preliminary work has focused on several approaches using novel PET tracers, including tumor perfusion and angiogenesis, drug delivery and transport, tumor receptor expression, and early response to treatment in breast cancer (reviewed in reference 137). It is beyond the scope of this article to discuss all of these approaches; however, the ability to image tumor receptors is particularly relevant to breast cancer. Two receptors routinely measured by in vitro assay of biopsy material, both with significant prognostic and therapeutic implications, are estrogen receptor (ER) and HER2. Most of the work to date for breast cancer tumor receptor imaging has been undertaken for steroid receptors using 16 alpha-[18F]-fluoro-17 beta-estradiol (FES).138 This tracer looks promising in preliminary trials because it can identify and quantitatively measure ER heterogenous expression—for example loss of ER expression in metastases arising from ER-expressing primary tumors139,140—and predict response to endocrine therapy.140,141 Likewise, PET imaging approaches for measuring regional HER2 expression in breast cancer also look promising.142

Summary
FDG PET and PET-CT are not sensitive enough to replace the current methods for detection and staging primary breast cancer or for staging the axilla. FDG PET-CT should not be routinely used for the initial staging exam in patients who present with early breast cancer since the prevalence of disease outside the breast and axilla is low and the false-positive/true-positive rate is too high with any staging exam in this group of patients. FDG PET-CT is helpful in staging patients who present with locally advanced and inflammatory breast cancer and for restaging patients who have undergone primary treatment and are suspected of having locoregional or distant recurrence. FDG PET has shown promise in its ability to predict response to systemic therapy both for patients with locally advanced disease receiving neoadjuvant treatment and in the metastatic setting. Finally, advances in PET imaging in breast cancer continue to be made with the use of novel tracers that can help characterize important biologic properties of an individual patient’s tumor and, in this way, direct therapy.

Article Information:
Disclosure

The authors have no conflicts of interest to declare. This work was supported in part by National Institutes of Health (NIH) grants RO1CA42045, RO1CA72064, RO1CA90771, and S10RR177229.

Correspondence

William B Eubank, MD, Department of Radiology (S-114-RAD), Puget Sound VA Health Care System, 1660 South Columbian Way, Seattle, WA 98108-1597. E: weubank@u.washington.edu

Received

2010-04-03T00:00:00

References

  1. Scheidhauer K, Walter C, Seemann MD, FDG PET and other imaging modalities in the primary diagnosis of suspicious breast lesions, Eur J Nucl Med Mol Imaging, 2004;31(Suppl. 1):S70–79.
  2. Elmore JG, Armstrong K, Lehman CD, et al., Screening for breast cancer, JAMA, 2005;293(10):1245–56.
  3. Lehman CD, Blume JD, Weatherall P, et al., Screening women at high risk for breast cancer with mammography and magnetic resonance imaging, Cancer, 2005;103(9):1898–905.
  4. Stavros AT, Thickman D, Rapp CL, et al., Solid breast nodules: use of sonography to distinguish between benign and malignant lesions, Radiology, 1995;196(1):123–34.
  5. Saslow D, Boetes C, Burke W, et al., American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography, CA Cancer J Clin, 2007;57(2):75–89.
  6. Avril N, Menzel M, Dose J, et al., Glucose metabolism of breast cancer assessed by 18F-FDG PET: histologic and immunohistochemical tissue analysis, J Nucl Med, 2001;42(1):9–16.
  7. Bos R, van Der Hoeven JJ, van Der Wall E, et al., Biologic correlates of [F-18]-fluorodeoxyglucose uptake in human breast cancer measured by positron emission tomography, J Clin Oncol, 2002;20(2):379–87.
  8. . Buck A, Schirrmeister H, Kuhn T, et al., FDG uptake in breast cancer: correlation with biological and clinical prognostic parameters, Eur J Nucl Med Mol Imaging, 2002;29(10):1317–23.
  9. Gil-Rendo A, Martinez-Regueira F, Zornoza G, et al., Association between [F-18]-fluorodeoxyglucose uptake and prognostic parameters in breast cancer, Br J Surg, 2009;96(2):166–70.
  10. Oshida M, Uno K, Suzuki M, et al., Predicting the prognoses of breast carcinoma patients with positron emission tomography using [F-18]-2-fluoro-2-deoxy-D-glucose, Cancer, 1998;1:2227–34.
  11. Ueda S, Tsuda H, Asakawa H, et al., Clinicopathological and prognostic relevance of uptake level using 18Ffluorodeoxyglucose positron emission tomography/ computed tomography fusion imaging (18F-FDG PET/CT) in primary breast cancer, Jpn J Clin Oncol, 2008;38(4):250–8.
  12. Avril N, Rose CA, Schelling M, et al., Breast imaging with positron emission tomography and [fluorine-18]- fluorodeoxyglucose: use and limitations, J Clin Oncol, 2000;18(20):3495–502.
  13. Kumar R, Chauhan A, Zhuang H, et al., Clinicopathologic factors associated with false negative FDG-PET in primary breast cancer, Breast Cancer Res Treat, 2006;98(3):267–74.
  14. Rosen EL, Eubank WB, Mankoff DA, FDG PET, PET/CT, and breast cancer imaging, Radiographics, 2007;27(Suppl. 1):S215–29.
  15. Berg WA, Weinberg IN, Narayanan D, et al., High-resolution 18Ffluorodeoxyglucose positron emission tomography with compression (“positron emission mammography”) is highly accurate in depicting primary breast cancer, Breast J, 2006;12(4):309–23.
  16. Rosen EL, Turkington TG, Soo MS, et al., Detection of primary breast carcinoma with a dedicated, large-field-of-view FDG PET mammography device: initial experience, Radiology, 2005;234(2):527–34.
  17. Raylman RR, Majewski S, Smith MF, et al., The positron emission mammography/tomography breast imaging and biopsy system (PEM/PET): design, construction and phantom-based measurements, Phys Med Biol, 2008;53(3):637–53.
  18. Singletary SE, Connolly JL, Breast cancer staging: working with the sixth edition of the AJCC Cancer Staging Manual, CA Cancer J Clin, 2006;56(1):37–47.
  19. . Gerber B, Seitz E, Muller H, et al., Perioperative screening for metastatic disease is not indicated in patients with primary breast cancer and no clinical signs of tumor spread, Breast Cancer Res Treat, 2003;82(1):29–37.
  20. Myers RE, Johnston M, Pritchard K, et al., Baseline staging tests in primary breast cancer: a practice guideline, CMAJ, 2001;164(10):1439–44.
  21. Puglisi F, Follador A, Minisini AM, et al., Baseline staging tests after a new diagnosis of breast cancer: further evidence of their limited indications, Ann Oncol, 2005;16(2):263–6.
  22. Ravaioli A, Pasini G, Polselli A, et al., Staging of breast cancer: new recommended standard procedure, Breast Cancer Res Treat, 2002;72(1):53–60.
  23. Schneider C, Fehr MK, Steiner RA, et al., Frequency and distribution pattern of distant metastases in breast cancer patients at the time of primary presentation, Arch Gynecol Obstet, 2003;269(1):9–12.
  24. Port ER, Yeung H, Gonen M, et al., [F-18]-2-fluoro-2-deoxy-Dglucose positron emission tomography scanning affects surgical management in selected patients with high-risk, operable breast carcinoma, Ann Surg Oncol, 2006;13(5):677–84.
  25. Pugliese M, Shivaram G, Rogers J, et al., PET-CT imaging in the initial management of high-risk breast cancer patients: who did it help? The Breast Journal, 2009;15(5):554–6.
  26. Fisher B, Slack NH, Number of lymph nodes examined and the prognosis of breast carcinoma, Surg Gynecol Obstet, 1970;131(1):79–88.
  27. Nemoto T, Vana J, Bedwani RN, et al., Management and survival of female breast cancer: results of a national survey by the American College of Surgeons, Cancer, 1980;45(12):2917–24.
  28. Newman EA, Newman LA, Lymphatic mapping techniques and sentinel lymph node biopsy in breast cancer, Surg Clin North Am, 2007;87(2):353–64, viii.
  29. Krag DN, Anderson SJ, Julian TB, et al., Technical outcomes of sentinel-lymph-node resection and conventional axillary-lymphnode dissection in patients with clinically node-negative breast cancer: results from the NSABP B-32 randomised phase III trial, Lancet Oncol, 2007;8(10):881–8.
  30. Liberman L, Pathologic analysis of sentinel lymph nodes in breast carcinoma, Cancer, 2000;88(5):971–7.
  31. Veronesi U, Paganelli G, Viale G, et al., A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer, N Engl J Med, 2003;349(6):546–53.
  32. Adler L, Faulhaber P, Schnur K, et al., Axillary lymph node metastases: screening with [F-18]-2-deoxy-2-fluoro-D-glucose (FDG) PET, Radiology, 1997;203:323–7.
  33. Crippa F, Agresti R, Donne VD, et al., The contribution of positron emission tomography (PET) with 18Ffluorodeoxyglucose (FDG) in the preoperative detection of axillary metastases of breast cancer: the experience of the National Cancer Institute of Milan, Tumori, 1997;83(2):542–3.
  34. Greco M, Crippa F, Agresti R, et al., Axillary lymph node staging in breast cancer by [F-18]-2-fluoro-2-deoxy-D-glucose-positron emission tomography: clinical evaluation and alternative management, J Natl Cancer Inst, 2001;93(8):630–5.
  35. Smith IC, Ogston KN, Whitford P, et al., Staging of the axilla in breast cancer: accurate in vivo assessment using positron emission tomography with [fluorine-18]-2-fluoro-2-deoxy-Dglucose, Ann Surg, 1998;228(2):220–7.
  36. Utech CI, Young CS, Winter PF, Prospective evaluation of [F-18]- fluorodeoxyclucose positron emission tomography in breast cancer for staging of the axilla related to surgery and immunocytochemistry, Eur J Nucl Med, 1996;23(12):1588–93.
  37. Peare R, Staff RT, Heys SD, The use of FDG-PET in assessing axillary lymph node status in breast cancer: a systematic review and meta-analysis of the literature, Breast Cancer Res Treat, 2010;123(1):281–90.
  38. Wahl RL, Siegel BA, Coleman RE, et al., Prospective multicenter study of axillary nodal staging by positron emission tomography in breast cancer: a report of the staging breast cancer with PET Study Group, J Clin Oncol, 2004;22(2):277–85.
  39. Chae BJ, Bae JS, Kang BJ, et al., Positron emission tomographycomputed tomography in the detection of axillary lymph node metastasis in patients with early stage breast cancer, Jpn J Clin Oncol, 2009;39(5):284–9.
  40. Fehr MK, Hornung R, Varga Z, et al., Axillary staging using positron emission tomography in breast cancer patients qualifying for sentinel lymph node biopsy, Breast J, 2004;10(2):89–93.
  41. Gil-Rendo A, Zornoza G, Garcia-Velloso MJ, et al., Fluorodeoxyglucose positron emission tomography with sentinel lymph node biopsy for evaluation of axillary involvement in breast cancer, Br J Surg, 2006;93(6):707–12.
  42. Guller U, Nitzsche EU, Schirp U, et al., Selective axillary surgery in breast cancer patients based on positron emission tomography with [F-18]-2-fluoro-2-deoxy-D-glucose: not yet! Breast Cancer Res Treat, 2002;71(2):171–3.
  43. Lovrics PJ, Chen V, Coates G, et al., A prospective evaluation of positron emission tomography scanning, sentinel lymph node biopsy, and standard axillary dissection for axillary staging in patients with early stage breast cancer, Ann Surg Oncol, 2004;11(9):846–53.
  44. Taira N, Ohsumi S, Takabatake D, et al., Determination of indication for sentinel lymph node biopsy in clinical nodenegative breast cancer using preoperative 18Ffluorodeoxyglucose positron emission tomography/ computed tomography fusion imaging, Jpn J Clin Oncol, 2009;39(1):16–21.
  45. Ueda S, Tsuda H, Asakawa H, et al., Utility of 18Ffluorodeoxyglucose emission tomography/computed tomography fusion imaging (18F-FDG PET/CT) in combination with ultrasonography for axillary staging in primary breast cancer, BMC Cancer, 2008;8:165.
  46. van der Hoeven JJ, Hoekstra OS, Comans EF, et al., Determinants of diagnostic performance of [F-18]-fluorodeoxyglucose positron emission tomography for axillary staging in breast cancer, Ann Surg, 2002;236(5):619–24.
  47. Veronesi U, De Cicco C, Galimberti VE, et al., A comparative study on the value of FDG-PET and sentinel node biopsy to identify occult axillary metastases, Ann Oncol, 2007;18(3):473–8.
  48. Zornoza G, Garcia-Velloso MJ, Sola J, et al., 18F-FDG PET complemented with sentinel lymph node biopsy in the detection of axillary involvement in breast cancer, Eur J Surg Oncol, 2004;30(1):15–9.
  49. Davis J, Brill Y, Simmons S, et al., Ultrasound-guided fine-needle aspiration of clinically negative lymph nodes versus sentinel node mapping in patients at high risk for axillary metastasis, Ann Surg Oncol, 2006;13(12):1545–52.
  50. de Kanter AY, van Eijck CH, van Geel AN, et al., Multicentre study of ultrasonographically guided axillary node biopsy in patients with breast cancer, Br J Surg, 1999;86(11):1459–62.
  51. Hinson JL, McGrath P, Moore A, et al., The critical role of axillary ultrasound and aspiration biopsy in the management of breast cancer patients with clinically negative axilla, Ann Surg Oncol, 2008;15(1):250–5.
  52. Moore A, Hester M, Nam MW, et al., Distinct lymph nodal sonographic characteristics in breast cancer patients at high risk for axillary metastases correlate with the final axillary stage, Br J Radiol, 2008;81(968):630–6.
  53. Eubank WB, Mankoff DA, Current and future uses of positron emission tomography in breast cancer imaging, Semin Nucl Med, 2004;34(3):224–40.
  54. Kim J, Lee J, Chang E, et al., Selective sentinel node plus additional non-sentinel node biopsy based on an fdg-pet/ct scan in early breast cancer patients: single institutional experience, World J Surg, 2009;33(5):943–9.
  55. Kumar R, Zhuang H, Schnall M, et al., FDG PET positive lymph nodes are highly predictive of metastasis in breast cancer, Nucl Med Commun, 2006;27(3):231–6.
  56. Veronesi P, Rodriguez-Fernandez J, Intra M, Controversies in the use of sentinel nodes: microinvasion, post surgery and after preoperative systemic treatment, Breast, 2007;16 (Suppl. 2):S67–70.
  57. Brito RA, Valero V, Buzdar AU, et al., Long-term results of combined-modality therapy for locally advanced breast cancer with ipsilateral supraclavicular metastases: the University of Texas M.D. Anderson Cancer Center experience, J Clin Oncol, 2001;19(3):628–33.
  58. . Kuru B, Camlibel M, Dinc S, et al., Prognostic significance of axillary node and infraclavicular lymph node status after mastectomy, Eur J Surg Oncol, 2003;29(10):839–44.
  59. Bellon JR, Livingston RB, Eubank WB, et al., Evaluation of the internal mammary lymph nodes by FDG-PET in locally advanced breast cancer (LABC), Am J Clin Oncol, 2004;27(4):407–10.
  60. Carkaci S, Macapinlac HA, Cristofanilli M, et al., Retrospective study of 18F-FDG PET/CT in the diagnosis of inflammatory breast cancer: preliminary data, J Nucl Med, 2009;50(2):231–8.
  61. Fuster D, Duch J, Paredes P, et al., Preoperative staging of large primary breast cancer with [F-18]-fluorodeoxyglucose positron emission tomography/computed tomography compared with conventional imaging procedures, J Clin Oncol, 2008;26(29):4746–51.
  62. Groheux D, Moretti JL, Baillet G, et al., Effect of 18F-FDG PET/CT imaging in patients with clinical Stage II and III breast cancer, Int J Radiat Oncol Biol Phys, 2008;71(3):695–704.
  63. Mahner S, Schirrmacher S, Brenner W, et al., Comparison between positron emission tomography using [fluorine-18]-2- fluoro-2-deoxy-D-glucose, conventional imaging and computed tomography for staging of breast cancer, Ann Oncol, 2008;19(7):1249–54.
  64. Baslaim MM, Bakheet SM, Bakheet R, et al., 18Ffluorodeoxyglucose- positron emission tomography in inflammatory breast cancer, World J Surg, 2003;27(10): 1099–104.
  65. Yang WT, Le-Petross HT, Macapinlac H, et al., Inflammatory breast cancer: PET/CT, MRI, mammography, and sonography findings, Breast Cancer Res Treat, 2008;109(3):417–26.
  66. Eubank WB, Mankoff DA, Takasugi J, et al., 18Ffluorodeoxyglucose positron emission tomography to detect mediastinal or internal mammary metastases in breast cancer, J Clin Oncol, 2001;19(15):3516–23.
  67. Chia S, Swain SM, Byrd DR, et al., Locally advanced and inflammatory breast cancer, J Clin Oncol, 2008;26(5):786–90.
  68. van der Hoeven JJ, Krak NC, Hoekstra OS, et al., [F-18]-2-fluoro-2- deoxy-d-glucose positron emission tomography in staging of locally advanced breast cancer, J Clin Oncol, 2004;22(7):1253–9.
  69. Bender H, Kirst J, Palmedo H, et al., Value of 18Ffluorodeoxyglucose positron emission tomography in the staging of recurrent breast carcinoma, Anticancer Res, 1997;17(3B):1687–92.
  70. Gallowitsch HJ, Kresnik E, Gasser J, et al., [F-18]- fluorodeoxyglucose positron-emission tomography in the diagnosis of tumor recurrence and metastases in the follow-up of patients with breast carcinoma: a comparison to conventional imaging, Invest Radiol, 2003;38(5):250–6.
  71. Kamel EM, Wyss MT, Fehr MK, et al., [F-18]-fluorodeoxyglucose positron emission tomography in patients with suspected recurrence of breast cancer, J Cancer Res Clin Oncol, 2003;129(3):147–53.
  72. Kim TS, Moon WK, Lee DS, et al., 18F-fluorodeoxyglucose positron emission tomography for detection of recurrent or metastatic breast cancer, World J Surg, 2001;25(7):829–34.
  73. Lin WY, Tsai SC, Cheng KY, et al., [Fluorine-18]-FDG-PET in detecting local recurrence and distant metastases in breast cancer—Taiwanese experiences, Cancer Invest, 2002;20(5–6):725–9.
  74. Liu CS, Shen YY, Lin CC, et al., Clinical impact of 18F-FDG-PET in patients with suspected recurrent breast cancer based on asymptomatically elevated tumor marker serum levels: a preliminary report, Jpn J Clin Oncol, 2002;32(7):244–7.
  75. Lonneux M, Borbath II, Berliere M, et al., The place of wholebody FDG PET for the diagnosis of distant recurrence of breast cancer, Clin Positron Imaging, 2000;3(2):45–9.
  76. Moon DH, Maddahi J, Silverman DH, et al., Accuracy of wholebody [fluorine-18]-FDG PET for the detection of recurrent or metastatic breast carcinoma, J Nucl Med, 1998;39(3):431–5.
  77. . Siggelkow W, Zimny M, Faridi A, et al., The value of positron emission tomography in the follow-up for breast cancer, Anticancer Res, 2003;23(2C):1859–67.
  78. Suarez M, Perez-Castejon MJ, Jimenez A, et al., Early diagnosis of recurrent breast cancer with FDG-PET in patients with progressive elevation of serum tumor markers, Q J Nucl Med, 2002;46(2):113–21.
  79. Vranjesevic D, Filmont JE, Meta J, et al., Whole-body 18F-FDG PET and conventional imaging for predicting outcome in previously treated breast cancer patients, J Nucl Med, 2002;43(3):325–9.
  80. Weir L, Worsley D, Bernstein V, The value of FDG positron emission tomography in the management of patients with breast cancer, Breast J, 2005;11(3):204–9.
  81. Wolfort RM, Li BD, Johnson LW, et al., The role of whole-body [fluorine-18]-FDG positron emission tomography in the detection of recurrence in symptomatic patients with stages II and III breast cancer, World J Surg, 2006;30(8):1422–7.
  82. Isasi CR, Moadel RM, Blaufox MD, A meta-analysis of FDG-PET for the evaluation of breast cancer recurrence and metastases, Breast Cancer Res Treat, 2005;90(2):105–12.
  83. Coleman RE, Rubens RD, The clinical course of bone metastases from breast cancer, Br J Cancer, 1987;55(1):61–6.
  84. Cook GJ, Houston S, Rubens R, et al., Detection of bone metastases in breast cancer by 18F-FDG PET: differing metabolic activity in osteoblastic and osteolytic lesions, J Clin Oncol, 1998;16(10):3375–9.
  85. Abe K, Sasaki M, Kuwabara Y, et al., Comparison of 18F-FDG-PET with 99mTc-HMDP scintigraphy for the detection of bone metastases in patients with breast cancer, Ann Nucl Med, 2005;19(7):573–9.
  86. Nakai T, Okuyama C, Kubota T, et al., Pitfalls of FDG-PET for the diagnosis of osteoblastic bone metastases in patients with breast cancer, Eur J Nucl Med Mol Imaging, 2005;32(11):1253–8.
  87. Ohta M, Tokuda Y, Suzuki Y, et al., Whole body PET for the evaluation of bony metastases in patients with breast cancer: comparison with 99Tcm-MDP bone scintigraphy, Nucl Med Commun, 2001;22(8):875–9.
  88. Uematsu T, Yuen S, Yukisawa S, et al., Comparison of FDG PET and SPECT for detection of bone metastases in breast cancer, AJR Am J Roentgenol, 2005;184(4):1266–73.
  89. Yang SN, Liang JA, Lin FJ, et al., Comparing whole body [F-18]-2-fluorodeoxyglucose positron emission tomography and technetium-99m methylene diphosphonate bone scan to detect bone metastases in patients with breast cancer, J Cancer Res Clin Oncol, 2002;128(6):325–8.
  90. Even-Sapir E, Metser U, Flusser G, et al., Assessment of malignant skeletal disease: initial experience with 18F-fluoride PET/CT and comparison between 18F-fluoride PET and 18Ffluoride PET/CT, J Nucl Med, 2004;45(2):272–8.
  91. Petren-Mallmin M, Andreasson I, Ljunggren O, et al., Skeletal metastases from breast cancer: uptake of 18F-fluoride measured with positron emission tomography in correlation with CT, Skeletal Radiol, 1998;27(2):72–6.
  92. Schirrmeister H, Guhlmann A, Kotzerke J, et al., Early detection and accurate description of extent of metastatic bone disease in breast cancer with fluoride ion and positron emission tomography, J Clin Oncol, 1999;17(8):2381–9.
  93. Antoch G, Saoudi N, Kuehl H, et al., Accuracy of whole-body dual-modality [fluorine-18]-2-fluoro-2-deoxy-D-glucose positron emission tomography and computed tomography (FDG-PET/CT) for tumor staging in solid tumors: comparison with CT and PET, J Clin Oncol, 2004;22(21):4357–68.
  94. . Tatsumi M CC, Mortzikos KA, Fishman EK, Wahl RL, Initial experience with FDG-PET/CT in the evaluation of breast cancer, Eur J Nucl Med Mol Imaging, 2006;33(3):254–62.
  95. Grahek D, Montravers F, Kerrou K, et al., 18F-FDG in recurrent breast cancer: diagnostic performances, clinical impact and relevance of induced changes in management, Eur J Nucl Med Mol Imaging, 2004;31(2):179–88.
  96. Fueger B, Weber WA, Quon A, et al., Performance of [F-18]-2- fluoro-2-deoxy-d-glucose positron emission tomography and integrated PET/CT in restaged breast cancer patients, Mol Imaging Biol, 2005;7:369–76.
  97. Radan L B-HS, Bar-Shalom R, et al, The role of FDG-PET/CT in suspected recurrence of breast cancer, Cancer, 2006;107:2545–51.
  98. Piperkova E, Raphael B, Altinyay ME, et al., Impact of PET/CT in comparison with same day contrast enhanced CT in breast cancer management, Clin Nucl Med, 2007;32(6):429–34.
  99. Veit-Haibach P, Antoch G, Beyer T, et al., FDG-PET/CT in restaging of patients with recurrent breast cancer: possible impact on staging and therapy, Br J Radiol, 2007;80(955):508–15.
  100. Haug AR, Schmidt GP, Klingenstein A, et al., [F-18]-2-fluoro-2- deoxyglucose positron emission tomography/computed tomography in the follow-up of breast cancer with elevated levels of tumor markers, J Comput Assist Tomogr, 2007;31(4):629–34.
  101. Dirisamer A, Halpern BS, Flory D, et al., Integrated contrastenhanced diagnostic whole-body PET/CT as a first-line restaging modality in patients with suspected metastatic recurrence of breast cancer, Eur J Radiol, 2010;73(2):294–9.
  102. Gralow JR, Optimizing the treatment of metastatic breast cancer, Breast Cancer Res Treat, 2005;89(Suppl. 1):S9–15.
  103. Gralow JR, Burstein HJ, Wood W, et al., Preoperative therapy in invasive breast cancer: pathologic assessment and systemic therapy issues in operable disease, J Clin Oncol, 2008;26(5):814–19.
  104. Feldman LD, Hortobagyi GN, Buzdar AU, et al., Pathological assessment of response to induction chemotherapy in breast cancer, Cancer Res, 1986;46:2578–81.
  105. McCready DR, Hortobagyi GN, Kau SW, et al., The prognostic significance of lymph node metastases after preoperative chemotherapy for locally advanced breast cancer, Arch Surg, 1989;124:21–5.
  106. Wolmark N, Wang J, Mamounas E, et al., 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, 2001(30):96–102.
  107. Moscovic EC, Mansi JL, King DM, et al., Mammography in the assessment of response to medical treatment of large primary breast tumor, Clinical Radiology, 1993;47:339–44.
  108. Lee JH, Rosen EL, Mankoff DA, The role of radiotracer imaging in the diagnosis and management of patients with breast cancer: part 2—response to therapy, other indications, and future directions, J Nucl Med, 2009;50(5):738–48.
  109. Bassa P, Kim EE, Inoue T, et al., Evaluation of preoperative chemotherapy using PET with [fluorine-18]-fluorodeoxyglucose in breast cancer, J Nucl Med, 1996;37:931–8.
  110. Dunnwald LK, Gralow JR, Ellis GK, et al., Tumor metabolism and blood flow changes by positron emission tomography: relation to survival in patients treated with neoadjuvant chemotherapy for locally advanced breast cancer, J Clin Oncol, 2008;26(27):4449–57.
  111. Mankoff DA, Dunnwald LK, Gralow JR, et al., Changes in blood flow and metabolism in locally advanced breast cancer treated with neoadjuvant chemotherapy, J Nucl Med, 2003;44(11):1806–14.
  112. McDermott GM, Welch A, Staff RT, et al., Monitoring primary breast cancer throughout chemotherapy using FDG-PET, Breast Cancer Res Treat, 2007;102(1):75–84.
  113. Rousseau C, Devillers A, Sagan C, et al., Monitoring of early response to neoadjuvant chemotherapy in stage II and III breast cancer by 18F-fluorodeoxyglucose positron emission tomography, J Clin Oncol, 2006;24(34):5366–72.
  114. Schelling M, Avril N, Nahrig J, et al., Positron emission tomography using 18F-fluorodeoxyglucose for monitoring primary chemotherapy in breast cancer, J Clin Oncol, 2000;18:1689–95.
  115. Smith I, Welch A, Hutcheon A, et al., Positron emission tomography using 18F-fluorodeoxy-D-glucose to predict the pathologic response of breast cancer to primary chemotherapy, J Clin Oncol, 2000;18:1676–88.
  116. Wahl RL, Zasadny K, Helvie M, et al., Metabolic monitoring of breast cancer chemohormonotherapy using positron emission tomography: initial evaluation, J Clin Oncol, 1993;11:2101–11.
  117. Berriolo-Riedinger A, Touzery C, Riedinger JM, et al., 18F-FDGPET predicts complete pathological response of breast cancer to neoadjuvant chemotherapy, Eur J Nucl Med Mol Imaging, 2007;34(12):1915–24.
  118. Duch J, Fuster D, Munoz M, et al., 18F-FDG PET/CT for early prediction of response to neoadjuvant chemotherapy in breast cancer, Eur J Nucl Med Mol Imaging, 2009;36(10):1551–7.
  119. Schwarz-Dose J, Untch M, Tiling R, et al., Monitoring primary systemic therapy of large and locally advanced breast cancer by using sequential positron emission tomography imaging with 18F-fluorodeoxyglucose, J Clin Oncol, 2009;27(4):535–41.
  120. Burcombe RJ, Makris A, Pittam M, et al., Evaluation of good clinical response to neoadjuvant chemotherapy in primary breast cancer using 18F-fluorodeoxyglucose positron emission tomography, Eur J Cancer, 2002;38(3):375–9.
  121. Dose-Schwarz J, Tiling R, Avril-Sassen S, et al., Assessment of residual tumour by FDG-PET: conventional imaging and clinical examination following primary chemotherapy of large and locally advanced breast cancer, Br J Cancer, 2010;102(1):35–41.
  122. Kim SJ, Kim SK, Lee ES, et al., Predictive value of 18F-FDG PET for pathological response of breast cancer to neo-adjuvant chemotherapy, Ann Oncol, 2004;15(9):1352–7.
  123. Chen X, Moore MO, Lehman CD, et al., Combined use of MRI and PET to monitor response and assess residual disease for locally advanced breast cancer treated with neoadjuvant chemotherapy, Acad Radiol, 2004;11(10):1115–24.
  124. Emmering J, Krak NC, Van der Hoeven JJ, et al., Preoperative 18F-FDG-PET after chemotherapy in locally advanced breast cancer: prognostic value as compared with histopathology, Ann Oncol, 2008;19:1573–7.
  125. Therasse P, Arbuck SG, Eisenhauer EA, et al., New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada, J Natl Cancer Inst, 2000;92(3):205–16.
  126. Therasse P, Eisenhauer EA, Verweij J, RECIST revisited: a review of validation studies on tumour assessment, Eur J Cancer, 2006;42(8):1031–9.
  127. Couturier O, Jerusalem G, N’Guyen JM, et al., Sequential positron emission tomography using [F-18]-fluorodeoxyglucose for monitoring response to chemotherapy in metastatic breast cancer, Clin Cancer Res, 2006;12(21):6437–43.
  128. Dose-Schwarz J, Bader M, Jenicke L, et al., Early prediction of response to chemotherapy in metastatic breast cancer using sequential 18F-FDG PET, J Nucl Med, 2005;46(7):1144–50.
  129. Gennari A, Donati S, Salvadori B, et al., Role of 18Ffluorodeoxyglucose (FDG) positron emission tomography (PET) in the early assessment of response to chemotherapy in metastatic breast cancer patients, Clin Breast Cancer, 2000;1(2):156–61, discussion 62–3.
  130. Cachin F, Prince HM, Hogg A, et al., Powerful prognostic stratification by [F-18]-fluorodeoxyglucose positron emission tomography in patients with metastatic breast cancer treated with high-dose chemotherapy, J Clin Oncol, 2006;24(19):3026–31.
  131. Hamaoka T, Madewell JE, Podoloff DA, et al., Bone imaging in metastatic breast cancer, J Clin Oncol, 2004;22(14):2942–53.
  132. Coleman RE, Mashiter G, Whitaker KB, et al., Bone scan flare predicts successful systemic therapy for bone metastases, J Nucl Med, 1988;29(8):1354–9.
  133. Schneider JA, Divgi CR, Scott AM, et al., Flare on bone scintigraphy following Taxol chemotherapy for metastatic breast cancer, J Nucl Med, 1994;35(11):1748–52.
  134. Stafford SE, Gralow JR, Schubert EK, et al., Use of serial FDG PET to measure the response of bone-dominant breast cancer to therapy, Acad Radiol, 2002;9(8):913–21.
  135. Specht JM, Tam SL, Kurland BF, et al., Serial [F-18]-2-fluoro-2- deoxy-D-glucose positron emission tomography (FDG-PET) to monitor treatment of bone-dominant metastatic breast cancer predicts time to progression (TTP), Breast Cancer Res Treat, 2007;105(1):87–94.
  136. Tateishi U, Gamez C, Dawood S, et al., Bone metastases in patients with metastatic breast cancer: morphologic and metabolic monitoring of response to systemic therapy with integrated PET/CT, Radiology, 2008;247(1):189–96.
  137. . Mankoff DA, Lee JH, Eubank WB, Breast cancer imaging with novel PET tracers, PET Clin, 2009;4(4):371–80.
  138. Katzenellenbogen JA, Welch MJ, Dehdashti F, The development of estrogen and progestin radiopharmaceuticals for imaging breast cancer, Anticancer Res, 1997;17:1573–6.
  139. Dehdashti F, Mortimer JE, Siegel BA, et al., Positron tomographic assessment of estrogen receptors in breast cancer: comparison with FDG-PET and in vitro receptor assays, J Nucl Med, 1995;36(10):1766–74.
  140. Linden HM, Stekhova SA, Link JM, et al., Quantitative fluoroestradiol positron emission tomography imaging predicts response to endocrine treatment in breast cancer, J Clin Oncol, 2006;24(18):2793–9.
  141. Mortimer JE, Dehdashti F, Siegel BA, et al., Metabolic flare: indicator of hormone responsiveness in advanced breast cancer, J Clin Oncol, 2001;19(11):2797–803.
  142. Dijkers EC, Kosterink JG, Rademaker AP, et al., Development and characterization of clinical-grade 89Zr-trastuzumab for HER2/neu immunoPET imaging, J Nucl Med, 2009;50(6):974–81.

Further Resources

Share this Article
Related Content In Diagnostics and Screening
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72