Sei sulla pagina 1di 93

xi

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) xi

Preface
Breast Cancer

Norbert Avril, MD
Department of Nuclear Medicine
Barts and The London School of Medicine
Queen Mary, University of London
St. Bartholomew’s Hospital, West Smithfield (QE II)
London EC1A 7BE, UK

E-mail address:
n.e.avril@qmul.ac.uk
Norbert Avril, MD
Guest Editor

Breast cancer is one of the most devastating diseases The development of integrated PET and CT scan-
affecting women in Western countries. Although ners (PET/CT) was a milestone in the acceptance of
progress has been made in the development of new metabolic PET imaging in the clinical workup of
therapeutic agents, approximately 40,000 patients in cancer patient. The coregistered metabolic and ana-
the United States alone will die of breast cancer tomic information from PET/CT is resulting in
in 2005. The diagnosis of breast cancer—especially in improved sensitivity and specificity compared with
the early stages—is challenging, and mammography the single imaging procedures. FDG-PET and FDG-
and ultrasound play an important role in screening PET/CT have become an integral part of routine
asymptomatic women as well as in the initial workup patient care in breast cancer within the past few
of suspicious breast lesions. years. New applications such as radiation therapy
Positron emission tomography (PET) has become planning based on metabolic/anatomic informa-
widely available in the United States, and this first tion are on the horizon but still need careful evalua-
issue of the PET Clinics is devoted to the current tion and validation. PET tracers other than FDG
status and potential future applications of molecular targeting tumor cell proliferation and angiogenesis
PET imaging in breast cancer. Higher glucose con- as well as radiolabeled receptor ligands are now in-
sumption in cancer cells compared with normal creasingly being used to noninvasively characterize
tissue has made F-18 fluorodeoxyglucose (FDG) breast cancer.
the most commonly used PET radiopharmaceu- There are further potential applications of FDG-
tical. The metabolic information from FDG-PET pro- PET beyond diagnostic staging. Changes in tumor
vides sensitive and specific information about the glucose consumption occur early in the course of
spread of disease but is limited in evaluating pri- chemotherapy and ultimately predict treatment
mary breast cancer. New and improved dedicated outcome. The use of FDG-PET for monitoring
PET breast scanners have both higher sensitivity breast cancer therapy holds promise to reduce the
and spatial resolution. This necessitates a recon- number of ineffective therapies and unnecessary
sideration of the potential clinical application of side effects and to facilitate the effective evaluation
FDG-PET in primary breast cancer. of new therapeutic approaches.

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.10.001
pet.theclinics.com
1

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 1–13

F-18 Fluorodeoxyglucose-Positron
Emission Tomography Imaging
for Primary Breast Cancer and
Loco-Regional Staging
a,* b
Norbert Avril, MD , Lee P. Adler, MD

& Detection of primary breast tumors & Loco-regional staging


& Positron emission tomography imaging of & Positron emission mammography
breast cancer & Current clinical status of F-18
Positron emission tomography imaging fluorodeoxyglucose-positron emission
procedure and image analysis in the tomography in primary breast cancer
breast & References
Positron emission tomography imaging of
the breast
Biological features of breast cancer

Breast cancer is the most common female ma- distant metastases. Most patients who have locally
lignancy in most European countries, North Amer- advanced disease have axillary lymph nodes in-
ica, and Australia; it is less frequent in Asia and in volved with their tumors, but a subset of patients
Africa. In Europe, one out of every 10 to 15 women have large primary tumors without lymph node
will develop breast cancer in her lifetime, and the involvement. For patients who have lymph node
risk is even higher in the United States, where it is metastases, more than four lymph nodes involved
one out of every eight women. Approximately 95% predict poorer survival [2]. The College of Ameri-
of breast cancer cases occur sporadically without can Pathologists has recently considered prognostic
any known genetic mutation, and the causal and predictive factors in breast cancer and stratified
mechanisms underlying this disease have yet to them into categories reflecting the strength of pub-
be fully elucidated. Overall, 5-year survival rates lished evidence [3]. Factors ranked in Category I
are approximately 75%, with ranges of 92% for included tumor, node, metastases (TNM) staging;
Stage I (pT1, pN0, M0) to 15% for Stage IV (M1) histologic grade; histologic type; mitotic figure
disease [1]. counts; and hormone receptor status. Category II
The main prognostic factors in patients who have factors included c-erbB-2 (Her2-neu), proliferation
breast cancer are lymph node status, tumor size, markers, lymphatic and vascular channel invasion,
histologic grade, and the presence or absence of and p53. Factors in Category III included DNA

a
Department of Nuclear Medicine, Barts and the London School of Medicine, Queen Mary, University of
London, West Smithfield (QEII), London, EC1A 7BE, UK
b
Adler Institute for Advanced Imaging, The Pavilion, 261 Old York Road, Jenkintown, PA 19046, USA
* Corresponding author.
E-mail address: AvrilNE@upmc.edu (N. Avril).

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.008
pet.theclinics.com
2 Avril & Adler

ploidy analysis, microvessel density, epidermal mass, either by self-examination or routine breast
growth factor receptor, transforming growth factor- examination. Clinical signs include a fixed, hard
alpha, bcl-2, pS2, and cathepsin D. mass; asymmetry of the breast contour; a pro-
Invasive ductal carcinoma is the most common trusion; a subtle dimpling of the skin (peau d’
histological type (70%–80%), followed by invasive orange); or a bloody nipple discharge. Depending
lobular carcinoma (6%–10%) and medullary car- on the size of the breast and the density of breast
cinoma (~3%). The remaining tumors include a tissue, most tumors are not palpable smaller than
variety of histological types. Invasive breast cancer 1 cm in diameter. Breast carcinomas are often pres-
may be present as a single tumor, or as multifocal ent as irregularly shaped, firm or hard, yet painless
if tumors are growing in the same quadrant of nodules or masses. Physical examination typically
the breast, and as multicentric if they are detected does not allow an accurate differentiation between
in different quadrants. The disease can occur in a malignant and a nonmalignant mass. Therefore,
any part of the breast, but most frequently occurs imaging modalities are used to improve the diag-
in the upper outer quadrant. Locally advanced nostic accuracy, and various new and innovative
breast cancer remains a particular challenge, be- technologies are being investigated for advancing
cause the majority of patients who have this the early detection and diagnosis of breast cancer.
diagnosis develop distant metastases despite appro- Screening-mammography allows the detection of
priate therapy. Patients who have locally advanced breast cancer earlier than breast self-examination,
disease include primary tumors with direct exten- and is generally credited with earlier diagnosis and
sion to chest wall or skin (stage T4); advanced an overall improvement in survival for patients
nodal disease, such as fixed axillary nodes or who have newly diagnosed breast cancer. Mam-
involvement of ipsilateral supraclavicular, infracla- mography localizes and assesses the extent of a le-
vicular, or internal mammary nodes; and inflam- sion as well as identifying other suspicious masses.
matory carcinomas. Studies in large series have shown that mammog-
Noninvasive breast cancer consists of two histo- raphy, using mediolateral oblique and craniocau-
logical and clinical subtypes: ductal and lobular dal projections, is a useful tool to improve early
in-situ carcinomas. The carcinoma cells are con- detection of breast cancer. Depending on the lesion
fined within the terminal duct lobular unit and size and the radiographic appearance and breast
the adjacent ducts, but have not yet invaded tissue density, sensitivity ranges from 54% to 58%
through the basement membrane. Generally, lobu- in women under age 40, and from 81% to 94% in
lar carcinoma in situ (LCIS) does not present as those over 50 [4,5]. Malignant and benign breast
a palpable tumor and is usually found incidentally lesions often display similar radiographic appear-
in breast biopsies, often multifocal and bilateral. ance, resulting in a major limitation of mammog-
Ductal carcinoma in situ (DCIS) is increasingly raphy [6]. A Breast Imaging Reporting and Data
diagnosed because of microcalcifications seen on System (BI-RADS) has recently been introduced to
mammograms, and is more likely to be confined to characterize mammography [7]. The categories are
one breast or even to one quadrant of the breast. listed and explained in Box 1.
If breast cancer is suspected, a biopsy is necessary In some studies, approximately 6 to 8 out of
to confirm the diagnosis. The National Compre- 10 patients who have suspicious lesions in mam-
hensive Cancer Network has published guidelines mography and who undergo surgery have benign
for the work-up of women who have newly diag- histology [6,8]. A recent multicenter analysis of
nosed breast cancer. The recommendations include 332,926 diagnostic mammography examinations
history and physical examination, diagnostic bilat- [9] found the positive predictive value of a biopsy
eral mammogram and ultrasound and optional recommendation to be 31.5%, and that of a biopsy
breast MRI, pathology review, and determination
of estrogen receptor, progesterone receptor, nuclear
grade and HER-2/neu status. To evaluate distant Box 1: BI-RADS categories and definitions
metastases, chest radiograph, ultrasound of the 0: More information is needed to give a final
abdomen, bone scintigraphy, and CT or MRI may mammogram report.
be indicated. 1: Mammogram is normal.
2: Mammogram shows benign finding.
3: Probably benign finding—short interval
Detection of primary breast tumors follow-up suggested.
4: Suspicious abnormality—biopsy should
Improved methods to detect and diagnose breast
be considered.
cancer early are required to achieve a significant 5: Highly suggestive of malignancy—
impact on morbidity and mortality. More than appropriate action should be taken.
80% of cancers are detected because of a suspicious
FDG-PET Imaging for Breast Cancer 3

performed to be 39.5% This means that currently tion of dense breast parenchyma that often limits
almost two thirds have a negative biopsy. For the detection of breast cancer in mammography.
screening mammography, the positive predictive The use of paramagnetic contrast agents has
value is even lower. About 10% of breast carci- been found to be essential in characterizing breast
nomas are not identified in mammography, even masses. Signal enhancement following injection of
when they are palpable [10]. One reason is that intravenous (IV) contrast is a highly sensitive cri-
mammography is limited, because cancer can have terion to detect breast cancer, and sensitivity is
photon absorption similar to that of normal breast more than 90% in most studies. Unfortunately,
tissue, especially in younger women who have radio- the high sensitivity of MRI for invasive breast car-
graphically dense breasts. Despite these limitations, cinomas is coupled with a correspondingly low
mammography is viewed as the best tool currently specificity [14,15]. To improve upon the disap-
available for screening and early diagnosis. pointingly low positive predictive value of enhanc-
Ultrasound has become an important imaging ing lesions on MRI, high-speed dynamic imaging
modality in evaluating the breast [11]. Ultrasound of the breast must be performed in make use of the
is often used in addition to mammography, and differential washout rates of contrast agent gado-
provides differentiation between cystic lesions and linium (III)-diethyltriaminepentaacetic acid (Gd-
solid tumors. In younger patients who have dense DTPA) in benign and malignant breast lesions.
breasts, ultrasound can be superior in the detection Even with dynamic imaging, MRI has not proven
of breast cancer in comparison with mammog- to be a robust technique for discriminating benign
raphy. Breast cancer typically shows irregularly from malignant tumors in the community setting.
shaped hypoechoic masses, posterior acoustical MRI of the breast offers higher sensitivity for the
shadowing, and ill-defined demarcation against detection of multifocal or multicentric cancer,
the surrounding tissue. Doppler ultrasound may which is important in selecting patients appropriate
help distinguish benign from malignant breast dis- for breast-conserving surgery. It is also a valuable
ease; however, the diagnostic accuracy is often not tool for the screening of patients who have a high
sufficient enough to accurately characterize abnor- risk of breast cancer, or in whom there is axillary
mal tissue, and to specifically exclude malignancy disease or nipple discharge and conventional imag-
[12]. Another common application of ultrasound is ing has not revealed the primary focus. Lesions
to provide guidance for interventional procedures detected on MRI are frequently visible on mammog-
[12,13]. Less common uses include assisting in raphy or ultrasound in retrospect, if not prospec-
staging of breast cancer and evaluating patients tively, allowing for subsequent biopsy. Techniques
who have implants. Recently, there has been an are also now available to biopsy lesions only appar-
interest in using ultrasound to screen asymptomatic ent on MRI of the breast. MRI can differentiate scar
women for breast cancer, as is done with mam- tissue from tumor, and is specifically useful in pa-
mography. Further studies must be performed to tients suspected for local recurrent disease. Studies
assess if this reduces mortality from breast cancer. suggest that MRI can identify responders and non-
Although primarily used to image the female responders to neoadjuvant chemotherapy with
breast, ultrasound also can be used to evaluate more accuracy [16]. It is the modality of choice
breast-related concerns in men. Uses of contrast- for the assessment of breast implants for rupture,
enhanced ultrasound are still experimental and with accuracy higher than radiograph mammogra-
would add an invasive component to an otherwise phy and ultrasound. The most important limita-
noninvasive study. tions of MRI beside the low specificity are patient
MRI has become a valuable tool in breast disease, compliance, scan time, and cost.
especially in cases that are difficult to diagnose.
Recent progress in both spatial and temporal reso-
Positron emission tomography imaging of
lutions, the imaging sequences used, pharmacoki-
breast cancer
netic modeling of contrast uptake, and the use
of dedicated breast coils has contributed to the Positron emission tomography (PET) is a noninva-
advancement of this imaging technique. More re- sive imaging technique that measures the concen-
cently, phased-array breast coils, pulse sequences tration of positron-emitting radiopharmaceuticals
engineered to saturate signal from fat-containing within the body. Depending upon the radiolabeled
tissue (FATSAT), and gadolinium-based contrast tracer used, PET can be used to determine various
agents have done so as well [14]. MRI has several physiological and biochemical processes in vivo.
distinct advantages for breast imaging. These in- PET is highly sensitive, with the capacity to detect
clude three-dimensional visualization of breast tis- picomolar concentrations of radiotracer, and pro-
sue, information about tissue vascularity, and chest vides superior image resolution compared with con-
wall visualization. Moreover, MRI allows evalua- ventional nuclear medicine imaging with gamma
4 Avril & Adler

cameras. Currently, PET imaging can target several tracer retention in the axilla region, the tracer
biological features of cancer, including glucose should be injected into an arm vein contralateral
metabolism, cell proliferation, perfusion, and hyp- to the suspected tumor. Most of the studies re-
oxia. Approximately 95% of clinical PET examina- ported in literature are done in two-dimensional
tions are performed in patients who have known or mode data acquisition, and the influence of three-
suspected cancer, and virtually all of these are per- dimensional mode on the results of breast imaging
formed with a single radiotracer, the radiolabeled still needs to be studied. Imaging in prone position
glucose analog, [F-18] 2-deoxy-2-fluoro-D-glucose with both arms at the side and the breast hanging
(FDG). Following malignant transformation, vari- free is recommended to avoid compression and
ous tumors are characterized by elevated glucose deformation of the breast. Data acquisition should
consumption and subsequent increased uptake and be started approximately 60 minutes after tracer
accumulation of FDG. PET imaging using FDG injection. Boerner and colleagues [21] showed in-
provides more sensitive and more specific infor- creasing target-to-background ratios over time, sug-
mation about the extent of disease than morpho- gesting a benefit to longer waiting periods between
logical/anatomical imaging alone. FDG-PET has tracer injection and data acquisition. Lower image
become a standard imaging procedure for staging quality, due to radionuclide decay, has to be taken
and restaging of many types of cancer [17]. In the into account, however. Attenuation correction is
United States, PET scans performed on patients recommended for optimal tumor localization as
who have head and neck cancer, follicular thyroid well as subsequent quantification of regional tracer
cancer, solitary pulmonary nodules, lung cancer, uptake. The use of iterative reconstruction algo-
breast cancer, esophageal cancer, colorectal cancer, rithms results in better image quality; an increase
lymphoma, melanoma, and cervical cancer are in diagnostic accuracy has not yet been reported.
generally considered reimbursable by third-party Visual image interpretation should include analysis
payers. The metabolic activity of neoplastic tissue of transaxial, coronal, and sagittal views. Breast can-
assessed by PET offers additional information about cer is typically present with focally increased FDG
cancer biology, and can be used for the differentia- uptake, whereas benign tumors are negative in PET
tion between benign and malignant lesions, identi- imaging. Proliferative mammary dysplasia may re-
fication of early disease and staging of metastases, sult in moderate but diffuse increased tracer uptake.
assessment of therapeutic effectiveness, and to de- Attenuation-corrected PET images provide quan-
termine tumor aggressiveness. The uptake mecha- titative information about the tracer concentration
nism and biochemical pathway of the glucose in tissue. Various approaches of different complex-
analog FDG has been extensively studied both in ity can be applied for quantitative PET analysis.
vitro and in vivo. The transport of the radiotracer Standardized uptake values (SUV) are frequently
through the cell membrane via glucose transport being calculated, providing a semiquantitative mea-
proteins, particularly glucose transporter type 1 sure of FDG accumulation in tissue by normaliz-
(GLUT-1), and subsequent intracellular phosphory- ing the tissue radioactivity concentration measured
lation by hexokinase (HK) have been identified as with PET to injected dose and patient’s body
key steps for subsequent tissue accumulation [18]. weight. Quantitative methods may be used to com-
Because FDG-6-phosphate is not a suitable sub- plement visual image analysis for differentiation
strate for glucose-6-phosphate isomerase, and the between benign and malignant breast tumors;
enzyme level of glucose-6-phosphatase is generally that is, by using an SUV-normalized scale for
low in tumors, FDG-6-phosphate accumulates in image display [22]. In particular, SUV correction
cells and is visualized by PET. for partial volume effects and normalization to
blood glucose has been shown to yield the highest
diagnostic accuracy for breast imaging. Corre-
Positron emission tomography imaging sponding threshold values for optimal tumor char-
procedure and image analysis in the breast acterization have been published for various
To ensure a standardized metabolic state, including quantification methods [22]. Dynamic data acqui-
low plasma glucose levels, oncology patients must sition allows calculation of the tracer influx con-
fast for at least 4 to 6 hours before administration stant, although this procedure is more complex and
of FDG. Blood glucose level before tracer injection did not increase diagnostic accuracy.
should ideally not exceed 150 mg/100 ml. Intra- Whole-body imaging can be improved by in-
venous administration of about 300 to 400 MBq travenous injection of furosemide (20–40 mg) to
(~10 mCi) F-18 FDG is used in most studies; how- reduce tracer retention in the urinary system and
ever, Adler and colleagues [19,20] reported on a butylscopolamine (20–40 mg) to reduce FDG up-
higher breast cancer detection rate using up to take in the bowel [23]. Image evaluation requires
750 MBq (~20 mCi) F-18 FDG. To avoid artificial an appreciation of the normal physiologic FDG
FDG-PET Imaging for Breast Cancer 5

Fig. 1. Transaxial FDG-PET image (A), CT image (B), and fused PET/CT image (C), demonstrating a focal area of
intense FDG uptake in the left breast.

Fig. 2. Moderate FDG uptake in a local recurrence in the left breast. Transaxial FDG-PET (A), CT (B), and fused
PET/CT (C).
6 Avril & Adler

Fig. 3. Frontal maximum-intensity projection PET image (A) and sagittal slices from a PET/CT scan (B) show a
mammographically occult 8-mm breast cancer detected incidentally by FDG-PET during a workup for presumed
recurrent breast cancer in a patient with rising carcinoembryonic antigen. An ultrasound guided biopsy demon-
strated an invasive ductal carcinoma, which was resected with clear margins.

uptake distribution and of variation between indi- granulomatous disorders, healing fractures, and post-
viduals, as well as consideration of artifacts and radiation changes.
benign conditions that can mimic malignancy.
Increased FDG uptake is found within the brain Positron emission tomography imaging of
cortex, the myocardium, and the urinary tract. the breast
Low-to-moderate uptake is seen in the base of the The first FDG imaging in breast cancer patients,
tongue, salivary glands, thyroid, liver, spleen, gas- using a collimated gamma camera, was reported
trointestinal tract, bone marrow, musculature, and in 1989 by Minn and Soini [24]. Shortly afterwards,
reproductive organs. Of particular importance is Kubota and coworkers [25] reported on PET imag-
the inconsistent amount of normal uptake in glan- ing with FDG in one case with local recurrence.
dular breast tissue. The most common normal In a first series of 10 patients who had locally
cause of misinterpretation is related to muscle ac- advanced breast cancer, Wahl and colleagues [26]
tivity. Muscle tension may lead to increased FDG successfully identified all breast carcinomas. Subse-
uptake, and physical activity immediately before or quent studies including a limited number of pa-
after tracer injection can lead to spurious muscle tients, predominantly having advanced stages of
activity. In some patients, supraclavicular uptake disease, suggested a high accuracy of FDG-PET
has been shown to represent brown fat. Inflam- for the detection of primary breast carcinomas
matory and infectious processes also demonstrate [Figs. 1–7] [19,24–26]. The largest patient group
increased FDG uptake, as well as some benign reported to date includes 144 patients who had
diseases, such as Paget’s disease, Graves’ disease, 185 histologically confirmed breast tumors [27].

Fig. 4. Transaxial FDG-PET (A), and fused PET/CT image (B) with a small area of increased FDG uptake in a axillary
lymph node metastasis.
FDG-PET Imaging for Breast Cancer 7

Fig. 5. Coronal PET/CT images demonstrate a false-positive axillary lymph node finding in a patient 2 weeks
following a flu shot (arrows). There is also extensive, mild FDG uptake corresponding to metabolically active fat
in the supraclavicular soft tissues.

PET detected breast cancer with an overall sensitiv- accuracy (88% versus 84%) for both methods in
ity of 64.4% by conservative image reading (regard- 32 patients. The sensitivity of FDG-PET was 79%,
ing only definite FDG uptake as positive), and whereas MRI detected all primary breast carci-
80.3% by sensitive image reading (regarding equivo- nomas; however, the specificity of FDG-PET was
cal as well as definite FDG uptake as positive). When higher (94% versus 72%). Baslaim and coworkers
applying sensitive image reading, however, speci- [31] evaluated the usefulness of FDG-PET for diag-
ficity decreased from 94.3% to 75.5% [27,28]. nosing and staging of inflammatory breast cancer.
Schirrmeister and coworkers [29] found similar All 7 patients studied presented with diffuse breast
results in 117 patients, with a sensitivity of 93% enlargement, redness, and peau d’orange. PET
and specificity of 75%. The use of non-attenuation– showed diffuse FDG uptake in the involved breast,
corrected PET imaging combined with sensitive im- with intense uptake in the primary tumor as well as
aging reading may have contributed to the higher increased FDG uptake in the skin.
sensitivity and lower specificity in this study. A The ability of PET to detect breast cancer greatly
recent study compared MR imaging of the breast depends on tumor size. Regarding small tumors,
with FDG-PET [30] found a comparable diagnostic only 30 (68.2%) out of 44 breast carcinomas at

Fig. 6. Mediolateral radiograph mammograms of both breasts showed heterogeneously dense breast parenchyma
with bilateral masses (arrows). Ultrasound-guided right breast biopsy showed invasive ductal carcinoma
(A, arrow), whereas left breast ultrasound-guided biopsy showed papilloma (B, arrow). PEM of right breast
showed dominant known invasive cancer (C, arrow) and smaller foci in upper breast, proving multicentric cancer
with positive lymph node (arrowhead), and increased focal activity on left side (D, arrow) that was confirmed as
DCIS at surgery.
8 Avril & Adler

Fig. 7. (A–D) Mammogram, FDG-PEM, whole body PET, and CT in a patient with DCIS.

stage pT1 (<2 cm) were correctly identified, com- cancer were identified by PET [27]. Nevertheless,
pared with 57 (91.9%) out of 62 at stage pT2 (>2– Schirrmeister and colleagues [29] found that PET
5 cm) [27]. Sensitivity for tumors less than 1 cm was twice as sensitive in detecting multifocal
(pT1a and b) was only 25%, compared with 84.4% lesions (sensitivity 63%, specificity 95%) than the
for tumors between 1 and 2 cm in diameter (pT1c). combination of mammography and ultrasound
Table 1 provides more detailed information. (sensitivity 32%, specificity 93%).
Invasive lobular carcinomas were more often The diagnosis of in-situ carcinomas has increased
false-negative (65.2%) than invasive ductal carci- over the past decade, mainly due to increased use
nomas (23.7%). These results are consistent with a of and technological improvements in mammogra-
previous report from Crippa and colleagues [32], phy. There is little information available about
who found higher glucose metabolism for invasive the ability of PET imaging to detect noninvasive
ductal carcinomas (median SUV of 5.6) versus breast cancer. Tse and coworkers [36] studied
invasive lobular carcinomas (median SUV of 3.8). 14 patients and found that one out of two false-
This is of particular importance in the clinical negative cases had predominantly intraductal can-
application of PET, because lobular carcinomas cer with microscopic invasive foci. In 12 patients,
are more difficult to diagnose by imaging proce- (10 DCIS and 2 LCIS) none out of six in-situ
dures such as mammography, sonography, and carcinomas smaller than 2 cm could be identified
MRI [33–35]. The identification of multifocal or [27]. For larger in-situ carcinomas, three (50%)
multicentric breast cancer plays an important role out of six displayed increased FDG uptake. Al-
in the decision of therapy, because it limits breast- though the number of patients studied was small,
conserving surgery. Only 9 (50%) out of 18 pa- these data suggest that PET imaging cannot con-
tients who had multifocal or multicentric breast tribute to an improved diagnosis of noninvasive
breast cancer.
Vranjesevic and colleagues [37] evaluated the
Table 1: Sensitivity of FDG-PET and the size of influence of the breast tissue density on FDG
breast cancer uptake of normal breast tissue, and found signifi-
TNM Size n Sensitivity cantly lower SUVs for primarily fatty breasts than
for dense breasts. Benign conditions of the breast
pTis 12 42% are more common, and are often difficult to differ-
pT1 <2.0 cm 44 68% entiate from breast cancer in conventional imaging
pT1a <0.5 cm 4 25%
modalities. In general, benign breast masses display
pT1b >0.5–1.0 cm 8 25%
pT1c >1.0–2.0 cm 32 84% low FDG uptake. Only 3 out of 53 benign breast
pT2 >2.0–5.0 cm 62 92% masses presented with focally increased tracer
>2.0–3.0 cm 33 94% uptake, including one rare case of a ductal ade-
>3.0–4.0 cm 15 87% noma, one case with dysplastic tissue, and one
>4.0–5.0 cm 14 93% fibroadenoma [27]. Fibroadenomas are common
pT3 >5.0 cm 14 100% benign tumors, and only 1 out of 9 displayed in-
creased tracer uptake. Moreover, dysplastic tissue
Data from Avril N, Rose CA, Schelling M, et al. Breast im-
aging with positron emission tomography and fluorine-
often accounts for false-positive results in MRI,
18 fluorodeoxyglucose: use and limitations. J Clin Oncol predominantly showing a diffuse pattern of little
2000;18(20):3495–502. or moderate FDG uptake.
FDG-PET Imaging for Breast Cancer 9

Biological features of breast cancer differentiation, histopathologic grading, cell prolif-


FDG uptake in breast cancer shows considerable eration, or axillary lymph node status.
variation. To address that issue, the degree of tracer
accumulation was recently correlated with various Loco-regional staging
tumor characteristics [Table 2] [38]. Histologic sec-
tions of breast cancer specimens were analyzed for The axillary lymph node status is still considered
histologic type, microscopic tumor growth pattern, the single most important prognostic indicator in
percentage of tumor cells, presence of inflamma- patients who have breast cancer. Clinical examina-
tory cells, density of blood vessels, histopathologic tion is generally unreliable for staging the axilla
grading, tumor cell proliferation (mitotic rate and [39]. Lack of conventional imaging techniques to
antibody binding of MIB-1), expression of estrogen determine the axillary lymph node involvement
and progesterone receptors, and expression of the with acceptable accuracy has been the main reason
glucose transporter protein Glut-1. Invasive ductal for axillary lymph node dissection; however, up to
carcinomas displayed significantly higher FDG 70% of patients who have stage T1 and T2 tumors
uptake compared with invasive lobular carci- have negative axillary lymph nodes [40]. The ex-
nomas. The SUVs for clearly defined lesions were tent, morbidity, and cost of the staging procedure
higher compared with tumors with diffuse growth of axillary lymph node dissection are often greater
patterns. Lower densities of blood vessels cor- than those of the surgical treatment of the primary
responded to higher FDG uptakes. In addition, tumor. In anatomical based imaging modalities,
there was a positive correlation between FDG up- such as computed tomography, ultrasound, and
take and tumor cell proliferation, but only a weak MRI, the size of a particular lymph node is of
relationship between FDG uptake and the percen- crucial importance to determine the tumor involve-
tage of tumor cells. There was no relationship be- ment. Generally, lymph node enlargement over
tween FDG uptake and tumor size; axillary lymph 1 cm in diameter is the decisive criterion. In con-
node status; percentage of necrotic, fibrotic and trast, metabolic imaging with FDG-PET is suggested
cystic compounds; presence of inflammatory cells; to provide more specific information, based on de-
steroid receptor status; and expression of Glut-1. tecting increased glucose consumption of cancer
Histologic and immunohistochemical tissue tissue. In 1991, Wahl and coworkers [26] studied
analysis could not fully explain the variation of 12 patients who had locally advanced breast cancer,
FDG uptake in breast cancer. The study authors and found increased FDG uptake in axillary metas-
concluded that the degree of metabolic changes in tases. In 50 patients, Adler and colleagues [20]
malignant tumors is mainly caused by complex reported a sensitivity of 95%, a negative predictive
interactions between the cellular energy metabo- value of 95%, and an overall accuracy of 77% for
lism and the microenvironment of the tumor. axillary PET imaging. Greco and coworkers [41]
Hence FDG-PET uptake can not be used to predict studied 167 consecutive breast cancer patients,
the biologic behaviors of breast cancer, such as and axillary involvement was detected in 68 of
72 patients, resulting in a sensitivity of 94.4% and
a specificity of 86.3%; overall accuracy of lymph
node staging with PET was 89.8%. There is some
Table 2: FDG-uptake versus histology/ controversy about the sensitivity of axillary PET
immunohistochemistry imaging, however [42]. It is a well-known phe-
nomenon that the true FDG uptake is underesti-
SUV vs. p n
mated in small cancer deposits because of the
Histology (ductal versus lobular) 0.003 50 limited spatial resolution of current PET devices,
Tumor growth (nodular versus 0.007 49 (approximately 6–8 mm). Therefore it cannot be
diffuse) expected that PET will provide visualization of
Grading 0.69 50
micrometastases. Avril and colleagues [43] studied
% tumor cells 0.06 50
Inflammatory cells 0.74 50 51 patients and found overall sensitivity and speci-
Capillaries 0.08 50 ficity for detection of axillary lymph node metas-
Proliferation (MIB-1) 0.009 40 tases to be 79% and 96%, respectively. In patients
Estrogen receptor status 0.47 42 who had primary breast tumors larger than 2 cm
Progesterone receptor status 0.29 42 (>stage pT1), the sensitivity increased to 94%, with
Glut-1 transporter protein 0.21 45 a corresponding specificity of 100%. PET could
not identify lymph node metastases in 4 out of
Data from Avril N, Menzel M, Dose J, et al. Glucose me-
tabolism of breast cancer assessed by 18F-FDG PET: his-
6 patients who had small primary breast cancer
tologic and immunohistochemical tissue analysis. J Nucl (stage pT1), however, which led to a sensitivity of
Med 2001;42(1):9–16. only 33% in this group. Although the number of
10 Avril & Adler

patients studied is small, this study clearly points other hand, among patients who have larger tu-
out that the current achievable spatial resolution mors, sentinel biopsy can be avoided in those
of PET imaging limits the detection of microme- who have positive FDG-PET, in whom complete
tastases and small tumor-infiltrated lymph nodes. axillary lymph node dissection should be the pri-
This conclusion is also supported by others; for mary procedure. FDG-PET cannot replace the axil-
example, by Schirrmeister and coworkers [29], lary dissection, not only because of the limited
who studied 117 breast cancer patients and found sensitivity, but also because the number of involved
similar results (sensitivity 79%, specificity 92%). In lymph nodes and extranodal extension cannot be
a prospective multicenter study representing the determined. In patients who have locally advanced
largest patient cohort so far [44], 360 women disease and who are undergoing primary chemo-
who had newly diagnosed invasive breast cancer therapy, however, FDG-PET seems to be a reliable
underwent PET imaging. Three experienced readers method to determine the extent of disease.
blindly interpreted PET images, and the results Recently, the intrathoracic lymph node status has
from 308 axillae were compared with histopathol- been retrospectively analyzed comparing CT and
ogy. If at least one probably or definitely abnormal PET [48]. In 73 consecutive patients who had recur-
axillary focus was considered positive, the sensitiv- rent or metastatic disease, PET was able to correctly
ity for PET was 61% and the specificity was 80%. identify 40% of the patients who had intrathoracic
Patients who had false-negative PET had signifi- lymph node metastases, resulting in a sensitivity of
cantly smaller and fewer tumor-positive lymph 85% and a specificity of 90%. Only 23% of the
nodes than true-positive cases. Semiquantitative patients had suspiciously enlarged lymph nodes in
analysis of axillary FDG uptake showed that a CT, leading to a sensitivity of 54% and a specificity
nodal standardized uptake value (lean body mass) of 85%. PET and CT were both positive in 22% of
of more than 1.8 had a positive predictive value of the cases. Therefore overall diagnostic accuracy of
90% but a sensitivity of only 32%. Finding two or PET was higher (88%) than that of CT (73%).
more intense foci of tracer uptake in the axilla was Despite the limitation in detection of small tumor
highly predictive of axillary metastasis, but had a deposits, FDG-PET is currently the most sensitive
sensitivity of only 27%. imaging modality to detect lymph node metastases,
Sentinel node biopsy has become accepted as a including parasternal and mediastinal nodes.
reliable method of predicting the status of the axilla
in early stages of breast cancer [40]. There are few
Positron emission mammography
studies available directly comparing the diagnostic
accuracy of PET with sentinel node biopsy in breast Because of the limited resolution of conventional
cancer patients. In one study [45], 5 out of 15 pa- PET scanners (4.8 to 7.1 mm in plane resolution),
tients had sentinel lymph node metastases, but the even more limited resolution achieved with
PET identified only 1 of these patients. The size current clinical protocols (1 cm or greater), and
of missed metastases ranged from a small micro- the loss of contrast due to scatter, conventional
metastasis identified only by immunohistochemis- PET scanners are unlikely to detect the small carci-
try to an 11-mm, tumor-involved lymph node. nomas that are detectable with other imaging mo-
Another study [46] included 24 clinically node- dalities such as mammography and MRI. Dedicated
negative breast cancer patients who had primary breast PET—positron emission mammography
tumors smaller than 3 cm, and axillary staging by (PEM)—was developed to improve both resolution
PET was accurate in 15 of 24 patients (62.5%). PET and contrast of breast lesions and thereby improve
was false-negative in 8 of 10 node-positive patients, small lesion detectability, while reducing the cost
and false-positive in 1 patient. The sensitivity and of imaging [49]. By optimizing scanner geometry
specificity of FDG-PET were 20% and 93%, respec- for breast imaging, a dedicated breast PET scanner
tively. The mean diameter of false-negative axillary can achieve much higher resolution than a whole
lymph node metastases was 7.5 mm, and ranged body scanner, while maintaining higher count sen-
from 1 to 15 mm. In 32 breast cancer patients who sitivity and a marked reduction in scatter [50,51].
had clinically negative axillary nodes, sentinel Radiographic mammography uses compression to
lymph node biopsy was false-negative in 1 patient, reduce the mean radiographic travel path, thereby
whereas PET missed lymph node metastases in reducing scatter. In an analogous manner, incor-
11 patients, resulting in a sensitivity of 20% [47]. porating compression into PET imaging reduces
These studies clearly indicate the limitation of the mean travel path of gamma emissions, with a
FDG-PET for axillary staging of small primary resulting reduction in scatter. Because scatter rep-
tumors. FDG-PET is not accurate enough in clini- resents such a low proportion of incident pho-
cally node-negative breast cancer patients qualify- tons, septa or collimation is unnecessary, and a
ing for sentinel lymph node dissection. On the three-dimensional acquisition with full three-
FDG-PET Imaging for Breast Cancer 11

dimensional reconstruction is performed. An added when added to mammography, ultrasound, and


benefit of compression is that images can be pro- clinical examination, without reducing accuracy.
duced with a correspondence to radiograph mam- One of three lesions with atypical ductal hyper-
mograms, so PEM can be used to interrogate the plasia showed intense FDG uptake. Five (12%) of
functional status of abnormalities detected by mam- 41 other proven benign lesions showed intense
mography. Final output resolution of commercially FDG uptake, including two fibroadenomas, two
available equipment is approximately 1.5 mm in fibrocystic changes, and one fat necrosis in a parti-
plane. Resolution orthogonal to the imaging planes cipant who had transverse rectus abdominis muscle
is dependant on the distance between the flat plate (TRAM) flap reconstruction. The first two readers
detectors (ie, the compressed thickness of the breast) agreed on overall PEM assessment (ie, recommen-
and is generally not as good as in-plane resolution dation for biopsy or not) for 83% of the index
[52]. The dramatic improvement in resolution lesions and 80% of the incidental lesions. Examin-
achieved results in the ability to detect smaller ing individual reader performance, at least one
objects than would be detectable using a whole- reader missed a malignancy in 8 index lesions
body scanner (assuming identical object-to-back- and 3 incidental lesions. Examining all readings
ground activity ratios) or to detect small objects of index lesions, the average area under the receiver
with lower object-to-background activity ratios operating characteristic (ROC) curve was 0.91.
than would be detectable with whole body PET [19]. Qualitatively high and asymmetric FDG uptake,
A recent multicenter study [52,53] demonstrated as compared with radiograph mammograms, was
that high-resolution FDG-PEM detects in-situ com- the finding most predictive of malignancy. Al-
ponents of cancers better than any other modality. though promising, additional prospective studies
This fact has been documented in retrospective are needed to determine the clinical role of FDG-
surgical studies [52] as well as prospective imaging PEM imaging in the future.
studies [53]. In the last-cited study, 93 consecutive
subjects who had biopsy-proven breast cancer or
suspicious breast lesions were recruited from four Current clinical status of F-18
sites [53]. Only evaluable cases (eg, with proof of fluorodeoxyglucose-positron emission
pathology) were shown to a blinded panel of imag- tomography in primary breast cancer
ing specialists for review. Patients who had Type I Breast cancer often displays only moderately in-
or poorly controlled Type II diabetes were excluded creased FDG uptake, and considering the limited
from participating in the study. Other diabetic spatial resolution of FDG-PET, metabolic imaging
patients were included, with the prospective deci- results in a low sensitivity to detect small breast
sion that diagnostic performance of the PEM scans carcinomas, micrometastases, and small-tumor–
would be subsequently analyzed to determine infiltrated lymph nodes. The restricted sensitivity of
whether diagnostic accuracy would differ in certain FDG-PET does not allow the screening of asymptom-
subsets of patients (eg, those who had diabetes or atic women for breast cancer. Moreover, negative PET
other medical conditions). Among index cancers in results in patients who have suspicious breast masses
the 77 evaluable subjects reviewed, 39 (93%) out or abnormal mammography do not exclude breast
of 42 were PEM-positive. Including incidental cancer. Therefore, PET imaging may not be used as a
lesions, 43 (90%) out of 48 malignancies were routine application for evaluation of primary breast
PEM-positive, including 10 (91%) out of 11 lesions tumors, and currently cannot significantly reduce
of DCIS and 33 (89%) out of 37 invasive carcino- unnecessary invasive procedures in patients sus-
mas. Three index malignancies (a 3-mm Grade II/ pected of having breast cancer. Advances in technol-
III infiltrating and intraductal carcinoma, a 6-mm ogy such as the development of dedicated breast
Grade I/III tubular carcinoma, and a 10-mm imaging devices (eg, PEM) may improve the detec-
Grade I/III invasive ductal carcinoma with a positive tion of primary tumors with PET in the future. In
axillary lymph node) were occult on FDG-PEM, but patients who have locally advanced breast cancer,
were visible mammographically. One contralateral PET accurately determines the extent of disease, par-
25-mm invasive lobular carcinoma was visible mam- ticularly the loco-regional lymph node status. In
mographically, but was only recognized by one of smaller tumors, the sentinel node biopsy has become
three PEM readers. Three DCIS foci (Grade II/III) the standard of care in many institutions.
were visible on FDG-PEM, but were mammographi-
cally occult. The combination of mammography,
ultrasound, and FDG-PEM depicted 47 (98%) out References
of 48 cancers, with one case of contralateral Paget’s [1] Sant M, Allemani C, Berrino F, et al. Breast car-
disease caused by Grade II DCIS missed on all cinoma survival in Europe and the United States.
imaging modalities. FDG-PEM improved sensitivity Cancer 2004;100(4):715–22.
12 Avril & Adler

[2] Banerjee M, George J, Song EY, et al. Tree-based glucose positron emission tomography in breast
model for breast cancer prognostication. J Clin cancer. Eur J Nucl Med 1999;26(3):226–30.
Oncol 2004;22(13):2567–75. [22] Avril N, Bense S, Ziegler SI, et al. Breast imaging
[3] Fitzgibbons PL, Page DL, Weaver D, et al. Prog- with fluorine-18-FDG PET: quantitative image
nostic factors in breast cancer. College of Ameri- analysis. J Nucl Med 1997;38(8):1186–91.
can Pathologists Consensus Statement 1999. [23] Stahl A, Weber WA, Avril N, et al. Effect of
Arch Pathol Lab Med 2000;124(7):966–78. N-butylscopolamine on intestinal uptake of
[4] Sickles EA. Breast masses: mammographic evalua- fluorine-18-fluorodeoxyglucose in PET imaging
tion. Radiology 1989;173:297–303. of the abdomen. Nuklearmedizin 2000;39(8):
[5] Kopans DB, Feig SA. False positive rate of screen- 241–5.
ing mammography. N Engl J Med 1998;339(8): [24] Minn H, Soini I. F-18 fluorodeoxyglucose scintig-
562–4. raphy in diagnosis and follow up of treatment in
[6] Kopans DB. The positive predictive value of mam- advanced breast cancer. Am J Clin Pathol 1989;
mography. AJR Am J Roentgenol 1992;158:521–6. 91(5):535–41.
[7] Liberman L, Menell JH. Breast imaging reporting [25] Kubota K, Matsuzawa T, Amemiya A, et al. Imag-
and data system (BI-RADS). Radiol Clin North ing of breast cancer with F-18 fluorodeoxy-
Am 2002;40(3):409–30, v. glucose and positron emission tomography. J
[8] Meyer JE, Eberlein TJ, Stomper PC, et al. Biopsy Comput Assist Tomogr 1989;13(6):1097–8.
of occult breast lesions. Analysis of 1261 abnor- [26] Wahl RL, Cody RL, Hutchins GD, et al. Primary
malities. JAMA 1990;263:2341–3. and metastatic breast carcinoma: initial clinical
[9] Sickles EA, Miglioretti DL, Ballard-Barbash R, evaluation with PET with the radiolabeled
et al. Performance benchmarks for diagnostic glucose analogue 2-[F-18]-fluoro-2-deoxy- D-
mammography. Radiology 2005;235(3):775–90. glucose. Radiology 1991;179(3):765–70.
[10] Bird RE, Wallace TW, Yankaskas BC. Analysis [27] Avril N, Rose CA, Schelling M, et al. Breast
of cancer missed at screening mammography. imaging with positron emission tomography and
Radiology 1992;184:613–7. fluorine-18 fluorodeoxyglucose: use and limita-
[11] Fine RE, Staren ED. Updates in breast ultra- tions. J Clin Oncol 2000;18(20):3495–502.
sound. Surg Clin North Am 2004;84(4):1001–34, [28] Avril N, Dose J, Jänicke F, et al. Metabolic
v–vi. characterization of breast tumors with positron
[12] Mendelson EB. Problem-solving ultrasound. emission tomography using F-18 fluorodeoxy-
Radiol Clin North Am 2004;42(5):909–18, vii. glucose. J Clin Oncol 1996;14(6):1848–57.
[13] Simmons R. Ultrasound in the changing ap- [29] Schirrmeister H, Kuhn T, Guhlmann A, et al.
proaches to breast cancer diagnosis and treat- Fluorine-18 2-deoxy-2-fluoro-D-glucose PET in
ment. Breast J 2004;10(Suppl 1):S13–4. the preoperative staging of breast cancer: com-
[14] Lee CH. Problem solving MR imaging of parison with the standard staging procedures.
the breast. Radiol Clin North Am 2004;42(5): Eur J Nucl Med 2001;28(3):351–8.
919–34, vii. [30] Goerres GW, Michel SC, Fehr MK, et al. Follow-
[15] Hylton N. Magnetic resonance imaging of the up of women with breast cancer: comparison
breast: opportunities to improve breast cancer between MRI and FDG PET. Eur Radiol 2003;
management. J Clin Oncol 2005;23(8):1678–84. 13(7):1635–44.
[16] Chen X, Moore MO, Lehman CD, et al. Com- [31] Baslaim MM, Bakheet SM, Bakheet R, et al. 18-
bined use of MRI and PET to monitor response Fluorodeoxyglucose-positron emission tomogra-
and assess residual disease for locally advanced phy in inflammatory breast cancer. World J Surg
breast cancer treated with neoadjuvant chemo- 2003;27(10):1099–104.
therapy. Acad Radiol 2004;11(10):1115–24. [32] Crippa F, Seregni E, Agresti R, et al. Association
[17] Rohren EM, Turkington TG, Coleman RE. between F-18 fluorodeoxyglucose uptake and
Clinical applications of PET in oncology. Radi- postoperative histopathology, hormone receptor
ology 2004;231(2):305–32. status, thymidine labelling index and p53 in
[18] Brown RS, Wahl RL. Overexpression of Glut-1 primary breast cancer: a preliminary observation.
glucose transporter in human breast cancer. Eur J Nucl Med 1998;25(10):1429–34.
An immunohistochemical study. Cancer 1993; [33] Krecke KN, Gisvold JJ. Invasive lobular car-
72(10):2979–85. cinoma of the breast: mammographic findings
[19] Adler LP, Crowe JP, al-Kaisi NK, et al. Evaluation and extent of disease at diagnosis in 184 patients.
of breast masses and axillary lymph nodes with AJR Am J Roentgenol 1993;161(5):957–60.
[F-18] 2- deoxy-2-fluoro-D-glucose PET. Radiol- [34] Gilles R, Guinebretière J-M, Lucidarme O, et al.
ogy 1993;187(3):743–50. Nonpalpable breast tumors: diagnosis with con-
[20] Adler LP, Faulhaber PF, Schnur KC, et al. Axillary trast-enhanced subtraction dynamic MR imaging.
lymph node metastases: screening with [F-18] Radiology 1994;191:625–31.
2-deoxy-2-fluoro-D-glucose (FDG) PET. Radiol- [35] Paramagul CP, Helvie MA, Adler DD. Invasive
ogy 1997;203(2):323–7. lobular carcinoma: sonographic appearance and
[21] Boerner AR, Weckesser M, Herzog H, et al. Opti- role of sonography in improving diagnostic
mal scan time for fluorine-18 fluorodeoxy- sensitivity. Radiology 1995;195(1):231–4.
FDG-PET Imaging for Breast Cancer 13

[36] Tse NY, Hoh CK, Hawkins RA, et al. The ap- [45] Kelemen PR, Lowe V, Phillips N. Positron emis-
plication of positron emission tomographic sion tomography and sentinel lymph node dis-
imaging with fluorodeoxyglucose to the evalua- section in breast cancer. Clin Breast Cancer 2002;
tion of breast disease. Ann Surg 1992;216(1): 3(1):73–7.
27–34. [46] Fehr MK, Hornung R, Varga Z, et al. Axillary
[37] Vranjesevic D, Schiepers C, Silverman DH, et al. staging using positron emission tomography in
Relationship between 18F-FDG uptake and breast cancer patients qualifying for sentinel
breast density in women with normal breast lymph node biopsy. Breast J 2004;10(2):89–93.
tissue. J Nucl Med 2003;44(8):1238–42. [47] Barranger E, Grahek D, Antoine M, et al. Evalua-
[38] Avril N, Menzel M, Dose J, et al. Glucose me- tion of fluorodeoxyglucose positron emission
tabolism of breast cancer assessed by 18F-FDG tomography in the detection of axillary lymph
PET: histologic and immunohistochemical tissue node metastases in patients with early-stage
analysis. J Nucl Med 2001;42(1):9–16. breast cancer. Ann Surg Oncol 2003;10(6):
[39] Sacre RA. Clinical evaluation of axillar lymph 622–7.
nodes compared to surgical and pathological [48] Eubank WB, Mankoff DA, Takasugi J, et al.
findings. Eur J Surg Oncol 1986;12(2):169–73. 18fluorodeoxyglucose positron emission tomog-
[40] Veronesi U, Paganelli G, Galimberti V, et al. raphy to detect mediastinal or internal mammary
Sentinel-node biopsy to avoid axillary dissection metastases in breast cancer. J Clin Oncol 2001;
in breast cancer with clinically negative lymph- 19(15):3516–23.
nodes. Lancet 1997;349(9069):1864–7. [49] Murthy K, Aznar M, Thompson CJ, et al. Results
[41] Greco M, Crippa F, Agresti R, et al. Axillary lymph of preliminary clinical trials of the positron
node staging in breast cancer by 2-Fluoro-2- emission mammography system PEM-I: a dedi-
deoxy-D-glucose-positron emission tomography: cated breast imaging system producing glucose
Clinical evaluation and alternative management. metabolic images using FDG. J Nucl Med 2000;
J Natl Cancer Inst 2001;93(8):630–5. 41(11):1851–8.
[42] Torrenga H, Licht J, van der Hoeven JJ, et al. Re: [50] Bergman AM, Thompson CJ, Murthy K, et al.
axillary lymph node staging in breast cancer by Technique to obtain positron emission mam-
2-fluoro-2-deoxy-D-glucose-positron emission mography images in registration with x-ray
tomography: clinical evaluation and alternative mammograms. Med Phys 1998;25(11):2119–29.
management. J Natl Cancer Inst 2001;93(21): [51] Weinberg I, Majewski S, Weisenberger A, et al.
1659–61. Preliminary results for positron emission mam-
[43] Avril N, Dose J, Jänicke F, et al. Assessment of mography: real-time functional breast imaging in
axillary lymph node involvement in breast a conventional mammography gantry. Eur J Nucl
cancer patients with positron emission tomogra- Med 1996;23(7):804–6.
phy using radiolabeled 2-(fluorine-18)-fluoro- [52] Weinberg IN, Beylin D, Zavarzin V, et al. Posi-
2-deoxy-D-glucose. J Natl Cancer Inst 1996; tron emission mammography: high-resolution
88(17):1204–9. biochemical breast imaging. Technol Cancer Res
[44] Wahl RL, Siegel BA, Coleman RE, et al. Prospec- Treat 2005;4(1):55–60.
tive multicenter study of axillary nodal staging by [53] Tafra L, Cheng Z, Uddo O, et al. Pilot clinical
positron emission tomography in breast cancer: trial of FDG positron emission mammography
a report of the staging breast cancer with PET (PEM) in the surgical management of breast
Study Group. J Clin Oncol 2004;22(2):277–85. cancer. Am J Surg 2005;190:628–32.
15

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 15–24

Diagnosis of Recurrent and


Metastatic Disease Using F-18
Fluorodeoxyglucose-Positron
Emission Tomography
William B. Eubank, MD

& Loco-regional recurrence & Summary


& Intrathoracic lymphatic recurrences & Acknowledgments
& Distant metastases & References
& Impact of F-18 fluorodeoxyglucose-
positron emission tomography on
patient management

Breast cancer is the most common non-skin can- tients who have more advanced or recurrent disease
cer, and the second leading cause of cancer death in [14,15].
women [1]. There are 40,000 women per year The recognition that breast cancer is a systemic
dying of breast cancer in the United States, and disease, even in its early stages, led to the cur-
most breast cancer victims die of progressive meta- rent approach to treatment, which combines local
static disease [1]. Because optimal treatment of pa- measures such as surgery and radiotherapy with
tients who have recurrent breast cancer depends on systemic treatment [16]. For most clinical trial stud-
knowing the true extent of disease, accurate staging ies, local failure is defined as any recurrence of
of these patients is an important public health tumor in the ipsilateral chest wall or mastectomy
problem. This is a useful application of FDG-PET scar; regional failure is defined as any recurrence of
when it is performed to complement conventional tumor in the ipsilateral supraclavicular, infracla-
imaging (CI) such as CT, MRI, and bone scintigra- vicular, axillary, or internal mammary nodes; and
phy. The additional metabolic information pro- recurrence of tumor in any other site is considered
vided by FDG-PET increases the accuracy of detecting as distant failure [17]. In general, systemic therapy
recurrent or metastatic lesions [2–12]. This is par- is used at almost all disease stages; however, iso-
ticularly true in the evaluation of anatomic regions lated loco-regional disease or single sites of meta-
that have been previously treated by surgery or static recurrence are also treated with surgery and
radiation [13], in which the discrimination be- radiation therapy [18,19]. It is hoped that the
tween post-treatment scar and recurrent tumor potential of FDG-PET to provide more accurate
can be problematic. FDG-PET can also significantly and earlier detection of breast cancer recurrences
impact the choice of treatment, especially in pa- will translate into more effective treatment strate-

This work was supported in part by National Institutes of Health (NIH) grants RO1CA42045, RO1CA72064,
RO1CA90771, and S10RR177229.
Department of Radiology, Puget Sound Veterans Administration Health Care System, 1660 South Columbian
Way, Seattle, WA 98108-1597, USA
E-mail address: weubank@u.washington.edu

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.002
pet.theclinics.com
16 Eubank

gies and better health outcomes for these patients or larger, and extranodal extension of 2 mm or
in the future. more [25]. Supraclavicular node recurrence is tech-
nically considered Stage IV disease, and is generally
considered a harbinger to more widely dissemi-
Loco-regional recurrence nated disease; however, patients who have supra-
Recurrence in the breast, skin of the breast, axillary clavicular node involvement as the sole site of
nodes, chest wall, and supraclavicular nodes are the disseminated disease may benefit from aggressive
most common sites of first loco-regional recurrence local radiotherapy.
after primary surgical resection [20,21]. The shift One clinical situation where FDG-PET has been
toward breast-conserving surgery and local radia- shown to be helpful is in the evaluation of pre-
tion therapy for early breast cancer in recent years viously treated patients who have symptoms of
has heightened concern over loco-regional recur- brachial plexopathy. This debilitating condition
rence [22]. The incidence of loco-regional recur- can be secondary to tumor recurrence in the axilla
rence after breast conservation treatment ranges or chest wall, or caused by scarring of tissue neigh-
from 5% to 22% [23]. Independent risk factors asso- boring the brachial plexus from previous surgery
ciated with loco-regional recurrence in this group of or radiation. Because the signs and symptoms of
patients include positive margins at surgical resec- loco-regional recurrence often overlap with the side
tion, tumors with extensive intraductal component, effects of treatment [29,30] and patients who have
high grade ductal carcinoma in situ (DCIS), patient tumor recurrence may benefit from surgical resec-
age under 40 years, and absence of radiation after tion [31,32], it is important to distinguish one from
breast conservation therapy [23,24]. the other. Hathaway and colleagues [33] showed
Among patients treated with mastectomy, axillary the value of combining the functional information
node dissection, and adjuvant chemotherapy, the of FDG-PET and the anatomic information from
most common sites of loco-regional recurrence are dedicated MRI to decide whether patients would
the chest wall (68% of loco-regional recurrences) benefit from further surgery. Other studies [34]
and supraclavicular nodes (41% of loco-regional have confirmed these early findings.
recurrences) [Fig. 1] [25]. Recurrent disease at
both of these sites is associated with poor progno-
Intrathoracic lymphatic recurrences
sis in terms of survival after recurrence [26–28].
Factors that predict an increased risk of chest wall Lymphatic drainage to the internal mammary
or supraclavicular node recurrence include four nodal chain is an important pathway of spread of
or more positive axillary nodes, tumor size 4 cm disease, both at the time of initial diagnosis and

Fig. 1. A 49-year-old woman who underwent bilateral mastectomies and reconstruction with implants 7 years
before recurrence of disease. She originally had extensive lobular carcinoma in situ with two small (2–3 mm) foci
of invasive lobular carcinoma in the right breast. Sentinel node and limited low axillary lymph node dissection
revealed no pathologically involved nodes, and surgical margins were negative. She received no adjuvant
treatment, but opted for prophylactic left mastectomy. At time of recurrence, she presented with palpable left
chest wall mass (biopsy-proven invasive ductal carcinoma) and axillary nodes. Coronal FDG-PET image (A) shows
recurrent chest wall mass (arrowhead; standardized uptake value [SUV] = 8.2) lateral to breast implant, uptake in
several axillary nodes (small arrows; maximum SUV = 4.5), and left supraclavicular node (long arrow; SUV = 2.1). At
more anterior level, coronal FDG-PET image (B) shows uptake in a left internal mammary node (long arrow;
SUV = 2.0) in addition to uptake in left axillary nodes (short arrows) and the left breast (arrowhead).
FDG-PET Diagnosis 17

after primary treatment of breast cancer. Data from ternal mammary and axillary nodal metastasis is
sentinel node lymphoscintigraphy series in patients significantly worse compared with patients who
who had early breast cancer at the author’s institu- have only axillary node disease [40,41], suggesting
tion reveal that overall prevalence of drainage to that internal mammary nodal chain is a conduit for
the internal mammary nodes is 17% [35]. This is a more widespread dissemination of disease.
similar prevalence to that shown in early extended The importance of internal mammary nodal de-
radical mastectomy series [36–38], in which metas- tection and treatment remains controversial [42].
tasis to internal mammary nodes occurred in close Unlike axillary nodes, internal mammary nodes are
to one in five women who had operable (Stage II–- not routinely biopsied as part of an individual
III) breast cancer. Metastasis to internal mammary patient’s staging work-up, and their status is gener-
nodes can occur from tumor located anywhere ally unknown. There has been reluctance to biopsy
in the breast; however, in the series of the author internal mammary nodes because: (1) early radio-
and colleagues, internal mammary drainage was therapy trials (before the era of routine adjuvant
significantly less frequent in tumors located in the chemotherapy) failed to demonstrate a clear bene-
upper outer quadrant (10%) compared with the fit in survival with internal mammary chain radia-
other three quadrants and subareolar portion of tion, and (2) there is a relatively high complication
the breast (17%–29%) [35]. Metastasis to the in- risk (pneumothorax and bleeding) associated with
ternal mammary and axillary nodes usually occurs internal mammary nodal sampling. A recent large,
synchronously but infrequently (4%–6% incidence) prospective, randomized, radiotherapy trial [43]
may be isolated to the internal mammary chain has shown a benefit in systemic relapse-free and
[36,39]. The prognosis of patients who have in- overall survival from aggressive regional nodal irra-

Fig. 2. A 46-year-old woman who presented with sternal pain 5 years after undergoing left mastectomy for
invasive ductal carcinoma (T3, N0) followed by chemotherapy and radiation. Anterior coronal FDG-PET image (A)
shows intense uptake in the left parasternal region (long arrow) and sternum (short arrows; maximum SUV = 9.4)
consistent with disease in the internal mammary node chain with local spread to the sternum. Physiologic uptake
in the anterior heart (arrowhead) is also present. A posterior coronal FDG-PET image of the chest (B) shows linear
uptake along the supradiaphragmatic region (arrow; SUV = 4.1) consistent with pleural metastasis and axial
FDG-PET (C ) and CT (D) of the upper chest show a small lung metastasis (arrows; SUV = 2.0) in addition to sternal
uptake (arrowheads).
18 Eubank

diation (including internal mammary field) fol- section and radiation [Fig. 3]. As with internal
lowing lumpectomy or mastectomy, even in pa- mammary nodes, mediastinal nodes are rarely
tients who had fairly limited spread to the axilla. sampled in breast cancer patients. CT, the conven-
These data suggest that eradication of residual loco- tional method of staging these nodes, relies on
regional metastasis (including internal mammary size criteria to determine the presence or absence
nodal disease) has a strong systemic effect. of disease. This method has been proven signifi-
FDG uptake in the internal mammary nodal chain cantly less accurate than FDG-PET in patients who
has been anecdotally reported in some of the stud- have non-small–cell lung cancer in which histologic
ies that have focused on detection of primary analysis is used as the gold standard [48,49]. In the
tumor or axillary staging [44,45]. In one study of author and colleagues’ retrospective series of 73 pa-
85 patients who underwent FDG-PET before axil- tients who had recurrent or metastatic breast cancer
lary node dissection [44], 12 (14%) had uptake and who underwent both FDG-PET and chest CT
in the internal mammary region, but there was [50], FDG uptake in mediastinal or internal mam-
no histological confirmation of these nodes. The mary nodes was two times more prevalent than
author and colleagues’ experience with imaging suspiciously enlarged nodes by CT, suggesting that
patients who have locally advanced breast cancer PET is a much more sensitive technique at detecting
shows that the prevalence of internal mammary nodal disease. In the subset of patients who had
FDG uptake can be as high as 25%, and that the confirmation, the sensitivity of FDG-PET was sig-
presence of internal mammary FDG uptake pre- nificantly higher (85%) than CT (50%), with nearly
dicts treatment failure patterns of disease consis- the same level of specificity (90% for PET and
tent with internal mammary nodal involvement 83% for CT). Ten of 33 (30%) patients suspected
and progression [Fig. 2] [46]. A preliminary study of having only loco-regional recurrence by CI and
by Bernstein and coworkers [47] showed the fea- clinical examination had mediastinal or internal
sibility of detecting internal mammary nodal me- mammary FDG uptake. Risk factors associated
tastases in early-stage patients using FDG-PET. with mediastinal or internal mammary FDG uptake
FDG-PET may prove to be an ideal method of non- in these patients were recurrent chest wall invasion
invasively staging this important nodal region, and and three or more positive axillary nodes.
may aid in the selection of patients who would
potentially benefit most from directed internal mam-
Distant metastases
mary nodal radiotherapy; however, further work
needs to be done to confirm FDG-PET findings The skeleton is the most common site of distant
with histopathology. metastasis in breast cancer; nearly 70% of patients
Neoplastic spread to mediastinal nodes is also who have advanced disease have bone metastasis
common in patients who have advanced disease, [51]. Bone scintigraphy is considered the most sen-
and the mediastinum is a common site of recurrence sitive method of detecting and determining the
in patients who have undergone axillary node dis- extent of skeletal metastases; however, purely osteo-

Fig. 3. A 65-year-old man with history of multiple chest wall recurrences of estrogen receptor-positive invasive
ductal carcinoma of the right breast. He had undergone multiple chest wall resections, radiation, chemotherapy,
and hormonal therapy for these episodes of recurrent disease. Anterior coronal image from FDG-PET (A)
performed 11 years after original diagnosis of breast cancer shows a focus of uptake in the right anterior chest
(arrow; SUV = 6.0) consistent with pathologic involvement of an internal mammary node. A more posterior
coronal image (B) shows two foci of uptake (arrows) in the right hilum (SUV = 6.4) and the pretracheal region
of the mediastinum (SUV = 5.0).
FDG-PET Diagnosis 19

Fig. 4. A 43-year-old woman with bone-dominant metastatic breast cancer. The posterior and anterior projections
of bone scan (A) show widespread foci of uptake in the axial skeleton and ribs consistent with metastatic disease.
Anterior (B) and posterior (C ) coronal images from FDG-PET performed 1 week after the bone scan show much
more extensive involvement of the axial skeleton, with a predominant intramedullary uptake pattern in the spine.
The pattern of uptake in the ribs is also different compared with the bone scan. The bone scan shows discrete foci
in multiple ribs bilaterally that are not as apparent on FDG-PET; these sites represent areas of active cortical bone
remodeling from either sclerotic metastases or pathologic fractures. The rib activity in the FDG-PET scan is more
diffuse and less discrete, consistent with intramedullary metastases.

lytic lesions or metastases confined to the marrow detection of skeletal metastases in patients who
cavity may be difficult to detect on bone scintigra- have advanced disease have shown conflicting
phy, because of a lack of sufficient osteoblastic re- results [3,4,9,53–56]. Some studies have shown
sponse [52]. Retrospective studies comparing the FDG-PET to be equal or superior to planar bone
sensitivity of bone scintigraphy to FDG-PET in the scintigraphy in the detection of skeletal metastases
20 Eubank

[53,54], whereas others have shown FDG-PET to be help clarify staging in the case of difficult or equivo-
less sensitive [3,4,9,55] on a lesion-based analysis. cal conventional staging. Evolving data suggest that
The results from a study by Cook and colleagues [F-18]-fluoride PET may provide similar and likely
[56], in which skeletal metastases from breast can- improved bone metastasis detection in breast can-
cer patients were categorized into osteoblastic, cer and other tumors compared with bone scin-
osteolytic, or mixed disease subsets based on plain tigraphy [58,59], and may play a role in breast
film or CT findings, provides an explanation for cancer bone metastasis staging in the future.
these conflicting reports. In the study of 23 breast Several studies have shown that whole-body
cancer patients who had known skeletal metasta- FDG-PET is a highly accurate method compared
ses and who underwent both bone scintigraphy with CI for restaging patients who have previously
and FDG-PET, FDG-PET detected more lesions undergone primary treatment for breast cancer. Re-
than bone scintigraphy, except in a subgroup of sults of these studies are summarized in Table 1.
patients who had osteoblastic metastases. More- One advantage of FDG-PET is that large areas can
over, the level of FDG uptake in osteolytic lesions be surveyed; it is therefore able to evaluate sites of
was significantly greater compared with osteoblas- recurrent disease that can be extensive and sepa-
tic lesions, and the prognosis of patients who had rated by large anatomic distances. Several investiga-
osteolytic-predominant disease was significantly tions have shown the added benefit of FDG-PET to
worse. A more recent prospective study compar- CI in asymptomatic patients who had elevated
ing the sensitivity of FDG-PET with bone single tumor marker serum levels and negative or equivo-
photon emission computed tomography (SPECT) cal CI [4,7,8]. Although the ability of FDG-PET to
in 15 patients [57] showed a clear superiority of successfully detect skeletal metastases compared
bone SPECT over FDG-PET in sensitivity in detect- with bone scintigraphy has been mixed in these
ing skeletal metastases (85% for SPECT versus series, as previously discussed, FDG-PET has been
17% for FDG), and corroborated the findings of proven significantly more accurate in the evalua-
Cook and coworkers regarding the improvement in tion of nodal disease, and equal or superior to CI
detection of the subset of osteolytic lesions by for visceral metastases [3,9,50]. In a retrospective
FDG-PET. These data clearly show a complemen- study of 61 patients, Vranjesevic and colleagues
tary nature of bone scintigraphy and FDG-PET in [60] showed that FDG-PET is significantly more
the evaluation of skeletal metastases in breast can- accurate than a combination of CI studies in pre-
cer patients [Fig. 4]. These results also suggest that dicting the outcome (disease-free survival) of pa-
FDG-PET and bone scan should not be considered tients being re-evaluated after primary treatment of
substitutes for each other for bone metastasis stag- breast cancer. These data support the practice at the
ing in breast cancer. In the author’s center, bone author’s institution of using FDG-PET as a comple-
scintigraphy remains one of the routine studies in ment to CI for the evaluation of patients who have
breast cancer metastatic staging, with FDG-PET to suspected recurrent disease.

Table 1: Studies comparing the ability of whole-body FDG-PET with conventional imaging to detect
loco-regional and distant recurrences in patients who have previously undergone primary treatment
for breast cancer
Number of Confirmed positive/ FDG-PET Sensitivity FDG-PET Specificity
Study patients negative cases (TP/TP+FN)a (TN/TN+FP)a
Bender et al [2] 75 60/15 95% (41/43) 96% (213/221)
Moon et al [3] 57 29/28 93% (27/29) 79% (22/28)
Lonneux et al [4] 39b 33/6 94% (31/33) 50% (3/6)
Kim et al [5] 27 17/10 94% (16/17) 80% (8/10)
Lin et al [6] 36 11/25 85% (23/27) 96% (85/89)
Liu et al [7] 30b 28/2 89% (25/28) 50% (1/2)
Suarez et al [8] 38b 26/12 92% (24/26) 75% (9/12)
Gallowitsch et al [9] 62 34/28 97% (33/34) 82% (23/28)
Siggelkow et al [10] 57 31/26 81% (25/31) 96% (25/26)
Kamel et al [11] 60 43/17 89% (24/27) LRR 84% (16/19) LRR
100% (26/26) DM 97% (30/31) DM

Abbreviations: DM, distant metastases; FN, false negative; FP, false positive; LRR, loco-regional recurrence; TN, true
negative; TP, true positive.
a
Values calculated on patient analysis except in [2] and [6], in which values are calculated on lesion analysis.
b
Patients were mostly or all asymptomatic with elevated tumor markers.
FDG-PET Diagnosis 21

uptake is improved; this can potentially lead to an


Impact of F-18 fluorodeoxyglucose-positron
improvement in specificity (reduction in false posi-
emission tomography on patient
tive results) and staging of disease compared with
management
separate evaluation of CT and PET data. PET/CT
A distinguishing feature of the management of also shows promise in radiation treatment plan-
breast cancer is the need for a multidisciplinary ning by modifying target tissue volumes selected
approach for optimal local and regional control for irradiation, based on the combination of mor-
of the disease. Many strategies and agents offer phologic and metabolic data [64]. The impact of
patients who have advanced breast cancer a thera- PET/CT on patient outcomes needs to be addressed
peutic benefit, including surgery, radiation, che- in future studies.
motherapy, and hormonal therapy. Choosing the
most appropriate therapy depends primarily on ac-
curately defining the extent of disease. As of October, Summary
2002, PET has been approved for payment by the FDG-PET is a useful tool in the evaluation of pa-
Center for Medicare and Medicaid Services in the tients who have suspected or proven breast cancer
United States for staging or restaging of patients recurrence. In the clinical setting, it should be used
who have recurrent or metastatic disease, especially as a complement to conventional staging studies
when conventional staging studies are equivocal. and not as a replacement. FDG-PET is generally
Despite this approval, there is relatively little in- more sensitive in the detection of loco-regional
sight into which patients who have recurrent or and distant metastases than conventional imaging,
metastatic breast cancer benefit the most from and is therefore a more accurate method of deter-
FDG-PET. A previous investigation [14] showed mining the true extent of disease. One exception
that FDG-PET had a major impact on the manage- is the detection of sclerotic bone metastases; these
ment of patients who had breast cancer and who lesions are often not metabolically active enough
underwent restaging for local or distant recurrences. for FDG-PET detection, but are readily detected
FDG-PET changed the clinical stage in 36% of pa- by bone scintigraphy. To have the greatest impact
tients and induced changes in therapy in 58% of on patient management, FDG-PET should be used
the patients. In the author and colleagues’ retro- to answer specific clinical questions. In the clinical
spective study of 125 patients who had advanced setting of an asymptomatic patient who has ris-
breast cancer and who were undergoing CI and ing tumor markers and negative or equivocal find-
FDG-PET for staging [15], the extent of disease ings on clinical examination or CI, FDG-PET has
was changed in 67% of patients (increased in the ability to detect otherwise occult disease. Pre-
43% and decreased in 24%), and the therapeutic liminary investigations show that FDG-PET has
plan was altered in 32% of patients based on FDG- the greatest impact on the choice of treatment in pa-
PET findings. FDG-PET altered therapy most fre- tients who have suspected or proven loco-regional
quently in two subgroups of patients: (1) patients recurrence and who are being considered for ag-
who had suspected or proven loco-regional re- gressive curative treatment, and in the evaluation
currence, under consideration for aggressive local of treatment response in patients who have meta-
therapy (FDG-PET altered treatment in 44%), and static disease.
(2) patients who had known metastases being
evaluated for response to therapy (FDG-PET altered
treatment in 33%). These results indicate that FDG- Acknowledgments
PET should not be used as the sole restaging tool
in patients who have recurrent or metastatic dis- The authors wish to acknowledge Erin Schubert for
ease, but should be used to answer specific ques- help with figure reproduction.
tions that will likely impact their management.
Future prospective trials using oncologist-directed
References
questionnaires will help to further define the role
and provide data for the cost-benefit analysis of [1] Greenlee RT, Murray T, Bolden S, et al. Cancer Sta-
FDG-PET in staging patients who have advanced tistics, 2000. CA Cancer J Clin 2000;50(1):7–33.
breast cancer. [2] Bender H, Kirst J, Palmedo H, et al. Value of
18
fluoro-deoxyglucose positron emission tomog-
There have been a few preliminary studies show-
raphy in the staging of recurrent breast carci-
ing promise for FDG-PET/CT in the evaluation of noma. Anticancer Res 1997;17(3B):1687–92.
patients who have recurrent or metastatic breast [3] Moon DH, Maddahi J, Silverman DHS, et al.
cancer [61–63]. Because of the accurate registration Accuracy of whole-body [fluorine-18]-FDG-PET
of metabolic and morphologic images from these for the detection of recurrent or metastatic breast
integrated systems, the accuracy in localizing FDG carcinoma. J Nucl Med 1998;39(3):431–5.
22 Eubank

[4] Lonneux M, Borbath I, Berliere M, et al. The place breast cancer treated by mastectomy and adju-
of whole-body FDG-PET for the diagnosis of vant chemotherapy with or without tamoxifen
distant recurrence of breast cancer. Clin Positron and without radiotherapy: results from five
Imaging 2000;3(2):45–9. National Surgical Adjuvant Breast and Bowel
[5] Kim TS, Moon WK, Lee DS, et al. Fluorodeoxy- Project randomized clinical trials. J Clin Oncol
glucose positron emission tomography for de- 2004;22(21):4247–54.
tection of recurrent or metastatic breast cancer. [18] Schwaibold F, Fowble BL, Solin LJ, et al. The
World J Surg 2001;25(7):829–34. results of radiation therapy for isolated local
[6] Lin WY, Tsai SC, Cheng KY, et al. Fluorine-18 regional recurrence after mastectomy. Int J Radiat
FDG-PET in detecting local recurrence and dis- Oncol Biol Phys 1991;21(2):299–310.
tant metastases in breast cancer–Taiwanese ex- [19] Probstfeld MR, O’Connell TX. Treatment of
periences. Cancer Invest 2002;20(5–6):725–9. locally recurrent breast carcinoma. Arch Surg
[7] Liu CS, Shen YY, Lin CC, et al. Clinical impact 1989;124(10):1127–9 [discussion: 1130].
of [18F]FDG-PET in patients with suspected [20] Valagussa P, Bonadonna G, Veronesi U. Patterns
recurrent breast cancer based on asymptomati- of relapse and survival following radical mas-
cally elevated tumor marker serum levels: a pre- tectomy. Analysis of 716 consecutive patients.
liminary report. Jpn J Clin Oncol 2002;32(7): Cancer 1978;41(3):1170–8.
244–7. [21] Kamby C, Rose C, Ejlertsen B, et al. Stage and
[8] Suarez M, Perez-Castejon MJ, Jimenez A, et al. pattern of metastases in patients with breast
Early diagnosis of recurrent breast cancer with cancer. Eur J Cancer Clin Oncol 1987;23(12):
FDG-PET in patients with progressive elevation 1925–34.
of serum tumor markers. Q J Nucl Med 2002; [22] Schmolling J, Maus B, Rezek D, et al. Breast
46(2):113–21. preservation versus mastectomy—recurrence and
[9] Gallowitsch HJ, Kresnik E, Gasser J, et al. [18F]- survival rates of primary breast cancer patients
fluorodeoxyglucose positron-emission tomogra- treated at the UFK Bonn. Eur J Gynaecol Oncol
phy in the diagnosis of tumor recurrence and 1997;18(1):29–33.
metastases in the follow-up of patients with breast [23] Huston TL, Simmons RM. Locally recurrent
carcinoma: a comparison to conventional imag- breast cancer after conservation therapy. Am J
ing. Invest Radiol 2003;38(5):250–6. Surg 2005;189(2):229–35.
[10] Siggelkow W, Zimny M, Faridi A, et al. The value [24] Leborgne F, Leborgne JH, Ortega B, et al. Breast
of positron emission tomography in the fol- conservation treatment of early stage breast can-
low-up for breast cancer. Anticancer Res 2003; cer: patterns of failure. Int J Radiat Oncol Biol
23(2C):1859–68. Phys 1995;31(4):765–75.
[11] Kamel EM, Wyss MT, Fehr MK, et al. [18F]- [25] Katz A, Strom EA, Buchholz TA, et al. Locore-
fluorodeoxyglucose positron emission tomogra- gional recurrence patterns after mastectomy and
phy in patients with suspected recurrence of breast doxorubicin-based chemotherapy: implications
cancer. J Cancer Res Clin Oncol 2003;129(3): for postoperative irradiation. J Clin Oncol 2000;
147–53. 18(15):2817–27.
[12] Eubank WB, Mankoff DA, Vesselle HJ, et al. [26] Aristei C, Marsella AR, Chionne F, et al. Regional
Detection of locoregional and distant recurrences node failure in patients with four or more
in breast cancer patients by using FDG-PET. positive lymph nodes submitted to conservative
Radiographics 2002;22(1):5–17. surgery followed by radiotherapy to the breast.
[13] Eubank WB, Mankoff DA, Schmiedl UP, et al. Am J Clin Oncol 2000;23(3):217–21.
Imaging of oncologic patients: benefit of com- [27] Nikkanen TA. Recurrence of breast cancer. A re-
bined CT and [F-18]-fluorodeoxyglucose posi- trospective study of 569 cases in clinical Stages I–
tron emission tomogoraphy scan interpretation III. Acta Chir Scand 1981;147(4):239–45.
in the diagnosis of malignancy. Am J Roentgenol [28] Kiricuta IC, Willner J, Kolbl O, et al. The prog-
1998;171(4):1103–10. nostic significance of the supraclavicular lymph
[14] Yap CS, Seltzer MA, Schiepers C, et al. Impact of node metastases in breast cancer patients. Int J
whole-body 18FDG-PET on staging and manag- Radiat Oncol Biol Phys 1994;28(2):387–93.
ing patients with breast cancer: the referring [29] Iyer RB, Fenstermacher MJ, Libshitz HI. MR
physician’s perspective. J Nucl Med 2001;42(9): imaging of the treated brachial plexus. Am J
1334–7. Roentgenol 1996;167(1):225–9.
[15] Eubank WB, Mankoff DA, Bhattacharya M, et al. [30] Bagley FH, Walsh JW, Cady B, et al. Carcinoma-
Impact of [F-18]-Fluorodeoxyglucose PET on tous versus radiation-induced brachial plexus
defining the extent of disease and management neuropathy in breast cancer. Cancer 1978;41(6):
of patients with recurrent or metastatic breast 2154–7.
cancer. Am J Roentgenol 2004;83(2):479–86. [31] Toi M, Tanaka S, Bando M, et al. Outcome of
[16] Hortobagyi GN. Developments in chemotherapy surgical resection for chest wall recurrence in
of breast cancer. Cancer 2000;88(Suppl 12):3073–9. breast cancer patients. J Surg Oncol 1997;37(1):
[17] Taghian A, Jeong J, Mamounas E, et al. Patterns 853–63.
of locoregional failure in patients with operable [32] Hathaway CL, Rand RP, Moe R, et al. Salvage
FDG-PET Diagnosis 23

surgery for locally advanced and locally recurrent [46] Bellon JR, Livingston RB, Eubank WB, et al.
breast cancer. Arch Surg 1994;129(6):582–7. Evaluation of the internal mammary (IM) lymph
[33] Hathaway PB, Mankoff DA, Maravilla KR, et al. nodes by FDG-PET in locally advanced breast
The value of combined FDG-PET and magnetic cancer (LABC). Am J Clin Oncol 2004;27(4):
resonance imaging in the evaluation of suspected 407–10.
recurrent local-regional breast cancer: prelimi- [47] Bernstein V, Jones A, Mankoff DA, et al. Assess-
nary experience. Radiology 1998;210(3):807–14. ment of internal mammary lymph nodes by
[34] Ahmad A, Barrington S, Maisey M, et al. Use of fluorodeoxyglucose positron emission (FDG-
positron emission tomography in evaluation of PET) in medial hemisphere breast cancer [ab-
brachial plexopathy in breast cancer patients. Br J stract]. J Nucl Med 2000;41:289P.
Cancer 1999;79(3–4):478–82. [48] Vansteenkiste JF, Stroobants SG, De Leyn PR,
[35] Byrd DR, Dunnwald LK, Mankoff DA, et al. et al. Lymph node staging in non-small–cell lung
Internal mammary lymph node drainage pat- cancer with FDG-PET scan: a prospective study
terns with breast cancer documented by breast on 690 lymph node stations from 68 patients.
lymphoscintigraphy. J Surg Oncol 2001;8(3): J Clin Oncol 1998;16(6):2142–9.
234–40. [49] Scott WJ, Gobar LS, Terry JD, et al. Mediastinal
[36] Donegan WL. The influence of untreated internal lymph node staging of non-small–cell lung
mammary metastases upon the course of mam- cancer: a prospective comparison of computed
mary cancer. Cancer 1977;39(2):533–8. tomography and positron emission tomography.
[37] Veronesi U, Cascinelli N, Greco M, et al. Prog- J Thorac Cardiovasc Surg 1996;111(3):642–8.
nosis of breast cancer patients after mastectomy [50] Eubank WB, Mankoff DA, Takasugi J, et al.
18
and dissection of internal mammary nodes. Ann Fluorodeoxyglucose positron emission tomog-
Surg 1985;202(6):702–7. raphy to detect mediastinal or internal mammary
[38] Lacour J, Le M, Caceres E, et al. Radical mas- metastases in breast cancer. J Clin Oncol 2001;
tectomy versus radical mastectomy plus internal 19(15):3516–23.
mammary dissection. Ten year results of an in- [51] Coleman RE, Rubens RD. The clinical course of
ternational cooperative trial in breast cancer. bone metastases from breast cancer. Br J Cancer
Cancer 1983;51(10):1941–3. 1987;55(1):61–6.
[39] Noguchi M, Ohta N, Thomas M, et al. Risk of [52] Nielsen OS, Munro AJ, Tannock IF. Bone me-
internal mammary lymph node metastases and tastases: pathophysiology and management pol-
its prognostic value in breast cancer patients. icy. J Clin Oncol 1991;9(3):509–24.
J Surg Oncol 1993;52(1):26–30. [53] Ohta M, Tokuda Y, Suzuki Y, et al. Whole body
[40] Veronesi U, Marubini E, Del Vecchio M, et al. PET for the evaluation of bony metastases in
Local recurrences and distant metastases after patients with breast cancer: comparison with
conservative breast cancer treatments: partly 99Tcm-MDP bone scintigraphy. Nucl Med Com-
independent events. J Natl Cancer Inst 1995; mun 2001;22(8):875–9.
87(1):19–27. [54] Yang SN, Liang JA, Lin FJ, et al. Comparing
[41] Cody 3rd HS, Urban JA. Internal mammary node whole body [18F]-2-deoxyglucose positron emis-
status: a major prognosticator in axillary node- sion tomography and technetium-99m methy-
negative breast cancer. Ann Surg Oncol 1995; lene diphosphonate bone scan to detect bone
2(1):32–7. metastases in patients with breast cancer. J Can-
[42] Freedman GM, Fowble BL, Nicolaou N, et al. cer Res Clin Oncol 2002;128(6):325–8.
Should internal mammary lymph nodes in [55] Kao CH, Hsieh JF, Tsai SC, et al. Comparison and
breast cancer be a target for the radiation on- discrepancy of [18F]-2-deoxyglucose positron
cologist? Int J Radiat Oncol Biol Phys 2000;46(4): emission tomography and Tc-99m MDP bone
805–14. scan to detect bone metastases. Anticancer Res
[43] Ragaz J, Olivotto IA, Spinelli JJ, et al. Locoregional 2000;20(3B):2189–92.
radiation therapy in patients with high-risk [56] Cook GJ, Houston S, Rubens R, et al. Detection
breast cancer receiving adjuvant chemother- of bone metastases in breast cancer by 18FDG-
apy: 20-year results of the British Columbia ran- PET: differing metabolic activity in osteoblastic
domized trial. J Natl Cancer Inst 2005;97(2): and osteolytic lesions. J Clin Oncol 1998;16(10):
116–25. 3375–9.
[44] Schirrmeister H, Kuhn T, Guhlmann A, et al. [F- [57] Uematsu T, Yuen S, Yukisawa S, et al. Compari-
18]-2-deoxy-2-fluoro-D-glucose PET in the pre- son of FDG-PET and SPECT for detection of bone
operative staging of breast cancer: comparison metastases in breast cancer. Am J Roentgenol
with the standard staging procedures. Eur J Nucl 2005;84(4):1266–73.
Med 2001;28(3):351–8. [58] Schirrmeister H, Guhlmann A, Kotzerke J, et al.
[45] Greco M, Crippa F, Agresti R, et al. Axillary lymph Early detection and accurate description of extent
node staging in breast cancer by 2-fluoro-2-deoxy- of metastatic bone disease in breast cancer with
D-glucose-positron emission tomography: clinical fluoride ion and positron emission tomography.
evaluation and alternative management. J Natl J Clin Oncol 1999;17(8):2381–9.
Cancer Inst 2001;93(8):630–5. [59] Schirrmeister H, Glatting G, Hetzel J, et al.
24 Eubank

Prospective evaluation of the clinical value of morphological and functional imaging with
planar bone scans, SPECT, and [18]F-labeled FDG-PET/CT for restaging breast cancer—impact
NaF PET in newly diagnosed lung cancer. J Nucl on patient management [abstract]. J Nucl Med
Med 2001;42(12):1800–4. 2003;44:78P.
[60] Vranjesevic D, Filmont JE, Meta J, et al. Whole- [63] Tatsumi M, Cohade C, Mourtzikos KA, et al.
body 18F-FDG-PET and conventional imaging for Initial experience with FDG-PET-CT in the
predicting outcome in previously treated breast evaluation of breast cancer [abstract]. J Nucl
cancer patients. J Nucl Med 2002;43(3):325–9. Med 2003;44:394P.
[61] Pelosi E, Messa C, Sironi S, et al. Value of [64] Ciernik F, Dizendorf E, Baumert BG, et al.
integrated PET/CT for lesion localisation in can- Radiation treatment planning with an integrated
cer patients: a comparative study. Eur J Nucl Med positron emission and computer tomography
Mol Imaging 2004;31(7):932–9. (PET/CT): A feasibility study. Int J Radiat Oncol
[62] Buck AK, Wahl A, Eicher U, et al. Combined Biol Phys 2003;57(3):853–63.
25

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 25–32

Detection of Bone Metastases in


Breast Cancer by Positron
Emission Tomography
Holger Schirrmeister, MD

& F-18 fluoride positron emission & Summary


tomography & References
& F-18 fluorodeoxyglucose
& F-18 fluoride positron emission tomography
versus F-18 fluorodeoxyglucose-positron
emission tomography: which radiotracer
should be used in breast cancer?

Carcinoma of the breast is the most prevalent bution of bone metastases is a relevant prognostic
cancer in women in the United States and Western factor. Yamashita and colleagues [6,7] reported a
Europe, and is commonly associated with meta- significantly longer survival in patients who had
static bone disease. The incidence of breast cancer bone metastases exclusively to the upper regions
has been increasing in Western Europe and the of the skeleton than in patients who had metastases
United States, but the number of deaths resulting to the pelvis and the lower extremities. Further-
from breast cancer has remained relatively stable more, presence of osteoblastic features in bony
[1]. This is probably related to more effective treat- metastases was also reported to be associated with
ment strategies and to the detection of disease at prolonged survival [6,7].
early stages. It has been estimated that bone metastases be-
Although the incidence of bone metastases is as come visible on plain radiographs after 30% to
low as 1% to 2% at the time of primary diagnosis 50% of bone mineral loss [8]. Secondary formation
[2], bone metastases are found in one third of all of bone also occurs as response to osteolytic de-
patients who have recurrent disease [3]. In a study struction; hence, even osteolytic bone metastases
including 587 patients who died from breast can- can be detected with bone scintigraphy several
cer [4], 69% had radiological evidence of skeletal months before they are apparent on plain radio-
metastases before death. There are two different graphs [9,10]. Bone scintigraphy is not perfect in
types of bony metastases—osteolytic and osteoblas- detecting bone metastases, however; compara-
tic disease—depending on their radiographic ap- tive studies with MRI revealed significantly more
pearance. Patients who have breast cancer can vertebral metastases not detected on conventional
have either osteolytic or osteoblastic metastases, or planar bone scintigraphy [11,12]. Nevertheless,
can present with mixed lesions containing both conventional bone scintigraphy remains the most
features [5]. Interestingly, patients who have bone suitable technique for whole-body screening, be-
metastases only survive 2 to 3 years longer than cause whole-body MRI is currently not practical
patients who have visceral involvement. The distri- for routine whole-body surveys, and its sensitivity

Clinic of Nuclear Medicine, University of Kiel, Arnold-Heller Str. 9, Kiel 24105, Germany
E-mail address: hschirrmeister@nuc-med.uni-kiel.de

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.005
pet.theclinics.com
26 Schirrmeister

in detecting extra-vertebral metastases is not yet bone scintigraphy ranged between 80% and 90% in
sufficiently assessed. the skull, thorax, and the extremities. The most
Positron emission tomography (PET) is a rela- probable explanation for these findings is that the
tively new imaging technique for noninvasive as- detection of vertebral bone metastases was limited
sessment of several metabolic pathways. During the by superimposed tracer retention in soft tissue and
last decade, this method evolved from a research other parts of the skeleton. This drawback of con-
tool to a standard imaging modality for routine ventional gamma camera imaging is generally not
staging of several types of malignant tumors. PET present with tomographic imaging. Hence, in con-
using the glucose analog 2-(fluoro-18)-2-deoxy-D- trast to conventional bone scintigraphy, the sensi-
glucose (FDG) enables detection of primary breast tivity of F-18 fluoride PET was independent from
cancer and its metastases on the basis of increased the anatomical localization of skeletal lesions [16].
glucose metabolism in neoplastic tissues. Char- In another prospective study [17], F-18 fluoride
acterization of bone metastases is also possible PET was performed in 34 patients who had breast
with F-18 sodium fluoride, which shows ostoblastic cancer and high risk of metastatic bone disease.
bone reaction. The potential of PET with FDG F-18 fluoride PET revealed more metastases than
and F-18 fluoride in detecting bone metastases conventional bone scintigraphy in 11 of 17 patients
originating from breast cancer is discussed in who had bony metastases. Three patients were true-
this article. positive on F-18 fluoride PET, but had no signs of
metastatic bone disease on normal bone scintigra-
phy [Figs. 1, 2]. In another patient who had known
F-18 fluoride positron emission tomography metastases, an osteolytic lesion was depicted with
Skeletal uptake of the positron emitter F-18 fluo- F-18 fluoride PET that had been missed by bone
ride is based on the exchange of hydroxyl ions in
the hydroxyapatite crystal. Hence, the uptake of
F-18 fluoride is an indicator of bone mineraliza-
tion [13]. F-18 fluoride was introduced into clini-
cal practice by Blau in 1962 [14]. Subsequently,
F-18 fluoride was approved by the Food and
Drug Administration (FDA) as a radiotracer for
bone scintigraphy. One decade later, this radio-
pharmaceutical was replaced by Tc-99m-labeled
polyphosphonates, because conventional gamma
cameras performed better in terms of sensitivity
and spatial resolution of the 140 keV photons
from Tc-99m compared with the 511 keV photons
from F-18 fluoride.
In comparing the pharmacokinetics of Tc-99m
labeled polyphosphonates and F-18 fluoride, it is
important to note that the uptake of F-18 fluoride
into bone is approximately twofold higher and the
blood clearance is significantly faster compared
with technetium labeled polyphosphonates. This
leads to an increased bone-to-background ratio
for F-18 fluoride. The improved sensitivity and bet-
ter spatial resolution of modern PET scanners allow
performing highly sensitive and specific whole-
body screening for bone metastases [15–19]. In a
pilot study [16], F-18 fluoride PET revealed two
times more bone metastases than bone scintigra-
phy in patients who had osteolytic disease from
thyroid cancer or osteoblastic metastases from pros-
tate cancer. An important finding in this study
was that the sensitivity of planar bone scintigra-
phy was highly variable, depending on the skele- Fig. 1. Normal bone scintigraphy in a patient with
tal region. The sensitivity of bone scintigraphy was breast cancer (pT2,N0). Bone scintigraphy was per-
as low as 20% in the thoracic spine and 40% formed because of increasing serum alkaline phospha-
in the lumbar spine; in contrast, the sensitivity of tase and calcium levels.
PEM Detection of Breast Cancer Bone Metastases 27

Fig. 2. F-18 fluoride PET of the same patient as presented in Fig. 1. PET imaging was obtained 2 hours after
injection of 370 MBq F-18 fluoride. Two osteolytic metastases are present in the thoracic and lumbar spine.
Maximum intensity projection images (left). Sagittal sections of the thoracic and lumbar spine (middle). Corre-
sponding sagittal CT images (right).

scintigraphy. Because of the risk for pathologic lesions and a better differentiation between benign
bone fracture, this metastasis was surgically stabi- [22] and malignant lesions [23] compared with
lized. In this highly selected series, clinical manage- conventional gamma cameras.
ment was influenced in 4 out of 34 patients by the In breast cancer, both osteolytic and osteoblastic
results from F-18 fluoride PET. bone metastases have been reported to demon-
Along with the twofold higher sensitivity in de- strate focally increased F-18 fluoride uptake [15].
tecting bone metastases, there is also a twofold In the experience of the author’s group, osteolytic
higher rate of benign lesions visible on F-18 fluo- metastases were often characterized as photopenic
ride PET [16]. In a recent review article [20], it was lesions surrounded by a rim of increased activity
assumed that this would lead to an increased rate of [Fig. 3]. This pattern was observed exclusively
false-positive results. This assumption, however, is in osteolytic metastases and not in benign le-
not supported by the current literature, in which sions [23].
the specificity of F-18 fluoride PET was 90% and Both osteolytic metastases and benign lesions
higher in several studies [16–19]. appear as regions with focally increased tracer
It is important to note that uptake of F-18 fluo- uptake. Therefore, knowledge of the typical appear-
ride is not tumor-specific, and that numerous ance of benign lesions becomes crucial for differ-
benign lesions are detected by F-18 fluoride PET. entiation between benign lesions and metastases.
Differentiation between benign and malignant Arthritis of the articular facets and endplate frac-
bony lesions is not possible by means of quantita- tures account for approximately 80% of all benign
tive analysis of bone metabolism [21]. Neverthe- abnormalities detected with F-18 fluoride PET [22].
less, the superior spatial resolution of modern PET These abnormalities have a very characteristic F-18
scanners allows for exact anatomical localization of fluoride uptake pattern [Fig. 4]. Osteophytes are

Fig. 3. F-18 fluoride PET. Typical pattern of osteolytic (left) and osteoblastic metastases (right).
28 Schirrmeister

Fig. 4. F-18 fluoride PET. Typical pattern of benign lesions.

characterized as lesions with variable tracer uptake was approximately sevenfold higher in osteolytic
located at the superior and inferior edges of adja- bone metastases compared with osteoblastic me-
cent endplates, whereas increased F-18 fluoride tastases [Figs. 5, 6]. In the entire patient group,
uptake at adjacent surfaces of intervertebral joints FDG-PET detected more bone metastases than
is typical for intervertebral arthritis. On F-18 fluo- bone scintigraphy. Bone scintigraphy, however,
ride PET, lesions neither located at joint surfaces was more sensitive in a subgroup of patients who
nor showing the typical pattern of endplate frac- had predominantly osteoblastic disease.
tures, osteophytes, or serial rib fractures are suspi- Ohta and colleagues [25] compared the accuracy
cious for metastatic disease [23]. of conventional bone scintigraphy with FDG-PET
in 51 breast cancer patients. In this series, the sen-
sitivity and specificity of FDG-PET were 77.7% and
97.6%, respectively, compared with 77.7% and
F-18 fluorodeoxyglucose
80.3% for bone scintigraphy. A similar sensitivity
The glucose analog 2-(fluoro-18)-2-deoxy-D-glucose and an even higher specificity were found for FDG-
(FDG) is currently the most commonly used radio- PET compared with bone scintigraphy in a series of
tracer in oncologic PET imaging. This surro- 48 patients who had biopsy-proven breast cancer
gate marker for glucose metabolism enters the cell and suspected bone metastases [26]. Comparable
via the glucose transporter membrane proteins results for FDG-PET and bone scintigraphy were
Glut-1 and Glut-5. FDG is then posphorylated by also reported in a series of 24 breast cancer patients
the enzyme hexokinase. In contrast to glucose,
FDG-6-phospate is not a substrate for the glucose-
6-phophate isomerase, and is therefore trapped
within tumor cells.
Although the exact uptake mechanism of FDG in
bone metastases is not yet known, it is assumed
that FDG is taken up directly into the tumor tissue
of skeletal metastases and not into the surrounding
remodeled bone. There are only a few reports
addressing the accuracy of FDG-PET for detecting
skeletal metastases of breast cancer. This is proba-
bly related to the low incidence of bone metastases
at initial diagnosis, and the absence of a reliable
gold standard.
In a series of 23 breast cancer patients who had
metastatic bone disease, Cook and coworkers [24]
evaluated FDG-PET in osteoblastic and osteolytic Fig. 5. FDG-PET in a patient with recurrent breast
bone metastases by semiquantitative standardized cancer presenting with increased FDG uptake in a
uptake values (SUV). In this series, the FDG uptake small osteolytic metastasis (L-4).
PEM Detection of Breast Cancer Bone Metastases 29

Fig. 6. FDG-PET in a patient with recurrent breast cancer presenting with multiple osteoblastic bone metastases.
Intense increased FDG uptake is present in a liver metastasis, whereas the bone metastases show only little
FDG uptake.

by Kao and coworkers [27]. In their study, FDG-


F-18 fluoride positron emission tomography
PET was less sensitive but significantly more speci-
versus F-18 fluorodeoxyglucose-positron
fic than bone scintigraphy.
emission tomography: which radiotracer
One limitation of the studies described above is
should be used in breast cancer?
that planer bone scintigraphy was compared with
FDG-PET tomographic images. In general, tomo- A prospective comparison of FDG-PET and F-18
graphic imaging methods are more sensitive than fluoride PET in detecting bone metastases from
planar imaging, because lesions are detectable with- breast cancer has not yet been published. A re-
out superposition by other anatomical structures. cent study [19] evaluated the accuracy and cost-
For example, small lung metastases are seen on CT effectiveness of F-18 fluoride PET in 103 patients
but frequently not on plain radiographs, and ver- who had initial diagnosis of lung cancer. In this
tebral bone metastases are seen on single photon study, FDG-PET was performed for whole-body
emission computed tomography (SPECT), CT, or staging in 41 patients, 10 of whom had bone me-
MRI, despite negative planar bone scintigraphy or tastases. One of these patients had disseminated
plain radiographs. Accordingly, Kosuda and col- bone metastases, but was negative on FDG-PET.
leagues [28] demonstrated a significantly improved Furthermore, the number of bone metastases was
sensitivity for detection of bone metastases by underestimated in an additional 4 patients. The
SPECT imaging compared with planar bone scintig- results of this study are in line with the results of
raphy. The improved sensitivity of bone scintigra- the study reported by Uesuda and coworkers [30],
phy by using SPECT imaging was also found in a who compared bone SPECT and FDG-PET. Both
mixed series of 174 patients by Han and coworkers studies indicated a significantly better sensitivity by
[29], and in another prospective series of 103 using bone-specific radiotracers in combination
patients who had lung cancer [19]. with tomographic imaging (SPECT or PET). An in-
The first study comparing bone SPECT and FDG- teresting finding of that study was that 2 patients
PET was recently published by Uematsu and co- who had highly suspicious vertebral metastases on
workers [30]. In this study, 15 patients who had MRI were negative on F-18 fluoride PET. Skeletal
known metastatic bone disease were included. CT, metastases were ruled out by clinical follow-up in
MRI, and the subsequent clinical follow-up were 1 patient and by biopsy in the other patient. F-18
used as references. In a lesion-by-lesion analysis, fluoride PET might therefore become the first imag-
the sensitivity of SPECT was 95%, and only 17% for ing method that is sensitive and specific enough for
FDG-PET. The specificity was not statistically sig- confirmation of suspicious lesions that are seen
nificantly different between both imaging methods on MRI, but are negative on currently available
(SPECT 99%, PET 100%). conventional imaging methods.
The results reported by Cook and colleagues [24] One important advantage of FDG over F-18 fluo-
and the studies comparing bone scintigraphy with ride is that the FDG uptake is not limited to bone
FDG-PET [25–27,30] indicate that FDG-PET might metabolism. Several studies have shown that FDG-
not be more sensitive than bone scintigraphy but PET reliably detects primary tumors [31–36], and is
is probably more specific. Because FDG-PET and currently the most sensitive imaging technique for
bone scintigraphy have a different sensitivity for noninvasive assessment of axillary lymph nodes
detecting oteoblastic and osteolytic bone metas- and visceral metastases. Furthermore, Eubank and
tases, the imaging modalities should be regarded colleagues [37] have shown that FDG-PET is accu-
as complementary, but cannot replace each other. rate in evaluating the internal lymph nodes. FDG
30 Schirrmeister

Fig. 7. FDG-PET (left) and bone scintigraphy (right) was performed for restaging in a patient with recurrent
breast cancer. FDG-PET shows multiple lymph node metastases but fewer bone metastases compared with
bone scintigraphy.

PET was superior to conventional imaging and as F-18 fluoride is not limited to malignant bone
sensitive as MRI in the differentiation between uni- lesions. In turn, F-18 PET fluoride requires specific
focal and multifocal disease [38]. In a series of 117 experience with this method. This limitation might
patients who had suspected breast cancer [36], be overcome by using coregistered PET and CT im-
whole-body FDG-PET was as accurate as the cur- ages (PET/CT). Recently Even-Sapir and coworkers
rently employed imaging methods in identification [39] found a significantly higher accuracy of F-18
of the primary tumor, and significantly more accu- fluoride PET/CT compared with F-18 fluoride PET
rate in the detection of lymph node and distant in a mixed series of 44 cancer patients.
metastases. In that series, only 2 patients had bone
metastases. Both were true-positive on both bone
Summary
scintigraphy and FDG-PET. Because of the low inci-
dence of bone metastases, a potentially lower sen- FDG-PET might be less sensitive than F-18 fluoride
sitivity in detecting osteoblastic bone metastases PET in detecting bone metastases from breast can-
with FDG-PET seems less relevant at initial staging. cer, but FDG-PET allows accurate assessment of the
Regarding the specificity in evaluation of the primary tumor, lymph nodes, and visceral metas-
skeleton, there are some rare benign bone tumors tases in one single imaging study. The uptake of
and inflammatory lesions that demonstrate in- FDG is more specific for malignant lesions than
creased FDG uptake. In contrast, the uptake of bone metabolic radiotracers. Hence, FDG-PET in

Fig. 8. F-18 fluoride PET of the same patient as presented in Fig. 7. F-18 fluoride PET was performed because
the patient complained of neck stiffness and a suspicious lesion was seen on MRI in the second cervical vertebral
body. The F-18 fluoride PET scan confirms the bone metastasis by intense increased bone metabolism and reveals
an additional, previously unsuspected, bone metastasis in first cervical vertebral body (arrows).
PEM Detection of Breast Cancer Bone Metastases 31

combination with bone scintigraphy might become et al. Skeletal metastases from breast cancer:
the standard of care for staging and restaging of uptake of F-18 fluoride measured with positron
the skeleton in breast cancer. F-18 fluoride PET is emission tomography in correlation with CT.
currently the most sensitive whole-body imaging Skeletal Radiol 1998;27(2):72–6.
modality for bone metastases, but is associated [16] Schirrmeister H, Guhlmann CA, Elsner K, et al.
Planar bone imaging vs. 18F-PET in patients with
with a significant learning curve. In breast cancer,
cancer of the prostate, thyroid and lung. J Nucl
the the use of F-18 fluoride PET should be based Med 1999;40(10):1623–9.
on individual cases [Figs. 7, 8] depending on the [17] Schirrmeister H, Guhlmann CA, Kotzerke J, et al.
expected change of therapy management. Early detection and accurate description of ex-
tent of metastatic bone disease in breast cancer
with 18F-fluoride ion and positron emission to-
References mograpy. J Clin Onol 1999;17(8):2381–9.
[1] Newcomb PA, Lantz PM. Recent trends in breast [18] Schirrmeister H, Glatting G, Hetzel J, et al. Pro-
cancer incidence, mortality, and mammography. spective evaluation of the clinical value of pla-
Breast Cancer Res Treat 1993;28(2):97–106. nar bone scans, SPECT and F-18 labelled NaF
[2] Perez DJ, Powles TJ, Milan J, et al. Detection of PET in newly diagnosed lung cancer. J Nucl Med
breast carcinoma metastases in bone: relative 2001;42(12):1800–4.
merits of x-rays and skeletal scintigraphy in breast [19] Hetzel M, Arslandemir C, König HH, et al. F-18
cancer. Lancet 1983;10;2(8350):613–6. NaF PET for detection of bone metastases in lung
[3] Kunkler IH, Merrick MV. The value of non- cancer. Accuracy, cost-effectiveness, and impact
staging skeletal scintigraphy. Clin Radiol 1986; on patient management. J Bone Miner Res 2003;
37(6):561–2. 18(12):2206–14.
[4] Coleman RE, Rubens RD. the clinical course of [20] Buscombe JR, Holloway B, Roche N, et al.
bone metastases from breast cancer. Br J Cancer Position of nuclear medicine modalities in the
1987;55:61–6. diagnostic work-up of breast cancer. Q J Nucl
[5] Hortobagyi GN. Bone metastases in breast cancer Med Mol Imaging 2004;48(2):109–18.
patients. Semin Oncol 1991;18(Suppl 5):11–5. [21] Hoh CK, Hawkins RA, Dahlbom M, et al. Whole
[6] Yamashita K, Koyama H, Inaji H. Prognostic sig- body skeletal imaging with 18F fluoride ion and
nificance of bone metastasis from breast cancer. PET. J Comput Assist Tomogr 1993;17(1):34–41.
Clin Orthop 1995;312:89–94. [22] Schirrmeister H, Diederichs CG, Rentschler M,
[7] Yamashita K, Ueda T, Komatsubara Y, et al. et al. Die Positronenemissionstomographie des
Breast cancer with bone-only metastases. Visceral Skelettsystems mit 18FNa: Häufigkeit, Befund-
metastases-free rate in relation to anatomic dis- muster und Verteilung benigner Veränderungen
tribution of bone metastases. Cancer 1991;68(3): [Positron-emission tomography of the skeletal sys-
634–7. tem using 18FNa: the incidence, pattern of the find-
[8] Bachman AS, Sproul EE. Correlation of radio- ings and distribution of benign changes]. Fortschr
graphic and autopsy findings in suspected me- Röntgenstr 1998;169(3):310–4 [in German].
tastases in the spine. Bull N Y Acad Med 1940; [23] Schirrmeister H, Guhlmann CA, Elsner K, et al.
44:169–75. Die Positronenemissionstomographie des Ske-
[9] Silberstein ER, Saenger EL, Tofe AJ, et al. Imaging lettsystems mit 18FNa: Häufigkeit, Befundmuster
of bone metastases with 99mTc-Sn-HEDP (diphos- und Verteilung von Skelettmetastasen [Positron
phonate), 18F and skeletal radiography. Radi- emission tomography of the skeletal system
ology 1973;107:551–5. using 18FNa: frequency, distribution and appear-
[10] Green D, Jeremy R, Towson J, et al. The role of ance of skeletal metastases]. Rontgenpraxis 1999;
fluorine 18 scanning in the detection of skeletal 52(1):19–25 [in German].
metastases in early breast cancer. Aust N Z J Surg [24] Cook GJ, Houston S, Rubens R, et al. Detection
1973;43:251–4. of bone metastases in breast cancer by 18FDG
[11] Frank JA, Ling A, Patronas NJ, et al. Detection of PET: differing metabolic activity in osteoblastic
malignant bone tumors: MR imaging vs scin- and osteolytic lesions. J Clin Oncol 1998;16(10):
tigraphy. AJR Am J Roentgenol 1990;155:1043–8. 3375–9.
[12] Gosfield E, Alavi A, Kneeland B. Comparison of [25] Ohta M, Tokuda Y, Suzuki Y, et al. Whole body
radionuclide bone scans and magnetic resonance PET for the evaluation of bony metastases in
imaging in detecting spinal metastases. J Nucl patients with breast cancer: comparison with
Med 1993;34:2191–8. 99mTc-MDP bone scintigraphy. Nucl Med Com-
[13] Hawkins RA, Choi Y, Huang SC, et al. Evaluation mun 2001;22(8):875–9.
of skeletal kinetics of fluorine 18-fluoride ion [26] Yang SN, Liang JA, Lin FJ, et al. Comparing
with PET. J Nucl Med 1992;33:633–64. whole body 18F–2-deoxyglucose positron emis-
[14] Blau M, Nagler W, Bender MA. Fluorine-18 a new sion tomography and technetium-99m methy-
isotope for bone scanning. J Nucl Med 1962;3: lene diphophonate bone scan to detect bone
332–4. metastases in patients with breast cancer.
[15] Petren-Mallmin M, Andreasson I, Ljunggren O, J Cancer Res Clin Oncol 2002;128(6):325–8.
32 Schirrmeister

[27] Kao CH, Hsieh JF, Tsai SC, et al. Comparison and staging of the axilla in breast cancer. J Nucl Med
discrepancy of 18F-deoxyglucose-positron emis- 1998;39(1):4–8.
sion tomography and Tc-99m MDP bone scan to [34] Moon DH, Maddahi J, Silverman DHS, et al.
detect bone metastases. Anticancer Res 2000; Accuracy of whole-body fluorine-18 FDG PET for
20(3B):2189–92. the detection of recurrent or metastatic breast
[28] Kosuda S, Kaji T, Yokoyama H, et al. Does bone carcinoma. J Nucl Med 1998;39(3):431–5.
SPECT actually have lower sensitivity for detect- [35] Eubank WB, Mankoff DA, Vesselle HJ, et al. De-
ing vertebral metastasis than MRI? J Nucl Med tection of locoregional and distant recurrences
1996;37(6):975–8. in breast cancer patients by using FDG PET.
[29] Han LJ, Au-Yong TK, Tong WC, et al. Compari- Radiographics 2002;22(1):5–17.
son of bone single-photon emission tomogra- [36] Schirrmeister H, Kuhn T, Guhlmann A, et al.
phy and planar imaging in the detection of Fluorine-18 2-deoxy-2-fluoro-D-glucose PET in
vertebral metastases in patients with back pain. the preoperative staging of breast cancer: com-
Eur J Nucl Med 1998;25(6):635–8. parison with the standard staging procedures.
[30] Uematsu T, Yuen S, Yukisawa S, et al. Compari- Eur J Nucl Med 2001;28(3):351–8.
son of FDG PET and SPECT for detection of [37] Eubank WB, Mankoff DA, Takasugi J, et al.
bone metastases in breast cancer. AJR Am J 18fluorodeoxyglucose positron emission tomog-
Roentgenol 2005;184(4):1266–73. raphy to detect mediastinal or internal mam-
[31] Adler LP, Crowe JP, Al-Kaisi NK, et al. Evaluation mary metastases in breast cancer. J Clin Oncol
of breast masses and axillary lymph nodes with 2001;19(15):3516–23.
F-18 2-deoxy-3-fluoro-D-glucose. Radiology [38] Rieber A, Schirrmeister H, Gabelmann A, et al.
1993;187(3):743–50. Pre-operative staging of invasive breast cancer
[32] Avril N, Dose J, Jänicke F, et al. Assessment with MR mammography and/or PET: boon or
of axillary lymph node involvment in breast bunk? Br J Radiol 2002;75(898):789–98.
cancer patients with positron emission tomogra- [39] Even-Sapir E, Metser U, Flusser G, et al. Assess-
phy using radiolabeled 2-(fluorine-18)-deoxy- ment of malignant skeletal disease: initial experi-
2-fluoro-D-glucose. J Natl Cancer Inst 1996; ence with 18F-fluoride PET/CT and comparison
88(17):1204–9. between 18F-fluoride PET and 18F-fluoride PET/
[33] Crippa F, Agresti R, Seregni E, et al. Prospective CT. J Nucl Med 2004;45(2):272–8.
evaluation of fluorine-18-FDG PET in presurgical
33

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 33–38

Instrumentation for Positron


Emission Mammography
Christopher J. Thompson, DSc FCCPM

& Feasibility and promise of early & Recent clinical findings


instruments & Future prospects for positron emission
& Evolution of positron emission mammography
mammography instruments & Acknowledgments
& Early clinical findings & References

The use of positron emission tomography (PET) the parent nucleus before they lose energy and
to provide images of the glucose metabolism of annihilate with an electron in tissue. Another limi-
malignant disease is becoming more and more tation is the noncollinearity of the pair of gamma
widespread, and whole body PET (WB-PET) images rays that do not travel away from the point of
are now often used to choose appropriate therapy annihilation at exactly 180°, because of the energy
for cancer patients. The first use of F-18 fluorodeoxy- of the electron at the time of annihilation. Al-
glucose-positron emission tomography (FDG-PET) though this appears as a fundamental limit in con-
for the study of breast cancer was reported by Wahl ventional PET, it is much less of a problem in an
and colleagues [1], and since then many other re- instrument that has its detectors much closer to
ports have been made [2,3], to cite the first few only. each other than the separation needed in WB-
When combined with radiograph CT, PET images PET. The major instrumental limitation is the size
provide the equivalent of a ‘‘metabolic contrast of the detectors. Making the detectors smaller
agent,’’ which serves to highlight the abnormal glu- improves the resolution at the expense of increased
cose metabolism in tumors. Combined PET/CT complexity and resulting cost; however, if detectors
scanners now outsell conventional PET scanners, are placed closer, fewer detectors are required, so
and they have become the standard of care in instruments made especially for breast imaging can
many centers. be made to provide higher spatial resolution than
PET scanners have a limited spatial resolution those from WB-PET. The factors that limit spatial
compared with structural imaging modalities such resolution (SR) in PET can be combined as the sum
as CT and MRI. Because the identification of small of independent variables in the formula:
tumors leads to earlier diagnosis and treatment,
much effort has gone into trying to improve the
spatial resolution of PET. There are both instru- ðSRÞ2 ¼ R*½PR 2 þ ð0:005*DSÞ2 þ ðCW=2Þ2 þ BE2 
mental and fundamental factors that degrade spa-
tial resolution in PET. The fundamental limit is In this equation, R is a factor that relates to the
caused by the distance positrons move away from reconstruction method and filter, PR is the effective

The development and evaluation of the PEM-1 scanner was funded by grants from the National Cancer
Institute of Canada’s Canadian Breast Cancer Research Initiative #6139 and #9232 in 1997 and 1999.
Departments of Neurology and Neurosurgery, Medical Physics, and Biomedical Engineering, Suite #798,
Montréal Neurological Institute, McGill University, 3801 University Street, Montreal, Québec, Canada H3A 2B4
E-mail address: Christopher.Thompson@McGill.Ca

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.004
pet.theclinics.com
34 Thompson

positron range in tissue 0.005 = 1/2*tan(θ/2), θ is x-ray


the mean angle of noncollinearity, DS is the detec- source
co-registration
tor separation, CW is the crystal width, and BE tool
combines the effects of light sharing and under- upper
detector
sampling of the image.
One of the design factors in all PET scanners is
a trade-off between spatial resolution, sensitivity, compression
and cost. In the equation above, the factor CW compression breast
paddle
is the one that balances the cost versus the spa-

45 cm
tial resolution, and the one over which scanner
manufacturers have the most control. In WB-PET, lower
moving the detectors closer together is not possi- magnification detector
table
ble, because the patient would no longer fit in
the scanner; however, there is an option for This side faces
doing this in instruments designed exclusively for chest wall rails
breast imaging. x-ray film cassette
The idea of a dedicated PET scanner for breast holder
imaging was first proposed by Weinberg in 1993, in 45 cm
a US patent application [4], and a successful pro- Fig. 1. Layout of PEM-1 breast imaging system. The
posal for a small business incentive for research two detectors, mounted on rails, are moved out of the
(SBIR) grant from the US National Institutes of radiographic field for conventional mammography,
Health (NIH). and back over and under the breast during the acqui-
sition of a metabolic image.

Feasibility and promise of early instruments vious one. This paper provided the basic estimate
Through a collaboration with Weinberg, the first of both the signal-to-noise ratio and count-rate that
experiments to examine the concept were published could be expected from a clinical PEM instrument.
in 1993 [5], and the name ‘‘positron emission mam- Encouraged by these results, the author’s group
mography’’ (PEM) was coined to represent this applied for funding from the Canadian Breast
technique. The concept was to place two planar Cancer Research Initiative. We then built and per-
detectors capable of detecting the 511 keV annihi- formed a preliminary clinical trail of an instrument
lation photons in a conventional mammography known as ‘‘PEM-1.’’ Because we had concluded that
unit. Placing the breast on a ‘‘magnification table’’ back-projection images were sufficient to identify
sometimes used in these instruments provides regions of higher-than-surrounding uptake, we did
the possibility of having one detector between the not perform any sophisticated reconstruction, pre-
radiograph tube and the compression plate, and ferring to opt for an almost real-time image display
another between the lower aspect of the breast of the PEM image. The goal we set was to perform
and the Radiograph sensor. The two detectors the clinical trial using only 75 MBq (2 mCi) of
move out of the radiograph field for conventional FDG, and an imaging time of 2 minutes per breast,
mammography, and move back over and under because this is about the time it takes to develop a
the breast for the PEM acquisition [6]. This concept radiograph film in an automatic film processor.
predates PET/CT by several years [7], but the goal
was very much the same as that of PET/CT as it
Evolution of positron emission
has evolved today: to provide a coregistered anato-
mammography instruments
mical and functional image in the same procedure
with minimal movement of the patient. The de- When performing WB-PET scans, it is a simple
sign of the instrument is illustrated in Fig 1. matter to overscan the regions most likely to harbor
An important finding of this first PEM paper [5] metastases, and to overlap the bed positions to
was that a small hyperactive region was just as compensate for the reduced sensitivity toward the
visible in a superposition of a few near vertical pro- axial ends of each set of slices caused by the fall-off
jections as it was in fully reconstructed tomo- in three dimensional (3D) sensitivity in the scan-
graphic images. The experiments were performed ner. When imaging the breast with PET detectors in
in a 15-slice brain scanner on a box phantom con- a mammographic configuration, this is not possi-
taining four tubes of various sizes with either no ble, as illustrated in Fig. 2. Even when the detectors
activity or 9.3 times the background. The images are placed very close to the chest wall, some shield-
were made over different times, so that each con- ing is required. This is a serious problem when
secutive image contains half the counts of the pre- imaging small breasts, and for investigations close
Instrumentation for PEM 35

to the chest wall. PET imaging in the 3D mode is


always less sensitive toward the axial ends of the
field of view, because fewer detector pairs can be in
coincidence in these regions.
A variety of different PEM geometries have been
proposed, and several of these are currently in use
[Fig. 3]. Almost all have a larger field of view than
the first prototype. Complete coverage of the breast
while the breast remains in place has become
the norm. Fig. 3 shows the detector arrangement
in each device. Part A is the classic two-parallel
crystal arrays coupled to position-sensitive photo- Fig. 3. Four geometries used or proposed for PEM.
multipliers (PS-PMTs). One detector is moveable to (A) Two small planar detectors with variable separa-
allow positioning the breast and for variable com- tion. (B) Two multi-element detectors that move inside
pression to suit the patient’s anatomy. This geome- an enclosure during imaging. (C ) Rectangular box
try was first used in the PEM-1 scanner [8–12], and structure with moveable detectors. (D) Cylindrical crys-
later by Smith and coworkers of the Thomas Jeffer- tal array surrounding pendulous breast.
son Laboratory [13]. Part B shows a pair of linear
arrays of detectors that scan across the breast dur- allow a more complete reconstruction of the activ-
ing the examination [14]. This technique is used ity within the breast. This is the system proposed by
in the only commercially available PEM scanner Huber and colleagues [15] of the Berkeley National
at present, the PEM Flex from Naviscan PET Sys- Laboratory in Berkeley, California. A similar con-
tems in Rockville, Maryland. Part C is a boxlike cept encloses the breast in a small circular array
detector array that surrounds the breast, and should of detectors with the breast pendant through the
hole [16].
All of these instruments use very finely pixilated
detectors and a compact geometry designed to
reduce the blurring associated with the noncollin-
earity of the annihilation photons. Of special in-
terest is the proposal by Huber and coworkers [15],
which also encodes the depth at which each
gamma ray is detected in the crystal. This allows
for a very compact geometry while avoiding the
blurring associated with very oblique gamma ray
penetration of the detector.

Early clinical findings


In 1999, the author’s group published the results of
the first clinical trial of any PEM instrument [12].
During the clinical trial of the PEM-1 scanner, we
studied 14 patients, 10 of whom had various breast
cancers confirmed by pathological investigation of
the surgically excised specimens. Only 5 of these
had a clearly focal uptake (with a mean contrast of
5.8:1 with respect to the surrounding breast tissue).
Three other patients were considered PEM-positive
on the basis of a significant count-rate asymmetry,
Fig. 2. Geometry for conventional radiographic mam- after accounting for factors like isotope decay and
mography and PEM using two planar detectors. The volume of breast tissue in the field of view, and de-
radiograph beam is centered over the edge of the
tector separation.
image sensor near the chest wall, and collimated to
As an example of a typical patient from that
expose only the compressed breast. The response of
the sensor to the radiograph beam (plotted below the study, consider the case of a 75-year-old lady
geometry) is essentially constant over the entire sur- who presented with a suspicious mammogram of
face. In contrast, the response of the PEM detectors is her right breast, and was a candidate for lumpec-
triangular, with its peak in the geometric center of the tomy. She had a cranio-caudal (CC) PEM scan of
detector pair. her right breast starting 60 minutes after the admin-
36 Thompson

Fig. 4. PEM-1 images showing focal uptake in a 1.5 cm inter-ductal carcinoma (upper row) compared with normal
uptake in the contra-lateral breast (lower row). These images were scaled to the equivalent counting time and
isotope decay, showing an asymmetry in the overall uptake.

istration of 75 MBq of 18F-FDG. A CC scan of the The correlation between the PEM and mammo-
left breast was then acquired starting 70 minutes gram is illustrated in Fig. 5. The PEM image is first
postinjection. She underwent a right-breast seg- ‘‘windowed’’ such that only the suspicious region is
mental mastectomy 4 days after the PEM study. visible. The mammogram, following digitization
The pathologist’s report described a 1.5 × 1.5 × with a video frame digitizer, is then displayed.
1.3 cm intraductal carcinoma and infiltrating duc- The images are rescaled and fused by aligning the
tal carcinoma, with a histological grade of 3 on the top-right and bottom-left corners of a digital repre-
Bloom and Richardson scale. The appearance of the sentation of a wire grid with its image, which is
PEM images is shown in Fig. 4. Seven images are visible on the mammogram. A fused image is made
presented for each breast, with the right breast by displaying alternate pixels from the mammo-
displayed on the top row. The images are displayed gram and scaled PEM image in differing color
as if the breast were pendant, so that the chest wall scales. This technique is now used to display PET/
would be above the images presented. The left- CT images, although the image fusion software of
most image corresponds with the upper aspect the author’s group predates PET/CT.
of the breast on the CC mammogram. The focal
activity visible near the top (chest wall) in the
Recent clinical findings
upper row is most intense in image 4, but extends
further toward the upper breast surface. The lower The first report of clinical results from PEM Flex was
row of images from the normal breast is more published by Weinberg and coworkers earlier this
uniform, and was read as normal. year [14]. They reported on 94 cases performed at

Fig. 5. Correlation of the intensity-thresholded PEM-1 image and a hyperdense region of the mammogram using
the coregistration technique developed for this instrument. The field of view of the mammogram is bigger than
that of the PEM-1 detectors, and in the active PEM region, alternate pixels are assigned to PEM and radiograph
images in a manner identical to that commonly used for PET/CT image display.
Instrumentation for PEM 37

perhaps the most relevant to PEM. The technology


assessment report from the Blue Cross and Blue
Shield Association [17], which forms the basis of
the review of the utility of FDG-PET in breast can-
cer, contains over 70 references on the use of
PET and single photon imaging agents such as
99m
Tc sestamibi in the detection and staging of
breast cancer.
The major limitations of early PEM instruments
were their small field of view and reduced sensi-
tivity near the chest wall. The field of view was
limited by the PS-PMTS available in the early 1990s.
Recent demands for these devices and their deploy-
ment in small-animal PET scanners has led to much
improvement, both in uniformity and in ability
to image much closer to the edge without serious
distortion. Now instruments such as the PEM Flex
are able to capitalize on the advances in photon
Fig. 6. PEM image from the Naviscan imager. In addi- sensors. The problem of sensitivity loss near the
tion to one large and four smaller interductal lesions edges of the field of view is intrinsic to 3D PET
in the outer aspect of the breast, there is another acquisitions. During early PEM development, this
hypermetabolic region at the posterior wall (arrow),
was not considered seriously enough. Now that it
suggestive of axillary node involvement. (From Wein-
has been widely recognized, the situation has been
berg IN, Beylin D, Zavarzin V, et al. Positron emission
mammography: high-resolution biochemical breast improved through the use of higher density, thin-
imaging. Technol Cancer Res Treat 2005;4:55–60; ner shielding, better PMTs, and improved geome-
with permission.) try. This results in the effective field of view being at
least 3 cm closer to the chest in the latest instru-
four different sites during the first year of use of the ments than in the prototype developed by the
instrument. Analysis of these cases showed a sensi- author’s group.
tivity of 93% and a specificity of 83%.
Fig. 6 is an example of a PEM image from this Acknowledgments
instrument. Unlike the PEM-1 scanner, the field of
view is much larger, 24 × 18 cm rather than 5.5 × The author gratefully acknowledges support from
6.0 cm. The device also employs a sophisticated the Natural Science and Engineering Council of
limited-angle reconstruction algorithm, which pro- Canada over the last 15 years. PEM research at
duces much clearer 3D images. The five smaller the McGill University teaching hospitals has been
lesions would not have been visible on a conven- a team effort. The work of Dr. R Lisbona, Chairman
tional PET scan, or on the PEM-1 instrument. The of the Radiology Department, and Dr. Antoine
PEM Flex also has the ability of scanning closer to Loutfi of the Surgical Oncology unit of the McGill
the chest wall. University Health Centre: Royal Victoria Hospital
played vital roles in the success of this project.
Students Kavita Murthy, Yani Picard, James Robar,
Future prospects for positron emission Alanah Bergman, Raymond Clancy, Marie-Anne
mammography Aznar, Martin Hinse, and Nan Zhang all contrib-
In the United States, Medicare approval for use uted to the development of PEM under the au-
of PET imaging of breast cancer was given in late thor’s supervision.
2003 [17]. Unlike the approvals granted for PET in
other forms of malignant disease, the approval docu- References
ment refers to three specific areas: initial staging
of axillary lymph nodes, detection of loco-regional [1] Wahl RL, Cody RL, Hutchins GD, et al. Primary
recurrence or distant metastasis/recurrence, and and metastatic breast carcinoma: initial clinical
evaluation with PET with the radiolabeled glu-
evaluating response to treatment. Although this is
cose analogue 2-[F-18]-fluoro-2-deoxy-D-glucose.
intended for the use of conventional WB-PET scan- Radiol 1991;179:765–70.
ners, there is an opportunity for the assessment of [2] Adler LP, Crowe JP, Al-Kaisi NK, et al. Evaluation
dedicated PEM imaging instruments within the of breast masses and axillary lymph nodes with
guidelines of these new regulations. The ’’response [F-18] 2-deoxy-2- fluoro-D-glucose PET. Radiol
to treatments’’ aspect of the approval document is 1993;187:743–50.
38 Thompson

[3] Avril N, Schelling M, Dose J, et al. Utility of PET [11] Murthy K, Bergman AM, Thompson CJ, et al.
in breast cancer. Clin Positron Imaging 1999;2: Positron emission mammographic instrument:
261–71. initial results. Radiology 2000;215:280–5.
[4] Weinberg IN. US patent # 5,252,830. Dedicated [12] Murthy K, Aznar M, Thompson CJ, et al.
apparatus and method for emission mammogra- Preliminary clinical evaluation of an instrument
phy. October 12, 1993. for positron emission mammography (PEM-I).
[5] Thompson CJ, Murthy K, Weinberg IN, et al. J Nucl Med 2000;41:1851–8.
Feasibility study for positron emission mammog- [13] Smith MF, Majewski S, Weisenberger AG, et al.
raphy. Med Phys 1994;21:529–37. Analysis of factors affecting positron emission
[6] Thompson CJ, Murthy K, Picard Y, et al. Positron mammography (PEM) image formation. IEEE
emission mammography (PEM): a promising tech- Trans Nucl Sci 2003;50:53–9.
nique to detect breast cancer. IEEE Trans Nucl Sci [14] Weinberg IN, Beylin D, Zavarzin V, et al. Posi-
1995;NS42:1012–7. tron emission mammography: high-resolution
[7] Bergman A, Thompson CJ, Murthy K, et al. biochemical breast imaging. Technol Cancer Res
Technique to obtain positron emission mammo- Treat 2005;4:55–60.
graphy images in registration with X-ray mam- [15] Huber JS, Choong WS, Wang J, et al. Develop-
mograms. Med Phys 1998;25:2119–29. ment of the LBNL positron emission mammog-
[8] Townsend DW, Beyer T, Kinahan PE, et al. raphy camera. IEEE Trans Nucl Sci 2003;50:
The SMART scanner: a combined PET/CT tomo- 1650–3.
graph for clinical oncology. RSNA, Chicago, [16] Karimian A, Thompson CJ, Sarkar S, et al. CYBPET:
December 1998. a cylindrical PET system for breast imaging.
[9] Townsend DW, Nutt R. US patent #6,490,476. Nucl Instrum Methods Phys Res A 2005;545(2):
Combined PET and X-Ray CT tomograph and 427–35.
method for using same. December 3, 2002. [17] FDG positron emission tomography for evaluat-
[10] Robar JL, Thompson CJ, Murthy K, et al. Con- ing breast cancer. Blue Cross and Blue Shield
struction and calibration of detectors for high Technology Evaluation Center Report: 18:14.
resolution metabolic breast cancer imaging. Nucl Available at: http://www.bcbs.com/tec/Vol18/
Instrum Methods Phys Res A 1997;392:402–6. 18_14.pdf. Accessed August 11, 2005.
39

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 39–49

FDG-PET and PET/CT in Radiation


Therapy Simulation and
Management of Patients Who Have
Primary and Recurrent Breast Cancer
a,* a b
Dwight E. Heron, MD , Sushil Beriwal, MD , Norbert Avril, MD

& Conventional radiation therapy planning & Current limitations of F-18


& Image guidance for radiation therapy fluorodeoxyglucose-positron emission
planning tomography and positron emission
& Radiation therapy planning: use of F-18 tomography/computed tomography for
fluorodeoxyglucose-positron emission radiation therapy planning
tomography and positron emission & Radiosurgery treatment planning and
tomography/computed tomography positron emission tomography/computed
& F-18 fluorodeoxyglucose-positron emission tomography
tomography for radiation therapy of the & Future perspectives
breast and loco-regional lymph nodes & Summary
& F-18 fluorodeoxyglucose-positron emission & References
tomography/computed tomography for
radiation treatment planning

Precise anatomical information regarding the (eg, in the lungs); however, definition of tumor
location and extent of tumor tissue is essential for involvement, in lymph nodes for example, based on
radiation treatment planning. In the past, anatomi- the increased size is more difficult. Some enlarged
cal imaging has greatly improved the accuracy of lymph nodes may be reactive, whereas smaller nodes
delineating target structures and is currently the may harbor metastatic foci. MRI is better at outlining
basis of modern three dimensional (3D)-based ra- soft tissues, particularly in the brain, and has been
diation treatment [1]. Plain radiography, CT, and valuable in complementing CT-based radiation treat-
MRI provide structural or morphological informa- ment planning [2].
tion based on tissue density, size, vascularity, and Over recent years, functional imaging with posi-
fat or water content, and are routinely being used tron emission tomography (PET) has gained in-
for radiation treatment planning. The excellent spa- creased importance in determining biological or
tial resolution of anatomical imaging techniques molecular abnormalities in specific tumors. PET
allows the detection of subcentimeter lesions using [F-18] fluorodeoxyglucose (FDG) allows the

a
Department of Radiation Oncology, University of Pittsburgh School of Medicine, University of Pittsburgh
Cancer Institute, UPMC Shadyside Hospital, 5150 Centre Avenue, #545, Pittsburgh, PA 15232, USA
b
Department of Nuclear Medicine, Barts and the London School of Medicine, Queen Mary, University of
London, West Smithfield (QEII), London, EC1A 7BE, UK
* Corresponding author.
E-mail address: herond2@upmc.edu (D.E. Heron).

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.003
pet.theclinics.com
40 Heron et al

metastasis as well as for intracranial disease. Pallia-


tive radiation has been shown to be effective in im-
proving the quality of life and providing relief of
symptoms, especially the pain of bone metastasis.

Conventional radiation therapy planning


For the last half a century, breast irradiation has
been performed through parallel opposing tangen-
tial beams with wedges, with matching anterior
supraclavicular and posterior axillary fields when-
ever appropriate. Radiation simulation, the process
of selecting the area to be treated and beam se-
lection, is routinely performed through fluoro-
scopic placement of the tangential beams, based
upon clinically determined breast tissue borders.
Treatment planning has been single-slice, two-
Fig. 1. 2D tangential beam showing the isodose lines dimensional (2D) dosimetry, applying wedge beam
in the central axis. data onto manually obtained breast central axis con-
tours [Fig. 1].
characterization of suspicious masses as well as de- Although this unsophisticated technique initially
termination of the spread of disease to loco-regional produced a breakthrough in breast conservation
lymph nodes and distant sites [3]. The metabolic therapy, a number significant limitations of this
information derived from FDG-PET can influence technique are now apparent, including excess car-
the radiation treatment planning in several ways. diac and lung sequelae of breast irradiation [12]. In
First, FDG-PET may reveal a tumor target that was addition, patients desire better cosmetic results and
previously not detected by anatomical imaging. shorter treatment times. Increased scatter radiation
Second, FDG-PET may detect additional tumor re- to contralateral breast from wedges may increase
gions outside the tumor volume defined by CT or the incidence of second malignant neoplasm [13].
MRI. Third, FDG-PET may show subregions or foci Furthermore, some have suggested that poor dose
with increased or altered biological activity within distribution may contribute to late in-breast tumor
the gross tumor volume that could be preferentially failures. Data from Bhatnagar and colleagues [14]
targeted and treated with escalated radiation doses. suggest significant reduction of scatter radiation to
Therefore, the more precise 3D delineation of tumor the contralateral breast with the use of intensity-
volumes could translate into improved loco-regional modulated radiation therapy (IMRT). As a result,
control with radiation therapy. newer techniques using CT-based treatment plan-
Breast cancer is the most common malignancy ning have been used to create ‘‘beam’s eye’’ views to
among women in the United States, with approxi- avoid these underlying critical structures [Fig. 2].
mately 211,240 cases expected in 2005 [4]. Breast
irradiation was first used to minimize the risk of
loco-regional recurrence in patients who had lo-
cally advanced cancers [5]. Beginning in the late
1970s, breast irradiation has evolved into the main-
stay of therapy in patients treated with breast
conservation surgery. In recent years, breast conser-
vation therapy consisting of a segmental mastec-
tomy and definitive breast irradiation has assumed
an increasing role in the management of patients
who have both invasive and noninvasive breast
cancer. No fewer than six prospective, randomized
controlled trials have shown equivalent outcomes
of breast conservation therapy compared with mas-
tectomy [6–11]. Distant metastases of breast cancer
are frequently found in lymph nodes, lungs, liver,
and bones. ,
Fig. 2. Beam s eye view of 3D partial wide tangent
In the metastatic setting, radiation therapy is cur- covering chest wall and internal mammary nodes
rently used for treatment of soft tissue and bone (blue) and sparing heart (green).
FDG-PET and PET/CT Radiation Therapy Planning 41

These beams eye views allow the radiation oncolo- when selecting field arrangements for treatment of
gist to examine the target and nearby critical struc- these patients, and has become standard of care.
tures from the perspective of the beam’s path.

Radiation therapy planning: use of F-18


fluorodeoxyglucose-positron emission
Image guidance for radiation therapy tomography and positron emission
planning tomography/computed tomography
CT-based, 3D treatment planning represents an Currently, CT-assisted volume definition remains
evolution from 2D planning, and provides volu- the gold standard for external beam radiation ther-
metric information that permits a greater assess- apy with curative intent [1]. Accurate localization
ment of the beam’s path and subsequent dose to of the gross tumor volume (GTV) is critical to
target tissues and normal structures [Fig. 3]. CT- optimizing the therapeutic ratio, by sparing normal
based approaches also permit virtual simulation, a tissues and maximizing coverage of tumor volumes
process in which the CT data set is acquired rapidly, in conformal radiation techniques. Using current
thus significantly reducing the time the patient is available anatomical imaging for treatment plan-
spending on the simulation table. Treatment of ning has several limitations. These include the
chest wall and regional nodes poses significant chal- visualization and precise delineation of tumors in
lenges to radiation oncologists, particularly in the certain areas of the body, such as the head and
postmastectomy setting. The range of body habi- neck, but also in postsurgical distorted anatomy,
tus and close proximity of the internal mammary and in residual scar tissue after chemotherapy,
nodes to the heart often necessitates individualized specifically in previously irradiated areas. The in-
treatment planning, with complex field arrange- corporation of metabolic FDG-PET imaging in
ments. 3D treatment planning with volumetric radiation treatment planning has raised hopes for
information helps in clinical decision-making further improvement, which would lead to so-
called ‘‘multidimensional conformal radiotherapy.’’
FDG-PET is complementary to conventional meth-
ods of staging in that it has been shown to have
both higher sensitivity and specificity in detecting
nodal metastases and distance in a variety of ma-
lignancies, including breast cancer, when compared
with CT alone [15]. The sensitivity of FDG-PET in
the detection of axillary lymph node metastasis
from breast cancer varies from 20% to 100%
when compared with sentinel lymph node (SLN)
biopsy or axillary node dissection [16–22]. This
variation in the sensitivity of FDG-PET to detect
tumor-involved lymph nodes depends primarily
on the tumor stage, and subsequently on the extent
of disease. Step sectioning and immunohistochemi-
cal staining of sentinel lymph nodes detected
micrometastases in up to 45% of cases [21]. FDG-
PET can miss micrometastases in lymph nodes
when the number of tumor cells is too small to
provide a signal above the background activity.
Avril and coworkers [17] studied 51 patients and
found an overall sensitivity and specificity for de-
tection of axillary lymph node metastases of 79%
and 96%, respectively. In patients who had primary
breast tumors larger than 2 cm (>stage pT1), the
sensitivity increased to 94%, with a corresponding
specificity of 100%; however, in patients who had
small primary breast cancer (stage pT1), the sensi-
tivity of FDG-PET was only 33%. In a prospective
Fig. 3. 3D radiation treatment plan showing isodose multicenter study including 360 women [23] who
distribution covering the breast in axial and sagit- had newly diagnosed invasive breast cancer, the
tal planes. largest patient group so far, the sensitivity for
42 Heron et al

FDG-PET was 61%, with a corresponding specificity initial diagnosis, this area could be effectively tar-
of 80%. Despite the limitations of FDG-PET dis- geted with conformal radiation techniques.
cussed above, specifically in detecting micrometas- Several reports have suggested that FDG-PET is
tases, its accuracy for detecting tumor-involved superior to conventional imaging for detection of
axillary lymph nodes is superior to those of CT or distant metastatic disease. The use of FDG-PET
MRI alone. in patients who have locally advanced breast can-
Studies have consistently demonstrated that cer frequently results in the detection of unexpected
FDG-PET is better than CT in the detection of distant metastases, and has been shown to substan-
internal mammary as well as mediastinal lymph tially change the clinical management [25]. Recently,
nodal metastases [Fig. 4]. In 73 consecutive pa- Dose and colleagues [26] compared FDG-PET
tients who had recurrent or metastatic disease, with chest radiograph, bone scintigraphy, and ultra-
PET was able to correctly identify 40% of the pa- sound of the abdomen for detection of metastatic
tients who had intrathoracic lymph node metas- disease in 50 breast cancer patients. FDG-PET iden-
tases, resulting in a sensitivity of 85% and a tified metastatic disease with a sensitivity and a speci-
specificity of 90% [24]. Only 23% of the patients ficity of 86% and 90%, respectively, as compared
had suspiciously enlarged lymph nodes in CT, lead- with 35% and 95%, respectively, for conventional
ing to a sensitivity of 54% and a specificity of 85%. imaging procedures. In another retrospective analy-
Therefore, the overall diagnostic accuracy of PET sis of 62 patients [27], sensitivity and specificity for
(88%) was higher compared with that of CT (73%) detecting local recurrences or distant metastases
[24]. In locally advanced breast cancer patients, the were 97% and 82%, respectively, for FDG-PET, ver-
prevalence of internal mammary FDG uptake can sus 84% and 60% for conventional imaging.
be as high as 25% [25]. The potential importance The skeleton is a common site for distant metas-
of internal mammary nodal drainage has also been tases in breast cancer. Bone scintigraphy is an estab-
brought to the forefront with the widespread use lished screening method for bony metastases, and
of axillary sentinel node mapping, which fre- allows determining the extent of disease; however,
quently shows internal mammary lymph drainage tracer uptake in bone scintigraphy reflects osteo-
on lymphoscintigraphy. There is some speculation blastic activity, and is therefore limited in the detec-
that a subset of patients who have ‘‘sternal metas- tion of osteolytic lesions. Cook and coworkers [28]
tases’’ may in fact be internal mammary node fail- studied 23 breast cancer patients who had known
ures with erosion of the sternum. If detected at skeletal metastases, and FDG-PET was able to de-

Fig. 4. (A–C) PET, CT, and PET/CT demonstrating FDG-avid left chest wall metastasis.
FDG-PET and PET/CT Radiation Therapy Planning 43

tect more lesions than bone scintigraphy overall. has suggested more late cardiac death in patients
Higher FDG uptake was observed for osteolytic treated with radiation therapy compared with those
than for osteoblastic bone metastases, and FDG- treated surgery alone [31]. This effect appeared to
PET also detected more osteolytic lesions. On the be more pronounced in younger women who
other hand, PET detected fewer bone metastases had left-sided breast cancer and who were treated
than bone scintigraphy in patients who had osteo- with anthracycline-based chemotherapy. The excess
blastic metastases. In a larger series of 48 breast cardiac mortality is thought to be due to a com-
cancer patients [29], a total of 127 bone lesions, bined toxicity of chemotherapy and radiation of
including 105 metastatic and 22 benign lesions, the left ventricle and major coronary vessels. The
were found by either FDG-PET or bone scintigra- introduction of intensity-modulated radiation ther-
phy. The sensitivity and diagnostic accuracy of apy (IMRT) technology in breast treatment has the
FDG-PET were 95.2% and 94.5%, respectively, ver- potential to provide uniform dose coverage to the
sus 93.3% and 78.7% for bone scintigraphy. Al- left breast while limiting dose to the underlying
though the overall sensitivity for bone scintigraphy heart and lungs [Fig. 5]; however, this issue is
and PET are reported as comparable, bone scin- complicated by the respiratory motion of the
tigraphy seems to be superior in the detection of chest wall and physiologic cardiac motion. These
osteoblastic disease, whereas FDG-PET is superior motions may cause geometric deviations of the
in osteolytic metastases, suggesting a complemen- heart from the intended location, causing unde-
tary role for the imaging procedures. sired higher doses to the heart. The use of four-
dimensional (4D) CT and beam gating systems in
radiation therapy has proven effective in limiting
F-18 fluorodeoxyglucose-positron emission the motion impacts to radiation treatments in lung
tomography for radiation therapy of the cancer [32]. Similar techniques may be used to
breast and loco-regional lymph nodes manage chest wall and cardiac motion by the selec-
Breast cancer may be unique in that there are tion of phases that minimize the overlap of the
numerous loco-regional treatments using various treatment portal with the heart and lungs.
combinations of surgical techniques and radiother- The accurate evaluation and diagnosis of internal
apy regimens. Surgery may vary from excisional mammary metastases is of great importance in
biopsy with microscopically involved margins, to radiation treatment planning. The diagnosis of
lumpectomy, which usually indicates a more gen- internal mammary metastases can facilitate directed
erous resection (again with or without positive radiation or surgical intervention, thus allowing
margins), to quadrantectomy, simple mastectomy, for treatment planning tailored more closely to
modified radical mastectomy, and even to radical each individual patient’s disease and risk. Similarly,
mastectomy. There are several accepted radiation chemotherapy choices may also be affected, par-
therapy techniques, ranging from treatment of the ticularly in the patient who appears to have node-
entire breast with or without a boost to the lum- negative disease by conventional staging of the
pectomy cavity following breast conservation sur- axilla, but is found to have internal mammary
gery, to more comprehensive techniques including involvement. The routine use of radiation to treat
the regional nodes [30]. These nodal areas include the internal mammary lymph nodes in women
the axilla, supraclavicular region, and the upper who have locally advanced breast cancer remains
internal mammary region. Similarly, following the controversial; however, internal mammary disease
mastectomy, radiotherapy may be administered to has been associated with higher rates of distant me-
just the chest wall, or to the chest wall with or tastases and lower overall survival [33]. Although
without one or more of the regional nodal areas. older extended radical mastectomy series did not
Currently, FDG-PET cannot provide additional
information in the radiation treatment planning
of primary breast cancer following breast conser-
ving surgery, because there is no macroscopic (re-
sidual) disease. In the setting of local recurrence,
FDG-PET may aid in determining more precisely
the extent of tumor tissue, and may identify addi-
tional lymph node involvement in the axilla or
internal mammary chain, as well as assess meta-
static tumor to the chest wall.
Treatment-related toxicities in breast cancer pa- Fig. 5. (A, B) IMRT plan showing homogenous dose
tients treated with radiation therapy have improved to the breast in axial and sagittal plane with sparing
over the last 2 decades. Nevertheless, meta-analysis of heart.
44 Heron et al

demonstrate a survival advantage to internal mam- have first-episode loco-regional recurrence. In their
mary dissection, they did demonstrate that for study of 175 patients, they found that 16% had
medially located tumors the incidence of involve- distant metastases at the time of loco-regional recur-
ment was up to 30% to 40%, especially when the rence, and 24% developed distant metastases within
axilla was also positive for metastases [34,35]. In 18 months of confirmation of recurrent disease.
subset analyses, there was a suggestion of a poten- They estimated that FDG-PET would upstage and
tial disease-free advantage in patients who had likely change the therapeutic plan in up to 29%
medially located primary tumors and axillary me- of patients who have negative conventional stag-
tastases, those most likely to have internal mam- ing studies.
mary involvement when these areas were subjected
to treatment. Moreover, a recent large retrospective
F-18 fluorodeoxyglucose-positron emission
review from the British Columbia Cancer Agency
tomography/computed tomography for
[36] found a poorer overall survival and distant
radiation treatment planning
metastases-free survival in high-risk women who
had medial tumors, again indicating the presence Combining two imaging devices, PET and CT, into
of untreated internal mammary disease. Despite an one integrated PET/CT scanner has substantially
overall trend to less extensive surgery, interest in the broadened the clinical use of metabolic PET imag-
detection and treatment of internal mammary nodes ing, specifically in oncology, and particularly for
has resurfaced in recent years. Two large random- radiation treatment planning [41–46]. PET/CT is
ized trials published in the New England Journal of unique because it allows the acquisition of spatially
Medicine [37,38] demonstrated a survival benefit to and temporally registered PET and CT data in one
postmastectomy chest wall and regional nodal imaging procedure, providing combined anatomi-
radiation that included the internal mammary cal and functional imaging information that allows
nodes. The relative contribution of internal mam- accurate tissue characterization and determination
mary treatment, however, remains unclear. of the exact localization and extent of tumor tissue.
FDG-PET can contribute in significant ways to the The diagnostic accuracy of PET/CT is generally
clinical management and radiation planning of higher compared with either imaging procedure
patients who have suspected loco-regional recur- performed separately, because of the exact ana-
rences. The most common sites of loco-regional tomical correlation of abnormal FDG uptake and
recurrence among patients treated with mastec- the functional correlation of suspected morphologi-
tomy, axillary node dissection, or radiation therapy cal abnormalities [47]. The accuracy of FDG-PET/CT
are the chest wall and supraclavicular nodes. Be- is specifically higher in the neck and abdomen,
cause it provides biological and functional in- because it distinguishes physiological from patho-
formation, FDG-PET often is complementary to logical uptake by confirmation of metabolic abnor-
conventional staging methods such as physical ex- mality with anatomic correlation. Several reports
amination or cross-sectional imaging (CT or MR), indicate a distinct advantage of PET/CT in breast
which rely more on changes in morphology to cancer as well [47].
detect disease recurrence. This is particularly true Fused FDG-PET and CT images provide radiation
in the evaluation of anatomic regions that have oncologists with two pieces of critical information
been previously treated by surgery or radiation, with a single study: the extent of viable tumor, and
where the discrimination between post-treatment its exact location. Initial studies in patients who
scar and recurrent tumor is often problematic. Bra- had various tumor types have confirmed that
chial plexopathy, for example, is difficult to assess using FDG-PET/CT both in pretreatment planning
on conventional anatomical imaging. FDG-PET and in follow-up evaluations has a significant
and MRI were compared in 10 patients who had impact on radiotherapy management in up to
clinical findings suggestive of breast cancer me- 56% of patients [46]. In 24 patients who were
tastases [39]. Out of 9 patients who had local- planned with 3D conformal radiation therapy for
regional breast cancer metastases, MRI was positive lung cancer, FDG-PET clearly altered the radiation
in 5 patients and indeterminate in 4 patients, therapy volume in 14 (58%) patients. FDG-PET
whereas FDG-PET was positive in all patients. Simi- also helped to distinguish tumor from atelectasis
lar results were found in 19 breast cancer patients in 3 patients. Unsuspected nodal disease was de-
who had symptoms referable to the brachial plexus, tected in 10 patients, and 1 patient had a separate
and it was concluded that FDG-PET may particu- tumor focus detected within the same lobe of the
larly be useful in distinguishing between radiation- lung [45]. Similarly, Heron and colleagues [42]
induced and metastatic plexopathy [40]. Van Oost showed that in head and neck cancer, FDG-PET/CT
and colleagues [25] recently corroborated the need simulation provided valuable information that re-
for a more sensitive staging tool in patients who sulted in greater delineation of normal tissues from
FDG-PET and PET/CT Radiation Therapy Planning 45

tumor-bearing areas at high risk for recurrence. In treatment planning and potential FDG-PET treat-
this study of 21 consecutive patients, the FDG-PET ment planning. Although FDG-PET offers unique
portion of PET/CT demonstrated the primary tu- metabolic information about the tumor activity,
mor in all cases, whereas the coregistered CT did the limited spatial resolution of PET compared
not delineate the primary in three cases. In 8 pa- with CT or MRI may not characterize small lesions
tients, additional areas of disease were identified by with sufficient accuracy. Current PET technologies
FDG-PET. Because of the often poor anatomical do not allow identifying micrometastases, which
delineation of head and neck tumors, the volumes limits its use for narrowing the treatment volume
for the primaries were significantly larger on CT in lymph nodes areas at risk. The specificity of
treatment planning than on FDG-PET, which pro- FDG-PET is in the range of 80% to 95%, and gen-
vided more precise metabolic definition of the erally higher than that of CT or MRI.; however,
tumor extent. In contrast, the volumes for nodal the uptake of FDG is not specific for tumor tissue,
regions were smaller on CT because FDG-PET de- and granulomatous or acute inflammatory pro-
monstrated tumor outside the CT-defined target cesses can result in false-positive PET findings. An
volumes. The additional metabolic information important limitation of FDG-PET in radiation treat-
from FDG-PET resulted in a more accurate charac- ment planning is the precise delineation of tumor
terization of the extent of disease for both primary tissue based on the PET images. Unlike anatomical
tumors and nodal regions, with the potential clini- imaging, the size of metabolic abnormalities var-
cal implication of improved radiation treatment. ies depending on the scaling of the PET display.
Ciernik and coworkers [46] investigated the useful- Whereas tumors often have well-defined ana-
ness of FDG-PET/CT imaging for target volume tomic borders on CT images, the edges of tumors
definition in 39 patients who had solid tumors. on FDG-PET imaging appear indistinct to the con-
The gross tumor volume increased by 25% or more touring physician. Some have arbitrarily defined
because of FDG-PET in 17% of cases who had head- the FDG-avid volume as the region encompassed
and-neck and lung cancer, and in 33% of cases who by the 40% to 50% [45] intensity level relative
had cancer of the pelvis. The gross tumor volume to the tumor maximum, whereas others have nor-
was reduced 25% or more in 33% of patients who malized to the FDG uptake in the liver without
had head-and-neck cancer, in 67% who had lung background subtraction [42]; hence the FDG-avid
cancer, and in 19% who had cancer of the pelvis. volume may have significant interobserver variabil-
Overall, in 56% of cases, gross tumor volume delin- ity. The use of standardized uptake value (SUV)
eation was changed significantly if information from normalized parametric images may help to over-
metabolic PET imaging was used in the planning come this problem. Major limitations of FDG-PET
process. The modification of the gross tumor volume for radiation treatment planning are the limited
translated into altered planning target volume (PTV) anatomical landmarks, which are necessary for ex-
changes exceeding 20% in 46% of cases. act tumor target definition on the treatment plan-
An important implication from improved target ning system.
definition is avoiding geographic misses and unnec- As discussed above, the use of coregistered
essary exposure of nontarget tissue to radiation. In FDG-PET and CT images obtained from combined
addition, improved target definition may also PET/CT scanners may greatly improve the use of
allow dose escalation in 3D and IMRT. The authors PET for radiation treatment planning, by provid-
found a close correlation between the metabolic ing the metabolic tumor activity and the precise
activity on pretreatment FDG-PET and the sub- anatomical information. The sequential nature of
sequent risk of treatment failure (unpublished the PET/CT acquisition does not completely pre-
data, 2005). If this observation is confirmed in vent misregistration between the PET and the
larger trials, the intensity of FDG uptake in tumor CT portion of the examination. In most PET/CT
tissue could guide and tailor the radiation dose scanners, a CT scan is acquired first from the base
delivered by IMRT, and therefore improve radiation of the skull though the pelvis, followed within
treatment outcomes. approximately 20 to 60 seconds by PET data acqui-
sition. Because a PET emission scan takes between
3 and 5 minutes per bed position (which is ap-
Current limitations of F-18
proximately 15–20 cm, depending on the scanner
fluorodeoxyglucose-positron emission
manufacturer) the PET images represent an average
tomography and positron emission
over the period of data acquisition, whereas as
tomography/computed tomography for
the CT represents a snapshot from a fraction of
radiation therapy planning
a second. Misregistration of tumors on the PET
There are important differences between the cur- compared with the CT portion of PET/CT is com-
rently used anatomical-imaging–based radiation mon because of respiratory motion, specifically at
46 Heron et al

the base of the lungs and in the upper abdomen specific increased signal intensity in the bone mar-
(liver). In breast cancer, differences in the breath- row, limiting the definition of viable tumor.
ing cycle can cause misregistration, specifically of The potential advantage of FDG-PET and PET/CT
tumor lesions in the breast, chest wall, and internal is in providing more accurate treatment planning
mammary lymph node chain. Different filling of compared with CT and MRI alone. This could result
the renal collecting system and the urinary bladder in improved local control rates after radiosurgery,
are also frequently observed because of the sequen- and reduce subsequent tumor recurrence rates.
tial imaging of CT and PET. Finally, patient move- There is only very little information so far describ-
ment may occur between the PET and the CT ing the role of FDG-PET and PET/CT for CyberKnife
portion, specifically in the head and neck region. treatment planning, however. Prospective studies
Heron and colleagues [42] have used a thermoplas- are needed comparing volumetric dose planning
tic mask for radiation treatment planning with for target definition and radiosurgical treatment
FDG-PET/CT of head and beck cancers, and suc- planning using FDG-PET and PET/CT with cur-
cessfully immobilized patients to improve the rently used CT and MRI. For potential clinical ap-
precise coregistration between PET and CT. Laser- plication, it is important to demonstrate that the
assisted localization for treatment planning is often metabolic information is different from the ana-
used for current CT-based radiation treatment plan- tomical tumor delineation, and provides specific
ning, and might also be necessary for PET/CT radia- additional information. It has also to be shown
tion treatment planning. that the ability to tailor radiation doses to a specific
metabolic activity, as opposed to using conven-
tional doses based upon histology alone, results
in improved local control. It is also important to
Radiosurgery treatment planning and
determine specific features of FDG-PET and PET/CT
positron emission tomography/computed
that would predict local failure to standard radio-
tomography
surgical dose prescriptions.
There are several techniques being developed to
extend radiosurgery from the brain to the body.
Future perspectives
The CyberKnife (Accuray, Sunnyvale, California) sys-
tem [48] for example, consists of a lightweight linear Preliminary data provide early proof of the princi-
accelerator mounted on a robotic arm with 1-mm ple that multimodality imaging can facilitate indi-
spatial accuracy. Real-time radiograph imaging track- vidualized treatment by better delineation of target.
ing allows for correction of patient movement by In theory, better delineation of the target should
bringing the radiation beam into alignment with mean better tumor coverage, and hence better local
the observed position of the treatment target. An control. Better target delineation will not help if
important advantage of this type of radiosurgery we are missing the target because it moves as a
is that each beam is delivered independently, with- result of physiological or other mechanical forces.
out a fixed isocenter. Because of the spatial preci- Future studies are needed to integrate respiratory
sion with which radiation can be delivered, it is gating with PET/CT scanning to account for target
feasible to administer a tumoricidal radiation dose motion in radiation treatment planning and deliv-
in a single outpatient treatment. Radiosurgery is ery. The use of 4D PET/CT can correct for respira-
specifically helpful in spinal metastases from breast tory motion artifacts seen in conventional PET/CT
and other cancers, but can potentially be used for imaging. It has potential to reduce smearing and
any tumor localization within the body [49]. improve the accuracy in PET/CT coregistration.
Because of the high precision and the steep decline The metabolically active disease seen by PET can
in delivered tumor dose, extension beyond the ana- be treated with escalated dose to improve chances
tomical abnormality would lead to underdosing of of local control. Prospective trials are needed to
the tumor. evaluate and validate the role of dose escalation
The major potential benefits of radiosurgery are in this setting using IMRT-based technique. Besides,
the short treatment time in an outpatient setting, the studies performed to date using PET/CT in ra-
with rapid recovery and good symptomatic diation planning have only shown that treatment
response. Tumor target definition and dose plan- volume changes significantly with the use of func-
ning are currently based on CT, however, which tional imaging. It has yet to be determined whether
frequently provides poor delineation of tumor the PET-defined target volume leads to improved
involvement, specifically in bone. When possible, outcomes in terms of local control, survival, and
MRI is being used to more accurately define the reduced toxicities.
extension of tumor tissue; however, in previously PET/CT also has application in assessment of treat-
irradiated areas, MRI frequently demonstrates un- ment response. The utility of post-therapy FDG-PET
FDG-PET and PET/CT Radiation Therapy Planning 47

to monitor tumor response also has been evalu- [5] Recht A, Edge SB, Solin LJ, American Society of
ated in several tumors, including lymphoma, Clinical Oncology. Postmastectomy radiother-
breast, cervix, and colorectal cancers [50–53]. There apy: clinical practice guidelines of the American
are also encouraging results available for treatment Society of Clinical Oncology. J Clin Oncol 2001;
evaluation of radiation therapy. The concept of as- 19(5):1539–69.
[6] Jacobson JA, Danforth DN, Cowan KH, et al.
sessment and prediction of treatment response by
Ten-year results of a comparison of conservation
sequential FDG-PET imaging needs to be validated with mastectomy in the treatment of Stage I and
in larger studies to help in early identification of II breast cancer. N Engl J Med 1995;332(14):
treatment failure. This can lead to early intervention 907–11.
with potential for salvage therapy, which might im- [7] van Dongen JA, Bartelink H, Fentiman IS, et al.
prove outcome. Randomized clinical trial to assess the value of
From the authors’ perspective, there is no doubt breast-conserving therapy in Stage I and II breast
that FDG-PET/CT will become an important part of cancer, EORTC 10801 trial. J Natl Cancer Inst
modern radiation treatment planning in the near Monogr 1992;11:15–8.
future; however, it should be emphasized that cur- [8] Sarrazin D, Le MG, Arriagada R, et al. Ten-year
results of a randomized trial comparing a con-
rently, FDG-PET can only be used as complemen-
servative treatment to mastectomy in early breast
tary imaging modality for detecting disease not cancer. Radiother Oncol 1989;14(3):177–84.
identified by CT. The routine use of FDG-PET or [9] Blichert-Toft M, Rose C, Anderson JA, et al.
PET/CT for radiation treatment planning requires a Danish randomized trial comparing breast con-
thorough and careful evaluation in large prospec- servation therapy with mastectomy. Six years of
tive studies as it relates to the promise of improving life-table analysis. J Natl Cancer Inst Monogr
local control or increasing disease-free and over- 1992;11:19–25.
all survival. [10] Veronesi U, Banfi A, Salvadori B, et al. Breast
conservation is the treatment of choice in small
breast cancer. Long-term results of a randomized
trial. Eur J Cancer 1990;26(6):668–70.
Summary [11] Fisher B, Anderson S, Redmond C, et al.
PET-CT based imaging is a valuable tool, and one Reanalysis and results after 12 years of follow-
of the most useful tools in the staging and restaging up in a randomized clinical trial comparing total
mastectomy with lumpectomy with or without
of breast cancer, especially in patients who have
irradiation in the treatment of breast cancer.
recurrent or locally advanced breast cancer. Its great- N Engl J Med 1995;333(22):1456–61.
est clinical applications are in detection and defini- [12] Cuzick J, Stewart H, Rutqvist L, et al. Cause-
tion of extent of recurrent or metastatic disease, and specific mortality in long-term survivors of breast
in monitoring response to therapy. The role of PET- cancer who participated in trials of radiotherapy.
CT in radiation treatment planning for breast cancer J Clin Oncol 1994;12(3):447–53.
is still in the nascent stages, lacking controlled ran- [13] Gao X, Fisher S, Emami B. Risk of second pri-
domized trials data; however, the potential to mary cancer in the contralateral breast in women
improve radiation treatment planning by allowing treated for early stage breast cancer A population
for the tailoring of comprehensive radiation portals, based study. Int J Radiat Oncol Biol Phys 2003;
56:1038–45.
particularly for locally advanced breast cancer,
[14] Bhatnagar AK, Brandner E, Sonnik D, et al.
makes this one of the most promising tools are we Intensity-modulated radiation therapy (IMRT)
enter the era of image-guided radiation therapy. reduces the dose to the contralateral breast when
compared to conventional tangential fields for
primary breast irradiation: Initial Report. Cancer J
References
2004;10(6):381–5.
[1] Bucci MK, Bevan A, Roach 3rd M. Advances in [15] Schirrmeister H, Kuhn T, Guhlmann A. [F-18]-
radiation therapy: conventional to 3D, to IMRT, 2-deoxy-2-fluoro-D-glucose PET in the preopera-
to 4D, and beyond. CA Cancer J Clin 2005;55(2): tive staging of breast cancer: comparison with the
117–34. standard staging procedures. Eur J Nucl Med
[2] Jansen EP, Dewit LG, van Herk M, et al. Target 2001;28:351–8.
volumes in radiotherapy for high-grade malig- [16] Adler LP, Crowe JP, Al-Kaisi NK. Evaluation
nant glioma of the brain. Radiother Oncol 2000; of breast masses and axillary lymph nodes with
56(2):151–6. [F-18]-2-deoxy-2-fluoro-D-glucose PET. Radiology
[3] Quon A, Gambhir SS. FDG-PET and beyond: 1993;187:743–50.
molecular breast cancer imaging. J Clin Oncol [17] Avril N, Rose CA, Schelling M. Breast imaging
2005;23(8):1664–73. with positron emission tomography and [fluo-
[4] Greenlee RT, Murray T, Bolden S. Cancer sta- rine-18]-fluorodeoxyglucose: use and limitations.
tistics, 2000. CA Cancer J Clin 2000;50(1):7–33. J Clin Oncol 2000;18:3495–502.
48 Heron et al

[18] Adler LP, Faulhaber PF, Schnur KC. Axillary tectomy radiotherapy of the chest wall: dosi-
lymph node metastases: screening with [F-18]-2- metric comparison of common techniques. Int J
deoxy-2-fluoro-D- glucose (FDG) PET. Radiology Radiat Oncol Biol Phys 2002;52(5):1220–30.
1997;203:323–7. [31] Cuzick J, Stewart H, Peto R, et al. Overview
[19] Avril N, Dose J, Janicke F. Assessment of axil- of randomized trials of postoperative adjuvant
lary lymph node involvement in breast cancer radiotherapy in breast cancer. Cancer Treat Rep
patients with positron emission tomography 1987;71(1):15–29.
using radiolabeled 2-[fluorine-18]-fluoro- [32] Rietzel E, Chen GT, Choi NC, et al. Four-
2-deoxy-D-glucose. J Natl Cancer Inst 1996;88: dimensional image-based treatment planning:
1204–9. target volume segmentation and dose calculation
[20] Wahl RL, Siegel BA, Coleman RE. Prospective in the presence of respiratory motion. Int J Radiat
multi-center study of axillary nodal staging with Oncol Biol Phys 2005;61(5):1535–50.
FDG positron emission tomography in breast [33] Lacour J, Le M, Caceres E, et al. Radical mas-
cancer. J Clin Oncol 2004;22:277–85. tectomy versus radical mastectomy plus inter-
[21] Lovrics PJ, Chen V, Coates G. A prospective nal mammary dissection. Ten year results of an
evaluation of positron emission tomography international cooperative trial in breast cancer.
scanning, sentinel lymph node biopsy, and stan- Cancer 1983;51:1941–3.
dard axillary dissection for axillary staging in [34] Veronesi U, Valagussa P. Inefficacy of internal
patients with early stage breast cancer. Ann Surg mammary node dissection in breast cancer sur-
Oncol 2004;11:846–53. gery. Cancer 1981;47:170–5.
[22] Yang JH, Nam SJ, Lee TS. Comparison of [35] Lacour J, Le MG, Hill C. Is it useful to remove
intraoperative frozen section analysis of sentinel internal mammary nodes in operable breast can-
node with preoperative positron emission tomog- cer? Eur J Surg Oncol 1987;13:309–14.
raphy in the diagnosis of axillary lymph node [36] Lohrisch C, Jackson J, Jones A, et al. Relationship
status in breast cancer patients. Jpn J Clin Oncol between tumor location and relapse in 6781
2001;31:1–6. women with early invasive breast cancer. J Clin
[23] Wahl RL, Siegel BA, Coleman RE. Prospective Oncol 2000;18:2828–35.
multi-center study of axillary nodal staging with [37] Overgaard M, Hansen PS, Overgaard J, et al. Post-
FDG positron emission tomography in breast operative radiotherapy in high-risk premenopausal
cancer. J Clin Oncol 2004;22:277–85. women with breast cancer who receive adjuvant
[24] Eubank WB, Mankoff DA, Takasugi J. 18Fluoro- chemotherapy. N Engl J Med 1997;337:949–55.
deoxyglucose positron emission tomography to [38] Ragaz J, Jackson SM, Le N, et al. Adjuvant
detect mediastinal or internal mammary metas- radiotherapy and chemotherapy in node-positive
tases in breast cancer. J Clin Oncol 2001;19: premenopausal women with breast cancer.
3516–23. N Engl J Med 1977;377:956–62.
[25] van Oost FJ, van der Hoeven JJ, Hoekstra OS. [39] Hathaway PB, Mankoff DA, Maravilla KR, et al.
Staging in patients with locoregionally recurrent Value of combined FDG PET and MR imaging
breast cancer: current practice and prospects for in the evaluation of suspected recurrent local-
positron emission tomography. Eur J Cancer regional breast cancer: preliminary experience.
2004;40:1545–53. Radiology 1999;210(3):807–14.
[26] Dose J, Bleckmann C, Bachmann S, et al. [40] Ahmad A, Barrington S, Maisey M, et al. Use of
Comparison of fluorodeoxyglucose positron positron emission tomography in evaluation of
emission tomography and ‘‘conventional diag- brachial plexopathy in breast cancer patients. Br J
nostic procedures’’ for the detection of distant Cancer 1999;79(3–4):478–82.
metastases in breast cancer patients. Nucl Med [41] Esthappan J, Mutic S, Malyapa RS, et al. Treat-
Commun 2002;23(9):857–64. ment planning guidelines regarding the use of
[27] Heinisch M, Gallowitsch HJ, Mikosch P, et al. CT/PET-guided IMRT for cervical carcinoma with
Comparison of FDG-PET and dynamic contrast- positive paraaortic lymph nodes. Int J Radiat
enhanced MRI in the evaluation of suggestive Oncol Biol Phys 2004;58(4):1289–97.
breast lesions. Breast 2003;12(1):17–22. [42] Heron DE, Andrade RS, Flickinger J, et al. Hybrid
[28] Cook GJ, Houston S, Rubens R, et al. Detection PET-CT simulation for radiation treatment plan-
of bone metastases in breast cancer by 18FDG ning in head-and-neck cancers: a brief technical
PET: differing metabolic activity in osteoblastic report. Int J Radiat Oncol Biol Phys 2004;60(5):
and osteolytic lesions. J Clin Oncol 1998;16: 1419–24.
3375–9. [43] Calvo FA, Domper M, Matute R, et al. 18F-FDG
[29] Yang SN, Liang JA, Lin FJ, et al. Comparing positron emission tomography staging and re-
whole body (18)F-2-deoxyglucose positron emis- staging in rectal cancer treated with preoperative
sion tomography and technetium-99m methy- chemoradiation. Int J Radiat Oncol Biol Phys
lene diphosphonate bone scan to detect bone 2004;58(2):528–35.
metastases in patients with breast cancer. J Can- [44] Erdi YE, Rosenzweig K, Erdi AK, et al. Radio-
cer Res Clin Oncol 2002;128:325–8. therapy treatment planning for patients with
[30] Pierce LJ, Butler JB, Martel MK, et al. Postmas- non-small cell lung cancer using positron emis-
FDG-PET and PET/CT Radiation Therapy Planning 49

sion tomography (PET). Radiother Oncol 2002; of stereotactic radiosurgery and stereotactic ra-
62(1):51–60. diation therapy for patients with spine tumors.
[45] Bradley J, Thorstad WL, Mutic S, et al. Impact of J Neurooncol 2004;69(1–3):319–34.
FDG-PET on radiation therapy volume delinea- [50] Grigsby P, Siegel BA, Dehdashti F, et al.
tion in non-small-cell lung cancer. Int J Radiat Posttherapy [18F] fluorodeoxyglucose positron
Oncol Biol Phys 2004;59(1):78–86. emission tomography in carcinoma of the cervix:
[46] Ciernik IF, Dizendorf E, Baumert BG, et al. response and outcome. J Clin Oncol 2004;
Radiation treatment planning with an integrated 22(11):2167–71.
positron emission and computer tomography [51] Lowe VJ, Boyd JH, Dunphy FR, et al. Surveillance
(PET/CT): a feasibility study. Int J Radiat Oncol for recurrent head and neck cancer using posi-
Biol Phys 2003;57(3):853–63. tron emission tomography. J Clin Oncol 2000;
[47] Gorres GW, Steinert HC, von Schulthess GK. 18(3):651–8.
PET and functional anatomic fusion imaging [52] Kumar R, Xiu Y, Potenta S, et al. 18F-FDG PET for
in lung and breast cancers. Cancer J 2004;10(4): evaluation of the treatment response in patients
251–61. with gastrointestinal tract lymphomas. J Nucl
[48] Kuo JS, Yu C, Petrovich Z, et al. The CyberKnife Med 2004;45(11):1796–803.
stereotactic radiosurgery system: description, in- [53] Larson SM, Schoder H, Yeung H. Positron
stallation, and an initial evaluation of use and emission tomography/computerized tomogra-
functionality. Neurosurgery 2003;53(5):1235–9. phy functional imaging of esophageal and colo-
[49] Rock JP, Ryu S, Yin FF, et al. The evolving role rectal cancer. Cancer J 2004;10(4):243–50.
51

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 51–70

Steroid Receptor Imaging in


Breast Cancer
Jean-Mathieu Beauregard, MD, Éric Turcotte, MD,
François Bénard, MD*

& Molecular biology of the estrogen and & Role of estrogen receptor/progesterone
progesterone receptors receptor expression imaging in the
& Transport and delivery of steroid hormones management of breast cancer
in plasma & Radioligands for steroid receptor imaging
& Prognostic value of estrogen receptor and Estrogen receptors
progesterone receptor expression in Progesterone receptors
breast cancer Clinical imaging studies with 18F-FES
& Current treatment strategies for estrogen & Summary
receptor/progesterone receptor positive & References
breast cancers

Many breast cancers express high levels of estro- being used as first- or second-line agents. Because
gen receptors (ER) or progesterone receptors (PR), these drugs are relatively well-tolerated by patients,
which have both been linked with the pathogene- with low toxicity compared with most chemotherapy
sis of this disease. Estrogen acts as a stimulant on regimen, hormone therapy is recommended to all
the growth of ER-positive breast tumors, and the postmenopausal women who have ER- or PR-positive
reduction of this stimulatory effect remains the breast cancer.
key strategy in the treatment of ER- or PR-positive Because ER and PR are overexpressed in 60% to
breast cancers, either by reducing estrogen produc- 80% of breast cancers, these proteins were identi-
tion or by antagonizing the effects of estrogens on fied many years ago as excellent targets for imag-
the ER. ing breast cancer with radiopharmaceuticals. There
Hormone therapy has shown promise in the pre- have been several efforts undertaken over the past
vention of breast cancer, and is routinely used as an 25 years to identify suitable agents for imaging
adjuvant treatment to complement surgery and ER or PR expression in breast cancers, with the goal
radiation therapy in the treatment of primary breast of providing noninvasive characterization of breast
cancer. About 50% to 60% of patients will respond cancers to guide hormone therapy. Some highly
to hormone therapy [1]. Tamoxifen, a partial estrogen effective ligands have been identified and well-
antagonist, remains the most widely used pharmaco- characterized, but ER imaging is still not yet used as
logical agent in routine clinical use, but aromatase a routine clinical procedure because of the limited
inhibitors and pure antiestrogens are increasingly number of available clinical trials.

This work was supported by grant #015388 from the Canadian Breast Cancer Research Alliance.
Metabolic and Functional Imaging Center, Clinical Research Center, Centre Hospitalier Universitaire de
Sherbrooke, 3001 12th Avenue N., Sherbrooke, Québec, J1H 5N4 Canada
* Corresponding author.
E-mail address: francois.benard@usherbrooke.ca (F. Bénard).

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.006
pet.theclinics.com
52 Beauregard et al

the state of phosphorylation of ER, which is influ-


Molecular biology of the estrogen and
enced by ligand binding [2].
progesterone receptors
Ligands can be classified as pure agonists,
ER are nuclear proteins, members of the nuclear (eg, estradiol), pure antagonists (eg, fulvestrant)
superfamily of transcription factors that mediate or mixed agonist-antagonists, also called selective
cell response to estrogens (or antiestrogens) by ER modulators, or SERMs (eg, tamoxifen or raloxi-
the regulation of the synthesis of specific RNAs fene). Pure agonist and antagonist ligands activate
and proteins [2]. Estrogens regulate, via ER, the or block (via conformational change in ER) both
development and function of the reproductive sys- AF1 and AF2, whereas SERMs have different effects
tem, and also affect many other organs. ER was first on each AF. The ability of a ligand to initiate or
identified in rat uterus in 1966 [3]. In 1986, the block gene transcription in a specific cell type de-
human ER was cloned and sequenced from MCF-7 pends on the dominance of ERα versus ERβ and
human cancer cells [4]. This ER is now known as AF1 versus AF2. Initiation of transcription is much
ERα, since the discovery in 1996 of a second ER more complex, because it involves the interaction
subtype, named ERβ [5]. of many proteins that act as coregulators (general
ERα is 595 amino acids long (66 kDa), whereas transcription factors, coactivators, corepressors,
ERβ is 530 amino acids long. Other isoforms of cointegrators, histone acetyltransferases, and his-
both ER subtypes have been described [6,7]. ERα tone deacetylases), further refining this process
and ERβ have the same structure, composed of six and making it even more cell-specific [8]. Acom-
functional domains that are responsible for specific plete review of these coregulators is beyond the
functions. The A/B domain is the least conserved scope of this article. Moreover, other nonclassical
between ERα and ERβ, and only ERα A/B domain pathways of gene regulation by ER (at different
contains the activation function 1 (AF1). The C DNA sites and with other coactivators) and non-
domain, or DNA binding domain (DBD), is the genomic ER actions (involving membrane ER out-
most conserved between the two subtypes, suggest- side the nucleus) are studied, and these could provide
ing they interact with the same genes. The D some explanations in drug-resistance mechanisms,
domain is called ‘‘hinge region,’’ and contains the particularly in the case of selective ER modula-
nuclear localization signal. E/F domain (ligand bind- tors [9].
ing domain, LBD) contains AF2, which is found in Both ER subtypes are found in brain, cardiovas-
both ER subtypes [2,6]. cular system, urogenital tract, and bone, in various
ER α and β have slightly different affinities for ratios. ERα predominates in the liver, whereas ERβ
their natural endogenous or exogenous ligands. Affi- is the main ER subtype in the colon [2,6]. In nor-
nity is often expressed as relative binding affinity mal breast tissue, both ERα and ERβ are expressed,
(RBA), which is 100 times the ratio between the but ERβ predominates. In premalignant lesions,
quantities of the reference ligand (eg, estradiol in however, there is an increase in ERα, which pro-
the case of ER) and the tested ligand needed to motes cell proliferation under estrogen stimulation,
displace 50% of the activity of radioactive reference and a decrease in ERβ expression, which is asso-
ligand (eg, tritiated estradiol) in a competitive bind- ciated with lower proliferation and pathological
ing assay on ER (either isolated receptors, cell ex- grade, and thought to downregulate ERα, acting
tracts, or cell culture). The estradiol RBA is, by as a tumor suppressor [7,8,10]. Thus ERα is the
default, 100 for both ER subtypes. The potency of therapeutic target of choice in breast cancer. ERα
different ER ligands depends, in part, on their RBA. is routinely dosed in vitro by immunohistochem-
The binding of a ligand to the ligand binding istry on breast cancer biopsy specimens for treat-
domain (LBD) of ER causes dissociation of protec- ment planning, as is discussed later. The clinical
tion proteins (called heat shock proteins) from ER, value of ERβ, however, is not established [7,10].
dimerization and conformational change of ER, PR are, like ER, members of the nuclear receptor
resulting in increased affinity of its DBD for the superfamily. Like ER, they are ligand-activated nu-
estrogen response elements (ERE). ERE are short clear proteins that regulate expression of specific
DNA sequences located in the promoter region subset of genes [8,11]. PR are found in the uterus
on genes, near the transcriptional sequence, that and many other organs, including the brain. One
enhance the transcriptional potential of a gene of the main roles of progesterone is to prepare
[2]. This is not sufficient to activate transcription, the uterus for pregnancy. Two isoforms of PR are
which is the role of activation functions AF1 and known: Progesterone receptor A (PRA) and proges-
AF2. The activity of each AF is dependent on the terone receptor B (PRB). They are encoded by the
target gene promoter context, cellular environment, same gene. PRB is a longer version of PRA contain-
and ligand [2,8]. ER DNA binding affinity and ing 164 additional amino acids. Both subtypes are
transcriptional activation are also modulated by normally found in the normal breast [8]. In malig-
Steroid Receptor Imaging in Breast Cancer 53

nant transformation, there is an alteration of the along with the nodal status, tumor size, patient age,
PRA:PRB ratio, and both PRA and PRB dominance and pathologic grade [17]. Although other prog-
have been reported [8,11,12]. Loss of PR expression nostic indicators, such as p53, HER2/neu status,
in breast cancer is associated with more aggressive angiogenesis, and proliferation indices have also
tumors and worse prognosis [8]. shown potential, the value of these parameters
has not been as clearly established. Breast tumors
that express ER grow more slowly, have a lower
Transport and delivery of steroid hormones
thymidine-labeling index, and typically have a more
in plasma
differentiated phenotype than ER-negative tumors
In women, progestins (progesterone and 17-hydroxy- [18]. The prevalence of ER-positive tumors increases
progesterone) and estrogens (estrone, estradiol- with patient age [19,20], and is significantly higher
17β, and estriol) are synthesized from cholesterol, in postmenopausal women (among whom 80%
mainly by the ovaries, and during pregnancy by the of patients have ER positive tumors) than pre-
placenta. These hormones, like other steroids, are menopausal women (among whom approximately
delivered to target organs by the blood. Because 50% of patients have ER-positive tumors) [17]. The
they are lipophilic, they enter the cell and reach short-term recurrence rate is also much lower in
the nucleus by passive diffusion. In blood, they are ER-positive than negative breast tumors.
bound to plasma proteins, mainly albumin and sex Conversely, the ER status does not correlate with
hormone-binding globulin (SHBG). Sex steroids the absence of nodal and distant metastases, and
bind with low affinity to albumin. Nevertheless, some studies have questioned whether the long-
this nonspecific binding is significant because of term survival of women who have ER-positive
the high concentration of albumin in plasma. tumors is really lower than those who have ER-
Like albumin, SHBG is synthesized mostly by the negative cancers [17]. Thus the presence of estrogen
liver. This glycoprotein plays a role in the transport receptors does not correlate with the metastatic
of sex steroids in plasma by allowing solubiliza- potential of breast tumors, but rather with their
tion of these lipophilic hormones and by protecting growth rate. The predictive value of ER, therefore,
them against metabolic inactivation [13]. SHBG more accurately reflects the short-term outcome
might also regulate the bioavailability of sex steroids, of these patients rather than the absolute disease-
because only the free (unbound) fraction is generally free survival.
considered available for cellular uptake [14]. The real value of measuring the expression of ste-
More recently, a cell-membrane SHBG receptor roid hormone receptors in breast cancers, howo-
has been discovered. This receptor is thought to par- ever, lies in their ability to predict a successful
ticipate in the delivery of estrogens into cells. Two response to hormonal therapy. A meta-analysis
mechanisms have been proposed: (1) the steroid- published in 1998 of the available evidence from
SHBG complex binds to the SHBG receptor and 55 studies that included 37 000 women [21]
then the steroid enters the cell [15], or (2) SHBG showed that 5 years of adjuvant therapy with ta-
binds to its receptor and then the free estrogen moxifen reduced recurrences by 47% in women
binds to the SHBG-receptor complex [16]. There- who had ER-positive primary tumors. No signifi-
after, either the estrogen diffuses passively inside the cant benefit could be proven in women who had
cell to reach ER in the nucleus, as mentioned above, ER-negative tumors.
or alternatively, the estrogen-SHBG-receptor com- Approximately 60% of previously untreated pa-
plex is internalized. It is still not clear how SHBG tients who have ER-positive tumors will respond
influences the delivery of steroids to their receptors. to hormonal therapy, whereas a response is ob-
It is currently difficult to ascertain whether the affin- served in only 5% to 10% of patients who have ER-
ity of estrogens and progestins for SHBG influ- negative tumors [17].
ences their biodistribution in humans. SHBG is
not expressed in small rodents used for preclinical
Current treatment strategies for estrogen
biodistribution studies of radiolabeled estrogens.
receptor/progesterone receptor positive
As it will be discussed later, this limits the extrapo-
breast cancers
lation of results from animal studies to humans.
Surgery and radiation therapy play important
roles in breast cancer treatment. Many reports have
Prognostic value of estrogen receptor and
shown that radiotherapy can significantly reduce
progesterone receptor expression in breast
the locoregional recurrence rate, but does not sig-
cancer
nificantly improve the long-term survival of the pa-
The hormone receptor status is one of the few well- tient [22,23]. Systemic adjuvant therapy, such as
established prognostic indicators in breast cancer, chemotherapy and hormonal therapy, has been
54 Beauregard et al

proven useful to reduce the rate of recurrence of receptor–positive breast cancer can also benefit from
breast cancers. aromatase inhibitors as adjuvant therapy. Anas-
When compared with hormone receptor-negative trozole and letrozole are nonsteroidal aromatase
tumors, hormone receptor-positive breast cancers inhibitors compared with exemestane, which is a
exhibit stronger clinical responses to hormonal treat- steroidal aromatase inactivator. The main effect of
ment. Adjuvant tamoxifen prolonged both disease- aromatase inhibitors is to decrease the concentra-
free and overall survival in patients who had tion of circulating estrogen levels in postmeno-
ER-rich tumors, but had little benefit in patients pausal women. Aromatase inhibitors should not
who had ER-negative neoplasms [21]. According be employed as monotherapy in premenopausal
to Harvey and colleagues [24], patients who have women because estrogen suppression increases
breast cancers containing as few as 1% to 10% of gonadotrophin release by feedback mechanism, in
cells staining for ER respond to hormone therapy. return increasing the production of estrogen by the
Patients who are ER-negative but PR-positive can ovaries. Both anastrozole and letrozole have been
also benefit from tamoxifen, but the role of the shown to be equivalent or superior to tamoxifen in
progesterone status in selecting therapy remains a variety of clinical parameters, including the treat-
controversial. In 2003, Bardou and coworkers [25] ment of metastatic breast cancer [33,34]. In a ran-
showed that the combined measurement of ER and domized unblinded trial comparing anastrozole
PR is superior to ER alone in predicting benefit and letrozole in patients who had metastatic breast
from adjuvant hormone therapy, because the pre- cancer [35], there was no significant difference
sence of PR enhances the ER predictive value in between the two agents, even if letrozole was
both pre- and postmenopausal patients. known to have a more pronounced effect on re-
Tamoxifen is accepted as the standard treatment ducing estrogen levels. Letrozole was associated
for patients whose tumors express the estrogen or with a higher overall response rate, but this was
progesterone receptors [26]—regardless of age, meno- not statistically significant. The two agents did not
pausal status, axillary node involvement, or tumor differ significantly in terms of clinical benefit rate
size [21,27,28]—to increase recurrence-free survival or overall survival. Both aromatase inhibitors were
and overall survival irrespective of whether che- associated with significantly fewer endometrial can-
motherapy has been given [21]. According to trials, cers and venous and arterial vascular events com-
5 years of tamoxifen treatment appeared supe- pared with tamoxifen [36,37]. Although this was
rior to 2 years. The beneficial effect of 5 years of not statistically significant, both anastrozole and
adjuvant tamoxifen was demonstrated in both letrozole were associated with a higher incidence
receptor-positive postmenopausal patients [29] and of cardiovascular disease [37,38].
premenopausal patients under 50 years of age [21]. Results of the Arimidex, Tamoxifen, Alone or
Currently, there are no data justifying the use of ta- in Combination (ATAC) trial after completion of
moxifen for a longer period than 5 years [21,30,31], 5 years’ adjuvant treatment for breast cancer have
but trials are still ongoing to address this question. been published recently [39], and propose anastro-
Tamoxifen combined with chemotherapy further re- zole for 5 years to be the preferred initial adju-
duces the risk of disease recurrence, particularly in vant treatment over tamoxifen for postmenopausal
premenopausal women [21,30]. women having localized, hormone-receptor–positive
According to Colleoni and colleagues [32], cyclo- breast cancer. This recommendation is based on a
phosphamide, methotrexate and 5 fluorouracil significantly increase disease-free survival (hazard ra-
(CMF) combination chemotherapy given concur- tio [HR] 0.87), greater time to recurrence (HR 0.79),
rently (early, delayed, or both) with tamoxifen was greater time to distant recurrence (HR 0.86), and
more effective than tamoxifen alone for patients reduced distant metastases (HR 0.86). Overall sur-
who had node-positive, endocrine-responsive breast vival rates were similar in the two groups, however,
cancer, supporting late administration of chemo- despite a death reduction by 12% in the anastro-
therapy even after initiation of tamoxifen. In con- zole group that was not statistically significant.
trast, sequential CMF and tamoxifen for patients Aromatase inhibitors have not been evaluated in
who had node-negative, endocrine-responsive breast the adjuvant setting in women whose tumors lack
cancer was ineffective [32]. Thus all patients who hormone receptors. The Canadian MA-17 trial [38]
have hormone-receptor positive tumors receiving is so far the only study that could demonstrate a
adjuvant chemotherapy should also receive tamoxi- survival benefit for node-positive disease by extend-
fen, but the optimal timing between chemotherapy ing 5 years adjuvant therapy of tamoxifen with
and tamoxifen (during or after chemotherapy) is still letrozole in ER-positive postmenopausal women
under investigation [21,30]. [38,40,41]. Postmenopausal women concluding
Based on results from multiple large randomized 2 to 3 years of tamoxifen therapy may consider
trials, postmenopausal women who have hormone- crossover to an aromatase inhibitor, and should
Steroid Receptor Imaging in Breast Cancer 55

plan on a total of 5 years of adjuvant endocrine ther- likely to derive benefit from treatment with CMF-
apy. The optimal timing of transition between based regimes compared with no treatment. Several
tamoxifen to the aromatase inhibitor and the opti- studies suggest that overexpression of this gene may
mal duration of aromatase inhibition are still under predict enhanced sensitivity to anthracycline-based
investigation. According to the American Society of regimens in the adjuvant setting [55–57]. Thus the
Clinical Oncology (ASCO) [42], there are no data available data suggest that patients who have HER-2/
on the use of tamoxifen after an aromatase inhibitor neu–positive cancers are more likely to respond to
in the adjuvant setting. Patients intolerant to aroma- anthracycline regimens than HER-2/neu–negative.
tase inhibitors should receive tamoxifen as adjuvant According to the ASCO, the use of HER-2/neu to
therapy, and women who have hormone-receptor– decide whether to prescribe endocrine therapy either
negative tumors should not receive adjuvant endo- in the adjuvant or metastatic setting is not recom-
crine therapy. mended. HER-2/neu may identify patients who par-
Fulvestrant is one of a new type of ER antagonists ticularly benefit from anthracycline-based adjuvant
that bind to estrogen receptor monomers, inhibit therapy, but levels of HER-2/neu should not be
receptor dimerization [43], disable activating func- used to exclude patients from this type of treatment
tion AF1 and AF2 [44], reduce translocation of [42]. For postmenopausal women who have breast
receptors to the nucleus, and accelerate degrada- cancer overexpressing HER-2/neu, higher response
tion of the ER, ultimately resulting in a reduction rates have been reported for aromatase inhibitors
in cellular ER [45,46]. This results in pure anti- compared with tamoxifen [59,60]; however, the
estrogenic effects [47], and unlike tamoxifen, ful- role of HER-2/neu status in selecting optimal endo-
vestrant is devoid of any known agonist activity crine therapy remains controversial and needs fur-
[46]. Fulvestrant has a steroidal structure that com- ther investigations.
petitively binds to the ER with an affinity much
greater than that of tamoxifen (~100 times) [46].
Many trials are ongoing to evaluate the clinical effi-
Role of estrogen receptor/progesterone
cacy of fulvestrant after recurrence or progression
receptor expression imaging in the
on a nonsteroidal aromatase inhibitor. Current re-
management of breast cancer
sults suggest that fulvestrant is at least as effective as
anastrozole [48]. Fulvestrant has been approved for The variability of ER/PR measurements across labo-
clinical use in the United States for the treatment of ratories using radioligand binding assays on tissue
ER-positive metastatic breast cancer that fails to extracts, the heterogeneity of tumor sampling [61],
respond to tamoxifen. The efficacy of fulvestrant in and the limitations of the technique to provide
sequential endocrine therapy and in combination accurate results using very small samples were all
with other agents appears promising, and active inves- important factors that motivated the development
tigations are ongoing to explore the clinical potential of nuclear imaging to characterize the ER status of
of this medication. breast cancers. The use of immunohistochemistry
New markers to predict response to tamoxifen are to assess the steroid receptor status of breast cancers
continuously being evaluated, and several investi- has overcome many of the limitations of radioli-
gators have focused their attention on HER-2/neu gand binding assays, and has found widespread
[49–53]. In breast cell lines and in model tumor acceptance. Because the presence of estrogen and
systems, overexpression of the HER-2/neu gene has progesterone receptors is routinely assessed at pre-
been associated with increased mitogenesis, malig- sentation from small specimens, there is little jus-
nant transformation, increased cell motility, inva- tification to perform an imaging procedure that
sion, and metastasis [54]. In human breast cancer, would duplicate this information. Because up to
amplification of the HER-2/neu gene is found in 20% of postmenopausal and 50% of premeno-
15% to 30% of primary invasive tumors. For pa- pausal women who have breast cancer have tumors
tients who have both early and advanced disease, that do not exhibit ER-positivity, radioligand-based
overexpression of HER-2/neu correlates with either imaging methods would miss an unacceptably high
relative resistance or adverse outcome after treat- number of breast cancers if used for screening or
ment with hormonal therapy [55,56]. The relation- diagnostic purposes.
ship between HER-2/neu expression and response Although the status of ER and PR receptors in
to chemotherapy in breast cancers appears to de- primary breast cancers is usually well established at
pend on the type of drug administered. With adju- the time of diagnosis with the routine use of immu-
vant CMF, the majority of studies showed a nohistochemistry, this is not always the case at the
reduced benefit in HER-2/neu–positive compared time of recurrence. A biopsy of a recurrent lesion is
with negative patients [56–58]; however, patients always recommended, because the ER/PR status of
who have cancers overexpressing HER-2/neu are the recurrence can then be documented, and the
56 Beauregard et al

Fig. 1. (A–F ) Chemical structures of some estrogen-binding radiopharmaceuticals used in clinical studies. The
structure of the endogenous hormone, 17-ß-estradiol, is shown in A.

status of other antigens with important prognostic radiopharmaceuticals that have been proposed for
and therapeutic value, such as HER2/neu, can also this purpose.
be obtained on the specimen. Not all sites of recur-
rences are easily amenable to biopsy, however, and
heterogeneity of ER/PR expression across metastatic Radioligands for steroid receptor imaging
sites could have strong prognostic implications.
Furthermore, the mechanisms of hormone therapy Estrogen receptors
resistance are multiple and complex, with loss of Iodinated ligands
expression of ER being just one of multiple path- 16αα-Iodoestradiol Hochberg synthesized 16α-
ways by which malignant breast cancer cells [125I]iodoestradiol in 1979 [66]. This compound
develop resistance. Only about 20% to 40% of pa- showed specific ER-mediated uptake in vivo, in
tients who have a primary tumor that expressed rat uterus. It thus became one of the first poten-
ER will have lost ER expression at the time of re- tial single photon-emitting ER imaging agent, be-
currence [62–64]. With resistance acquired during cause it could be labeled with either 131I or 123I [see
hormone therapy, 28% of patients will lose ER ex- Fig. 1B]. 16α-[123I]iodoestradiol (123I-E2) could
pression in their tumors [65]. With the availability bind to ER-positive tissues in animals, including
of second-line agents with proven efficacy even 7,12-dimethylbenz[a]anthracene (DMBA)-induced
when tamoxifen has failed, the accurate assessment mammary tumors [67]. In a series of 21 patients
of the status of ER expression gains even more who had breast masses [68], 123I-E2 was positive in
importance. A diagnostic test that can provide func- 7 of 9 patients who had ER-positive malignant tu-
tional information about the status of ER or PR mors, and negative in all 12 ER-negative tumors (in-
across all tumor sites could have profound prog- cluding 7 benign tumors). Kenady and coworkers
nostic implications and influence the decision to [69] reported uptake in chest wall tumors and in-
treat individual patients with hormone therapy or flammatory breast cancer. In another study [70], in
chemotherapy. Fig. 1 illustrates several ER-binding 42 patients, sensitivity of 123I-E2 for ER+ tumors
Steroid Receptor Imaging in Breast Cancer 57

was 66%. For imaging purpose, 131I had less desir- Van de Wiele and coworkers [98] imaged nine pa-
able physical characteristics than 123I, which ac- tients who had primary breast cancer using [123I]
counted for disappointing results obtained with iodomethyl-N,N-dimethyltamoxifen (123ITX) and
131
I-E2 [71]. Rapid metabolism of I-E2 appears to SPECT. 123ITX lacked sensitivity—only four pa-
prevent its optimal localization in ER-positive tu- tients out of six who had ER+/PR+ tumors had
mors [71,72]. positive 123ITX scintigraphy. The three other pa-
tients (two who had ER+/PR-, and one who had
Z-MIVE Ali and colleagues [73] reported synthe-
ER-/PR- tumors) had negative 123ITX studies. Sen-
sis of (17α,20Z)-[125I]iodoestradiol, which showed
sitivity was also low for detecting nodal and dis-
higher ER-mediated uptake in target tissue than its
tant metastasis.
20E isomer. They also reported that the addition
of a 11β-methoxy substituent to these isomers,
Tc-99m ligands
producing 11β-methoxy-(17α,20E/Z)-[125I]iodovi-
Because of its wider availability, lower cost, and
nylestradiol (E- and Z-MIVE, see Fig. 1C), which
convenient physical characteristics, Tc-99m (140 keV,
increased their in vivo specific uptake [74]. Both
6 hours half-life), would be a very desirable single-
E- and Z-MIVE were labeled with I-123 [75]. Ribeiro-
photon emitting label for ER imaging agents.
Barras and coworkers [76] chose E-MIVE for the
Skaddan and colleagues [99] described the syn-
first clinical study, but only 2 of their 4 patients
thesis of four estradiol complexes, labeled with
who had high ER content primary breast cancers
Tc-99m at position 7α, along with their bio-
had positive E-MIVE uptake on single photon emis-
distribution in rodents. These compounds had
sion computed tomography (SPECT) imaging. Rijks
poor ER-mediated uptake in target organs, along
and colleagues [77,78] preferred Z-MIVE over E-MIVE
with high nontarget organs uptake. Others reported
on the basis of their animal experiments. In 25 pa-
on Tc-99m labeled estrogens, including tamoxi-
tients imaged with Z-MIVE SPECT, there was 90%
fen conjugates [100,101]. Again, low uterus uptake
agreement (9/10 patients, 1 false-positive) between
was observed.
SPECT and ER status for primary breast cancer,
Single-photon emitting ER ligands are advanta-
and 82% agreement (9/11 patients, 1 false-positive,
geous because they can be readily used in any nu-
1 false-negative) in metastatic/recurrent breast can-
clear medicine department, using both planar and
cer [79]. The Sherbrooke group [80,81] achieved
SPECT imaging protocols; however, iodine-123, the
similar results (80% agreement, 2 false-positive)
current label of choice, remains expensive. With the
using Z-MIVE scintimammography in 10 patients
increasing availability of positron emission tomog-
who had primary breast cancer. Bennink and co-
raphy (PET) scanners in many major cancer centers,
workers [82] reported a sensitivity of 94% for planar
PET is an attractive alternative to SPECT because of
scintigraphy and 100% for SPECT imaging in
its superior spatial resolution and sensitivity over
25 patients who had primary breast cancer (includ-
traditional nuclear medicine techniques. Not sur-
ing 10 patients from reference [79]). Thus Z-MIVE
prisingly, several ligands were developed for ER
appears to be more sensitive than E-MIVE for ER
imaging with positron emitters.
imaging. Recently, it has been demonstrated that
Z-MIVE can predict response to hormonal therapy.
C-11 ligands
Among 21 patients who had clear Z-MIVE uptake in
Carbon-11 can be used to label molecules without
their metastatic lesions, the 17 patients who had
introducing a halogen or other foreign atoms, mak-
complete blockade of uptake 1 month after starting
ing their structure closer, if not identical, to en-
tamoxifen had a longer progression-free survival than
dogenous ligands; however, the radiosynthesis is
the 4 others [83].
generally more difficult because the labeling reaction
Other estrogens labeled with single photon is easily contaminated with cold carbon, reducing
emitting halogens Ali and colleagues [84–88] and specific activity. Because specific activity generally
others [89,90] reported synthesis and biologi- decreases at the same rate as activity decays, this
cal activity study of many other iodine and bro- may further reduce the sensitivity for imaging satu-
mine radiolabeled steroidal estrogens. Of these, rable receptor systems, like ER. The short half-life
only 16α-[77Br]bromoestradiol has been studied of C-11 (20 minutes) also prevents the distribution
in humans, but the images suffered from the poor of this radionuclide to distant sites. Nevertheless,
imaging characteristics of bromine-77 [91,92]. Radio- several reports C-11 labeled estrogens have been
iodinated tamoxifen analogs have been synthesized published [102–104]. These tracers showed poor to
[93–98]. Imaging breast cancer in humans with [131I] moderate receptor-mediated target organ uptake and
iodotamoxifen was unsuccessful [93]. The high lipo- target/nontarget ratios in biodistribution studies in
philicity of this molecule resulted in high nonspe- rodents, however. Tamoxifen was also labeled with
cific uptake and reduced target-to-nontarget ratio. C-11, but specific activity was low [105].
58 Beauregard et al

F-18 ligands sulfate as precursor [111], and methods have


Fluorine-18 is a useful radioisotope for PET ER been published to automate this synthesis [112].
imaging: it has a convenient half-life (allowing Several groups now use this synthesis as a rou-
off-site radiosynthesis), and its presence on the tine procedure.
ligands is well-tolerated by the ER (particularly at
16β-fluoromoxestrol To improve the uptake char-
the 16α position). It is relatively easy to radio-
acteristics of FES, many substituted FES derivatives
fluorinate estrogens with specific activities over
have been synthesized. 11β-ethyl, 11β-methoxy and
1000 Ci/mmol.
17α-ethynyl substitutions were synthesized in both
16α- [113] and 16β-FES [114]. These derivatives have
16α-[18F]fluoroestradiol In 1984, Kiesewetter and
been studied in vitro (ER and SHBG RBA) and in
coworkers [106] published the preparation of
vivo (biodistribution in Sprague-Dawley female
16α-[18F]fluoroestradiol (18F-FES, see Fig. 1D),
rats). Among these compounds, 17α-ethynyl-11β-
along with other fluorine-18 labeled estrogens
methoxy-16β-fluoroestradiol (16β-fluoromoxestrol,
(16β-[18F]fluoroestradiol, 1-[18F]fluoropentestrol,
βFMOX) was the most promising ligand (see
1-[18F]fluorohexestrol). Their RBAs for ER were 80,
Fig. 1E), with a very good ER relative binding affinity
30, 129 ,and 127, respectively. All these compounds
(78 at 25°C), very low SHBG relative binding affinity
showed ER mediated uptake in vivo in immature
(0.037), high uterus uptake (18.26 ± 7.850% I.D./g),
female rats, manifested by uterus uptake that can
and high uterus/blood and uterus/muscle ratios
be blocked by excess estradiol co-injection. In this
(66.26 ± 9.38 and 24.31 ± 6.14, respectively)
biodistribution study, 18F-FES showed the high-
[114,115]. These encouraging results were attri-
est selectivity for ER in vivo, manifested by the high-
buted to the greater metabolic stability of βFMOX
est uterus/blood and uterus/nontarget ratios over
over 16α-FES in vivo, contributing to high specific up-
2 hours (39 ± 16 and 28 ± 4.8, respectively, 1 hour
take and lower background activity from metabolites.
postinjection of 50 μCi of 18F-FES). 18F-FES also
These improved characteristics over 16α-FES led
had the highest blood clearance [106]. Good 18F-FES
to the first clinical study with βFMOX in 12 patients
uptake in DMBA-induced mammary tumors was
who had breast masses [116]. Only 3 of these
shown, although this uptake did not correlate
patients had biopsy-confirmed ER-positive malig-
well with the ER content of these tumors [107].
nant tumors. Unfortunately, βFMOX-PET was nega-
Mintun and colleagues [108] published the re-
tive in these 3 cases. These disappointing results
sults of the first clinical study with 18F-FES in 1988.
were attributed to the very low affinity of βFMOX
Thirteen patients who had breast masses under-
for SHBG. This supports the hypothesis that SHBG
went 18F-FES PET before biopsy. Except for one
favors steroid binding to ER in vivo in humans, by
benign lesion and one mass that was out of field
protecting estrogens from metabolism [13] and
of view, 18F-FES PET showed elevated uptake in all
facilitating their uptake via a cell-surface SHBG
other breast masses proven to be breast carcinoma,
receptor [16]. The authors compared FES and
and in some axillary metastases as well [108]. Since
βFMOX metabolic rates in rat, primate and hu-
then, 18F-FES has been considered the ER imaging
man isolated females hepatocytes. Results were
agent of choice.
consistent with a protective effect of sex hormone-
Mankoff and coworkers [109] established the ra-
binding globulin (SHBG) [116].
diation dosimetry in 18F-FES PET studies of 49 pa-
tients. The effective dose equivalent was 0.022 mSv/ 4-fluoro-11β-methoxy-fluoroestradiol In an effort
MBq, which would give 4.8 mSv for a typical activ- to improve metabolic stability of FES, the authors’
ity of 220 MBq (6 mCi). The critical organ was the group described the synthesis of a new series of
liver (0.13 mGy/MBq). Thus the radiation dose is fluorine-18 labeled estrogens, the 2- and 4-fluoro
reasonable and well below generally accepted limits derivatives of 16α-18F-FES, with or without a 11β-
for research studies. methoxy group [117]. Biodistribution studies in
Metabolism studies of 18F-FES in both animals immature Sprague-Dawley female rats identified
[107] and humans [110] showed that 18F-FES is 4-fluoro-11β-methoxy-fluoroestradiol (4F-MFES) as
metabolized rapidly, leaving conjugated circulating the most promising of these potential ER imag-
labeled metabolites that do not bind to ER, increas- ing agents [118]. Despite a more than fourfold
ing background activity. This rapid metabolism reduction of 4F-MFES relative binding affinity for
is also manifested by a rapid blood clearance of ER when compared with 18F-FES (on both isolated
18
F-FES, leaving only 20% of unmetabolized ERα and MCF7 cells), 4F-MFES achieved the highest
18
F-FES 20 minutes after injection [110], thus po- uterus uptake (14.66 ± 3.22% ID/g) and highest
tentially limiting the maximum achievable uptake uterus/blood and uterus/nontarget ratios (136.32 ±
in ER-containing cells. The F-18 labeling method 22.00 and 47.65 ± 7.23, respectively) at 1 hour
of 18F-FES has been improved by using a cyclic postinjection [118]. The performance of 4F-MFES
Table 1: Comparative characteristics of selected radiolabeled estrogens
RBA (estradiol = 100) Uterus uptakeb Ratios at 1 h
a d
ER SHBG 1h 1 h (+ E2 ) (uterus/blood) (uterus/nontargete) Reference
f
FES 80 – 4.67 ± 1.50 0.61 ± 0.12 39 ± 16 28 ± 4.8 [106]
FES 107.7 ± 26 4.5 ± 1 11.75 ± 3.20 0.70 ± 0.11 66.41 ± 20.44 36.10 ± 6.85 [118]
FES 54 9.47 – – – – [115]
βFMOX 78 0.037 18.26 ± 6.80 2.548 ± 0.593 66.27 ± 9.38 24.31 ± 6.14g [115]
4F-MFES 24.4 ± 5 <0.0017 14.66 ± 3.22 1.14 ± 0.29 136.32 ± 22.00 47.65 ± 7.23 [118]
Z-MIVE 32.0 ± 6.6 – 17.49 (15)c 1.87 (17)c 22.71 13.81 [74]

Abbreviations: CV, coefficient of variation; ID/g, percent injected dose per gram of tissue; SD, standard deviation.
a
at 25°C or room temperature.
b
% ID/g ± SD 1 hour after 50 μCi injection in immature female Sprague-Dawley rats.
c
% ID/g (% CV) 1 hour after 3 μCi injection in immature female Fischer rats.
d
coinjection of excess unlabeled estradiol.
e
nontarget organs: lung, spleen and muscle.
f
nontarget organs: oesophagus, lung, spleen and muscle.
g
nontarget organ: muscle.
Steroid Receptor Imaging in Breast Cancer
59
60 Beauregard et al

Fig. 2. (A–C ) Chemical structure of progesterone and other analogs.

relative to other ER-binding radiopharmaceuticals Fluorotamoxifen Because the ultimate goal of


is summarized in Table 1. Higher binding selec- imaging ER in breast cancer is to guide the choice
tivity, greater ability to cross membrane barriers, of therapy, it appears attractive to use radiolabeled
and reduced interaction with metabolic enzymes forms of SERM drugs used in breast cancer treat-
could all explain the superior localization properties ment. To date, Tamoxifen remains the most widely
of 4F-MFES. Biodistribution studies in Balb/c mice used antiestrogen drug for this purpose. The syn-
bearing MC7-L1 and MC4-L2 ER+ murine breast thesis of a fluorine-18 labeled derivative of ta-
cancer tumors also showed high uptake of 4F-MFES moxifen, [18F]fluoromethyl-N,N-dimethyltamoxifen
in these tumors. This uptake was ER mediated be- (18FTX), was described by Yang and colleagues
cause it could be blocked by coinjection of excess [96]. Biodistribution studies of this compound in
estradiol [119]. rats showed some ER-mediated uptake in both
4F-MFES has very low relative binding affinity for uterus and inoculated mammary tumors, although
SHBG (< 0.0017), like the other 11β-methoxy de- there was only slight to moderate (less than 50%)
rivatives in this series [118] and like, as discussed decrease in uptake when blocking with an excess
above, βFMOX. Because the role of SHBG in trans- of cold estradiol. Also, the absolute uptake in the
port and metabolism of steroids is not completely target organs was rather low compared with the
elucidated, we cannot predict how 4F-MFES will very high uptake in liver, lung, and kidney [96]. A
behave in human. Accordingly, 4F-FMES remains clinical study was conducted in 10 patients who
a promising but speculative ER imaging PET agent. had ER-positive breast tumors [120]. 18FTX PET was
A Phase I clinical trial is currently underway in the able to detect 16 of the 20 lesions (in five of seven
authors’ center with this agent. patients) considered to be truly positive for breast

Fig. 3. Typical 18F-FDG (A) and 18F-FES (B) maximum-intensity projection images in a patient with recurrent breast
cancer and multiple bone metastases. Most of the disease sites are concordant between both tracers. Note the
high liver, biliary tract, and bowel caused by excretion of 18F-FES metabolites.
Steroid Receptor Imaging in Breast Cancer 61

Fig. 4. Discordant 18F-FDG (A) and 18F-FES (B) findings in a patient with recurrent breast cancer in the left axilla.
Although the primary tumor was ER-positive, a biopsy of a left axillary lymph node confirmed that the recurrent
disease no longer expressed ER. The panels show coronal, sagittal, and transaxial images from left to right.

cancer. Equivocal lesions on 18FTX PET were in- response, and the other nonresponder had mixed
cluded in the positive results. 18FTX PET predicted results on PET [120]. Similarly to what was ob-
response to tamoxifen therapy after the scan in five served in the animal studies, markedly high up-
of six patients: three had positive lesions and a take in liver, lung, and heart was observed. This
good response, two had negative lesions and poor nonspecific uptake, attributable to the high lipo-

Fig. 5. 18F-FDG PET maximum-intensity projection images in a patient with diffuse bone metastasis (A). The 18F-FES
PET images were negative in this patient (B). Note the uptake of both 18F-FDG and 18F-FES in the apex of the
left lung, which is the site of radiation pneumonitis.
62 Beauregard et al

compounds have been synthesized for ER imaging


[90,121,122]. The authors’ group recently reported
the synthesis three fluorine-18 labeled analogs of
fulvestrant, a pure ER antagonist [123]; however,
the fluorine substitution at position 16α reduced
considerably the RBA (16α-fluoro-fulvestrant = 1.01;
fulvestrant = 89; estradiol = 100). Likewise, low ER-
mediated uptake was observed in immature female
rats [123].

Progesterone receptors
21-[18F]fluoro-16α-ethyl-norprogesterone ([18F]FENP)
was the first progestin labeled with fluorine-18
[124], and is the most studied positron emitting
PR imaging agent to date [Fig. 2]. Its RBA value of
6000 (progesterone = 100), and its highly selective
uptake in rat uterus were encouraging. It is the only
PR radiopharmaceutical studied in humans to date
Fig. 6. 18F-FES PET study (anterior projection image) of [125]. This study included eight patients who had
a patient. The dose of 18F-FES was injected in a port-a-
primary breast carcinoma. [18F]FENP PET was only
cath, with retention of the tracer in the tubing.
50% accurate in identifying PR-positive primary
tumors (3/6 patients). Rapid metabolism, mani-
philicity of 18FTX and its liver metabolism, contrib-
fested by high bone (defluorination) and hepatic
uted to reduce its sensitivity, particularly for tho-
uptake, contributed to these disappointing results.
racic lesions.
A few other fluorine-18 labeled progestins have
Other fluorinated PET ER imaging agents Many been synthesized and studied in small animals
other flurine-18 labeled steroidal and nonsteroidal [126]. Radioiodinated PR imaging agents have

Fig. 7. Patient with recurrent breast cancer manifested by serum tumor marker CA-125 elevation. The 18F-FDG PET
images showed subtle bone metastases (A – coronal and sagittal images; left), that are much more numerous
and obvious on the 18F-FES images (A – right). During aromatase inhibitor therapy, the 18F-FDG-PET study became
almost completely negative (B, left), while extensive residual disease was still apparent of 18F-FES PET (B, right).
Steroid Receptor Imaging in Breast Cancer 63

been proposed by Ali and coworkers [127] and Mankoff and coworkers [133] showed that
18
others [128]. Tc-99m labeled progestins are re- F-FES uptake could be heterogeneous across
viewed in reference [129]. tumor sites in a given patient, with a coefficient
of variation on the order of 30%. They also showed
18
that 18F-FES uptake tended to be higher in post-
Clinical imaging studies with F-FES menopausal patients who had low serum estradiol
As discussed above, Mintun and colleagues [108] and SHBG levels, and in patients who received
showed that 18F-FES uptake could detect ER-positive prior hormonal therapy. Recently, the same group
breast tumors in a series of patients who had rela- showed in a study conducted on 46 patients who
tively large primary tumors. They observed a good had ER-positive primary and recurrent breast can-
correlation (r = 0.96) between ER concentration cers [134] that 18F-FES uptake could predict re-
and the uptake of 18F-FES. These results were sponse to hormonal therapy (mostly aromatase
further supported in a subsequent study of 16 pa- inhibitors) depending on the HER2/neu status of
tients who had recurrent or metastatic breast the tumors. 18F-FES uptake was predictive of re-
carcinoma eligible to antiestrogen therapy [130]. sponse in HER2/neu-negative patients, but could not
18
F-FES uptake was seen in 14 of 16 patients and predict the outcome when HER2/neu was positive.
53 of 57 known lesions. Interestingly, three of In practical terms, 18F-FES is usually prepared in
the four false-negative lesions occurred in 2 pa- a saline solution containing from 5% to 10% of
tients injected with 18F-FES of low specific activity ethanol. 18F-FES can also be prepared in a lipid
(278 and 230 Ci/mmol). This study showed that emulsion without significant alterations in the bio-
18
F-FES could also assess ER expression in nodal
and distant metastases. Dehdashti and coworkers
[131] reported a good 88% agreement between
estrogen receptor status determined by 18F-FES imag-
ing and in-vitro analysis of biopsy samples. Their
study included 32 women who had primary breast
lesions and 21 patients who had recurrent breast
cancer. In primary breast tumors, they observed an
agreement of 82%. None of the benign breast
lesions incorporated 18F-FES. In recurrent or meta-
static sites, the rate of agreement between in vitro
results and 18F-FES-PET was 94%. They observed
intrapatient discordance (some lesions incorpo-
rating 18F-FES whereas others did not) in 2 out of
13 18F-FES positive patients in whom multiple sites
were evaluated, illustrating possible ER expression het-
erogeneity across metastatic sites. No correlation was
observed visually or quantitatively between 18F-FES
and 18F-Fluorodeoxyglucose (18F-FDG) uptake.
Mortimer and colleagues [132] reported a study
with combined 18F-FDG and 18F-FES performed
before and after treatment with tamoxifen to detect
hormone-induced changes in tumor metabolism
(metabolic flare), changes in available levels of
estrogen receptor (ER), and to correlate if those find-
ings would predict hormonally responsive breast
cancer. The 18F-FES uptake values were higher on
average for responders than nonresponders, but
there was considerable overlap between baseline
18
F-FES uptake values of responders and nonrespon-
ders. A successful response to tamoxifen was asso- Fig. 8. Patient recurrent metastatic breast cancer in
ciated with ER blockage confirmed by 18F-FES and a the mediastinum and bones. The 18F-FDG PET study
metabolic ‘‘flare’’ response corresponding to an (A) was obtained before (left) and after two months
of aromatase inhibitors (right), with a moderate re-
increased 18F-FDG uptake 7 to 10 days after the
duction in the metabolic activity of the lesions. The
initiation of tamoxifen. Results of PET were predic-
decrease in 18F-FES uptake (B) was also significant,
tive of responsiveness to tamoxifen therapy in pa- although more disease sites were still apparent on
tients who had advanced ER-positive breast cancer. the 18F-FES study after 2 months of therapy.
64 Beauregard et al

distribution of this compound, but there was a versely, loss of ER expression is demonstrated
trend for slightly higher tumor uptake with the by a complete lack of 18F-FES uptake at disease
ethanol formulation in animal studies [119]. The sites identified on 18F-FDG PET images [Fig. 4].
typical injected activity is 6 mCi (220 MBq). Although the uptake of 18F-FES is generally highly
18
F-FES uptake reaches a plateau between 60 to specific, some inflammatory lesions such as radia-
90 minutes after injection, and remains stable for tion pneumonitis can accumulate low amounts of
up to 2 hours. 18F-FES is rapidly metabolized from this radiopharmaceutical [Fig. 5]. Injections into
the plasma by the liver, and there is intense liver implanted central venous lines (such as a port-
and biliary excretion that interferes with the evalua- a-cath; Smiths Medical Canada Ltd., Markham, On-
tion of lesions in the abdomen and pelvis. 18F-FES tario) should be avoided, because this lipophilic
is also partly excreted by the kidneys, and the compound tends to stick to tubing and can inter-
bladder is usually apparent in the images. Apart fere with lesion visualization in the thorax [Fig. 6].
from these two excretory pathways, there is little In most cases, the distribution of 18F-FES will cor-
background activity in other organs. The brain respond to sites of disease that are highlighted on
takes up 18F-FES early after injection, presumably 18
F-FDG PET imaging [see Fig. 3], but the extent of
by blood-flow–related diffusion, and washes out disease is sometimes much more pronounced on
progressively afterwards, with minimal residual 18
F-FES images [Fig. 7A]. In general, the response
uptake on delayed images. The respective biodistri- to aromatase inhibitors is much more apparent on
bution of 18F-FES relative to 18F-FDG is illustrated 18
F-FDG PET studies [see Fig. 7; Fig. 8], and despite
in Fig. 3, which shows several sites of recurrent an almost complete resolution of 18F-FDG uptake,
breast cancer that express estrogen receptors. Con- residual tumors can still be depicted under ther-

Fig. 9. Cross-sectional images of a patient with an isolated site of recurrent disease in the sternum, obtained on
a PET/CT scanner. The recurrence in the sternum is readily identified on the 18F-FDG PET study (A). The presence
of estrogen receptors in that lesion is also obvious on the 18F-FES PET study obtained the following week (B).
(C ) shows the fused 18F-FES and CT studies.
Steroid Receptor Imaging in Breast Cancer 65

apy by 18F-FES imaging [see Fig. 7B]. Tamoxifen estrogen receptors. In: Jucker E, editor. Progress
typically blocks 18F-FES uptake by ER; in this case, in drug research, vol 59. Basel (Switzerland):
18 Birkhäuser Verlag; 2002. p. 201–32.
F-FES is useful in demonstrating the successful
blockage of the receptors by ER antagonists [83,132]. [3] Toft D, Gorski J. A receptor molecule for es-
With the availability of PET/CT, the interpreta- trogens: isolation from the rat uterus and pre-
liminary characterization. Proc Natl Acad Sci
tion of 18F-FDG and 18F-FES PET studies is greatly
USA 1966;55(6):1574–81.
facilitated to identify the precise sites of disease [4] Greene GL, Gilna P, Waterfield M, et al.
involvement [Fig. 9]. The CT component could Sequence and expression of human estrogen
improve the accuracy of these tests by identify- receptor complementary DNA. Science 1986;
ing some small pulmonary metastases and FES- or 231(4742):1150–4.
FDG-negative lesions that are occasionally unde- [5] Mosselman S, Polman J, Dijkema R. ER beta:
tected by PET imaging. identification and characterization of a novel hu-
Thus there is a wide spectrum of 18F-FES uptake man estrogen receptor. FEBS Lett 1996;392(1):
across tumor sites, and heterogeneity can be ob- 49–53.
served within a single patient. The current data [6] Pearce ST, Jordan VC. The biological role of
estrogen receptors alpha and beta in cancer.
suggest that 18F-FES is sensitive to detect ER-posi-
Crit Rev Oncol Hematol 2004;50(1):3–22.
tive tumor sites, and complements the information [7] Speirs V, Carder PJ, Lane S, et al. Oestrogen
provided by 18F-FDG to assess or predict response receptor [beta]: what it means for patients with
to therapy. Various studies are conducted at the breast cancer. Lancet Oncol 2004;5(3):174–81.
atuhors’ center and at least three other institutions [8] Clarke RB, Anderson E, Howell A. Steroid re-
to assess whether 18F-FES imaging has clinical value ceptors in human breast cancer. Trends Endo-
to select patients who have recurrent breast cancer crinol Metab 2004;15(7):316–23.
and who can benefit from second-line hormone [9] Osborne CK, Schiff R. Estrogen-receptor biol-
therapy by aromatase inhibitors or pure antagonists ogy: continuing progress and therapeutic im-
such as fulvestrant. With sufficient follow-up infor- plications. J Clin Oncol 2005;23(8):1616–22.
[10] Balfe PJ, McCann AH, Welch HM, et al. Estrogen
mation, the precise role of ER imaging agents in
receptor beta and breast cancer. Eur J Surg
this patient population will likely become clear. Oncol 2004;30(10):1043–50.
[11] Keen JC, Davidson NE. The biology of breast
Summary carcinoma. Cancer 2003;97(3):825–33.
[12] Mote PA, Bartow S, Tran N, et al. Loss of
ER expression measured by competitive binding co-ordinate expression of progesterone recep-
assays or immunohistochemistry has a proven util- tors A and B is an early event in breast carci-
ity in identifying a subset of patients who have nogenesis. Breast Cancer Res Treat 2002;72(2):
slower growing tumors and who will benefit from 163–72.
hormonal therapy, either with tamoxifen or second- [13] Tait JF, Tait SA. The effect of plasma protein
binding on the metabolism of steroid hor-
line agents such as aromatase inhibitors or pure
mones. J Endocrinol 1991;131(3):339–57.
antagonists. Several promising ligands that bind [14] Siiteri PK, Murai JT, Hammond GL, et al. The
selectively to the estrogen receptors are available serum transport of steroid hormones. Recent
for use in clinical research for PET and SPECT Prog Horm Res 1982;38:457–510.
imaging. The search for a suitable PR ligand is [15] Strel’chyonok OA, Avvakumov GV. Specific
still ongoing, but PR imaging might provide a bet- steroid-binding glycoproteins of human blood
ter estimate of tumor response to hormonal ther- plasma: novel data on their structure and func-
apy than ER imaging. Although the number of tion. J Steroid Biochem 1990;35(5):519–34.
published clinical studies remains limited, the [16] Hryb DJ, Khan MS, Romas NA, et al. The
available data suggest that ER imaging, as an control of the interaction of sex hormone-
binding globulin with its receptor by steroid
adjunct to 18F-FDG PET imaging, might be useful
hormones. J Biol Chem 1990;265(11):6048–54.
to predict the response to second-line hormonal [17] Osborne CK. Steroid hormone receptors in
therapy in recurrent or metastatic breast cancer. breast cancer management. Breast Cancer Res
Treat 1998;51(3):227–38.
[18] Henderson IC, Patek AJ. The relationship
References
between prognostic and predictive factors in
[1] Fisher B, Osborne K, Margolese R, et al. the management of breast cancer. Breast Cancer
Neoplasms of the breast. In: Holland J, Blast Res Treat 1998;52(1–3):261–88.
R, Morton D, et al, editors. Cancer Medicine. [19] Yasui Y, Potter JD. The shape of age-incidence
4th edition, vol 1. Baltimore (MD): Williams & curves of female breast cancer by hormone-
Wilkins; 1997. p. 2349–429. receptor status. Cancer Causes Control 1999;
[2] Ray S, Rastogi R, Kumar A. Current status of 10(5):431–7.
66 Beauregard et al

[20] Anderson WF, Chatterjee N, Ershler WB, et al. [33] Bonneterre J, Buzdar A, Nabholtz JM, et al. Anas-
Estrogen receptor breast cancer phenotypes in trozole is superior to tamoxifen as first-line
the Surveillance, Epidemiology, and End Results therapy in hormone receptor positive advanced
database. Breast Cancer Res Treat 2002;76(1): breast carcinoma. Cancer 2001;92(9):2247–58.
27–36. [34] Nabholtz JM, Bonneterre J, Buzdar A, et al.
[21] Early Breast Cancer Trialists’ Collaborative Anastrozole (Arimidex) versus tamoxifen as
Group. Tamoxifen for early breast cancer: an first-line therapy for advanced breast cancer
overview of the randomised trials. Lancet 1998; in postmenopausal women: survival analysis
351(9114):1451–67. and updated safety results. Eur J Cancer 2003;
[22] Overgaard M. Radiotherapy as part of a multi- 39(12):1684–9.
disciplinary treatment strategy in early breast [35] Geisler J, Haynes B, Anker G, et al. Influence
cancer. Eur J Cancer 2001;37(Suppl 7):S33–43. of letrozole and anastrozole on total body aro-
[23] Olson JE, Neuberg D, Pandya KJ, et al. The role matization and plasma estrogen levels in post-
of radiotherapy in the management of operable menopausal breast cancer patients evaluated in
locally advanced breast carcinoma: results of a a randomized, cross-over study. J Clin Oncol
randomized trial by the Eastern Cooperative 2002;20(3):751–7.
Oncology Group. Cancer 1997;79(6):1138–49. [36] Coombes RC, Hall E, Gibson LJ, et al. A ran-
[24] Harvey JM, Clark GM, Osborne CK, et al. domized trial of exemestane after two to three
Estrogen receptor status by immunohistochem- years of tamoxifen therapy in postmenopausal
istry is superior to the ligand-binding assay for women with primary breast cancer. N Engl J
predicting response to adjuvant endocrine Med 2004;350(11):1081–92.
therapy in breast cancer. J Clin Oncol 1999; [37] Baum M, Buzdar A, Cuzick J, et al. Anastrozole
17(5):1474–81. alone or in combination with tamoxifen ver-
[25] Bardou VJ, Arpino G, Elledge RM, et al. Pro- sus tamoxifen alone for adjuvant treatment of
gesterone receptor status significantly improves postmenopausal women with early-stage breast
outcome prediction over estrogen receptor sta- cancer: results of the ATAC (Arimidex, Tamoxi-
tus alone for adjuvant endocrine therapy in fen Alone or in Combination) trial efficacy and
two large breast cancer databases. J Clin Oncol safety update analyses. Cancer 2003;98(9):
2003;21(10):1973–9. 1802–10.
[26] Early Breast Cancer Trialists’ Collaborative [38] Goss PE, Ingle JN, Martino S, et al. A random-
Group. Systemic treatment of early breast ized trial of letrozole in postmenopausal
cancer by hormonal, cytotoxic, or immune women after five years of tamoxifen therapy
therapy. 133 randomised trials involving for early-stage breast cancer. N Engl J Med 2003;
31,000 recurrences and 24,000 deaths among 349(19):1793–802.
75,000 women. Lancet 1992;339(8785):71–85. [39] Howell A, Cuzick J, Baum M, et al. Results of the
[27] Goldhirsch A, Glick JH, Gelber RD, et al. Inter- ATAC (Arimidex, Tamoxifen, Alone or in Com-
national consensus panel on the treatment of bination) trial after completion of 5 years’
primary breast cancer. V: Update 1998. Recent adjuvant treatment for breast cancer. Obstet
Results Cancer Res 1998;152:481–97. Gynecol Surv 2005;60(5):307–9.
[28] Abrams JS. Adjuvant therapy for breast cancer— [40] Kahlert S, von Koch F, Ditsch N, et al. [Adjuvant
results from the USA consensus conference. treatment of breast cancer in 2004. A review of
Breast Cancer 2001;8(4):298–304. the 8th consensus conference of St. Gallen and
[29] Ferno M, Stal O, Baldetorp B, et al. Results of new study results]. Gynakol Geburtshilfliche
two or five years of adjuvant tamoxifen corre- Rundsch 2004;44(2):102–12.
lated to steroid receptor and S-phase levels. [41] Simpson D, Curran MP, Perry CM. Letrozole:
South Sweden Breast Cancer Group, and South- a review of its use in postmenopausal women
East Sweden Breast Cancer Group. Breast Can- with breast cancer. Drugs 2004;64(11):1213–30.
cer Res Treat 2000;59(1):69–76. [42] Bast Jr RC, Ravdin P, Hayes DF, et al. 2000
[30] National Institutes of Health Consensus Devel- update of recommendations for the use of tu-
opment Conference statement: adjuvant ther- mor markers in breast and colorectal cancer:
apy for breast cancer, November 1–3, 2000. clinical practice guidelines of the American So-
J Natl Cancer Inst Monogr 2001;(30):5–15. ciety of Clinical Oncology. J Clin Oncol 2001;
[31] Fisher B, Dignam J, Bryant J, et al. Five versus 19(6):1865–78.
more than five years of tamoxifen therapy for [43] Parker MG. Action of ‘‘pure’’ antiestrogens in
breast cancer patients with negative lymph inhibiting estrogen receptor action. Breast Can-
nodes and estrogen receptor-positive tumors. cer Res Treat 1993;26(2):131–7.
J Natl Cancer Inst 1996;88(21):1529–42. [44] Webb P, Nguyen P, Kushner PJ. Differential
[32] Colleoni M, Li S, Gelber RD, et al. Timing of SERM effects on corepressor binding dictate
CMF chemotherapy in combination with ta- ERalpha activity in vivo. J Biol Chem 2003;
moxifen in postmenopausal women with breast 278(9):6912–20.
cancer: role of endocrine responsiveness of the [45] Pink JJ, Jordan VC. Models of estrogen recep-
tumor. Ann Oncol 2005;16(5):716–25. tor regulation by estrogens and antiestrogens in
Steroid Receptor Imaging in Breast Cancer 67

breast cancer cell lines. Cancer Res 1996;56(10): trogen receptor assays in human breast cancer.
2321–30. Cancer Res 1983;43:413–6.
[46] Howell A, Osborne CK, Morris C, et al. ICI [62] Castagnetta L, Traina A, DiCarlo A, et al.
182,780 (Faslodex): Development of a novel, Heterogeneity of soluble and nuclear oestrogen
pure antiestrogen. Cancer 2000;89(4):817–25. receptor status of involved nodes in relation to
[47] Wakeling AE, Bowler J. Ici 182,780, a new anti- primary breast cancer. Eur J Cancer Clin Oncol
oestrogen with clinical potential. J Steroid Bio- 1987;23:31–5.
chem Mol Biol 1992;43(1–3):173–7. [63] Johnston S, Saccani-Jotti G, Smith E, et al. Changes
[48] Mauriac L, Pippen JE, Quaresma Albano J, et al. in estrogen receptor, progesteron receptor, and
Fulvestrant (Faslodex) versus anastrozole for the pS2 expression in tamoxifen-resistant human
second-line treatment of advanced breast cancer breast cancer. Cancer Res 1995;55:3331–8.
in subgroups of postmenopausal women with [64] Encarnacion C, Ciocca D, McGuire W, et al.
visceral and non-visceral metastases: combined Measurement of steroid hormone receptors in
results from two multicentre trials. Eur J Cancer breast cancer patients on tamoxifen. Breast
2003;39(9):1228–33. Cancer Res Treat 1993;26:237–46.
[49] Menard S, Fortis S, Castiglioni F, et al. HER2 as [65] Johnston S, Haynes B, Smith I, et al. Acquired
a prognostic factor in breast cancer. Oncology tamoxifen resistance in human breast cancer
2001;61(Suppl 2):67–72. and reduced intra-tumoral drug concentration.
[50] Duffy MJ. Predictive markers in breast and Lancet 1993;342:1521–2.
other cancers: a review. Clin Chem 2005; [66] Hochberg RB. Iodine-125–labeled estradiol: a
51(3):494–503. gamma-emitting analog of estradiol that binds
[51] Borg A, Baldetorp B, Ferno M, et al. ERBB2 am- to the estrogen receptor. Science 1979;205(4411):
plification is associated with tamoxifen resis- 1138–40.
tance in steroid-receptor positive breast cancer. [67] Pavlik EJ, Nelson K, Gallion HH, et al. Charac-
Cancer Lett 1994;81(2):137–44. terization of high specific activity [16 alpha-
[52] Arpino G, Green SJ, Allred DC, et al. HER-2 123I]Iodo-17 beta-estradiol as an estrogen
amplification, HER-1 expression, and tamoxifen receptor-specific radioligand capable of imaging
response in estrogen receptor-positive meta- estrogen receptor-positive tumors. Cancer Res
static breast cancer: a southwest oncology group 1990;50(24):7799–805.
study. Clin Cancer Res 2004;10(17):5670–6. [68] Schober O, Scheidhauer K, Jackisch C, et al.
[53] Agrup M, Stal O, Olsen K, et al. C-erbB-2 over- Breast cancer imaging with radioiodinated
expression and survival in early onset breast oestradiol. Lancet 1990;335(8704):1522.
cancer. Breast Cancer Res Treat 2000;63(1):23–9. [69] Kenady DE, Pavlik EJ, Nelson K, et al. Images
[54] Olayioye MA, Neve RM, Lane HA, et al. The of estrogen-receptor-positive breast tumors
ErbB signaling network: receptor heterodimer- produced by estradiol labeled with iodine I
ization in development and cancer. EMBO J 123 at 16 alpha. Arch Surg 1993;128(12):
2000;19(13):3159–67. 1373–81.
[55] Piccart MJ, Di Leo A, Hamilton A. HER2. a ’pre- [70] Scheidhauer K, Muller S, Smolarz K, et al. Tumor-
dictive factor’ ready to use in the daily manage- Szintigraphie mit 123J-markiertem Ostradiol
ment of breast cancer patients? Eur J Cancer beim Mammakarzinom–Rezeptorszintigraphie
2000;36(14):1755–61. [Tumor scintigraphy using 123I-labeled estradiol
[56] Yamauchi H, Stearns V, Hayes DF. When is a in breast cancer-receptor scintigraphy]. Nucl Med
tumor marker ready for prime time? A case (Stuttg) 1991;30(3):84–99 [in German].
study of c-erbB-2 as a predictive factor in breast [71] Symes EK, Coulson WF, Ralphs DN. The
cancer. J Clin Oncol 2001;19(8):2334–56. synthesis of 16 alpha-[131I]iodo-oestradiol
[57] Winston JS, Ramanaryanan J, Levine E. HER-2/ and evaluation of its use as a radiotracer for
neu evaluation in breast cancer are we there yet? oestrogen receptor positive breast tumours.
Am J Clin Pathol 2004;121(Suppl):S33–49. J Steroid Biochem 1985;22(2):155–60.
[58] Piccart MJ, Awada A. State-of-the-art chemother- [72] Gatley SJ, Shaughnessy WJ, Inhorn L, et al.
apy for advanced breast cancer. Semin Oncol Studies with 17 beta(16 alpha-[125i]iodo)-
2000;27(5, Suppl 9):3–12. estradiol, an estrogen receptor-binding radio-
[59] Ellis MJ, Coop A, Singh B, et al. Letrozole is more pharmaceutical, in rats bearing mammary
effective neoadjuvant endocrine therapy than tumors. J Nucl Med 1981;22(5):459–64.
tamoxifen for ErbB-1- and/or ErbB-2-positive, [73] Ali H, Rousseau J, Ghaffari MA, et al. Synthesis,
estrogen receptor-positive primary breast cancer: receptor binding, and tissue distribution of
evidence from a phase III randomized trial. (17 alpha,20E)- and (17 alpha,20Z)-21-[125I]
J Clin Oncol 2001;19(18):3808–16. iodo-19-norpregna-1,3,5(10), 20-tetraene-3,
[60] Ellis MJ, Coop A, Singh B, et al. Letrozole in- 17-diol. J Med Chem 1988;31(10):1946–50.
hibits tumor proliferation more effectively than [74] Ali H, Rousseau J, Ghaffari MA, et al. Synthesis,
tamoxifen independent of HER1/2 expression receptor binding, and tissue distribution of 7
status. Cancer Res 2003;63(19):6523–31. alpha- and 11 beta-substituted (17 alpha,20E)-
[61] Hull D, Clark G, Osborne C, et al. Multiple es- and (17 alpha,20Z)-21-[125I]iodo-19-norpreg-
68 Beauregard et al

na-1,3,5(10),20-tetraene-3,17-dio ls. J Med rivatives: synthesis and effect on estrogen recep-


Chem 1991;34(2):854–60. tor binding and receptor-mediated target tissue
[75] Foulon C, Guilloteau D, Baulieu JL, et al. Es- uptake. J Med Chem 1993;36(26):4255–63.
trogen receptor imaging with 17 alpha-[123I] [87] Ali H, Rousseau J, van Lier JE. Synthesis, recep-
iodovinyl-11 beta-methoxyestradiol (MIVE2)– tor binding and biodistribution of the gem-
Part I. Radiotracer preparation and charac- 21-chloro-21-iodovinylestradiol derivatives. J
terization. Int J Rad Appl Instrum B 1992; Steroid Biochem Mol Biol 1993;46(5):613–22.
19(3):257–61. [88] Ali H, Rousseau J, Paquette B, et al. Synthe-
[76] Ribeiro-Barras MJ, Foulon C, Baulieu JL, et al. sis and biological properties of 7alpha-cyano
Estrogen receptor imaging with 17 alpha-[123I] derivatives of the (17alpha,20E/Z)-[125I]iodo-
iodovinyl-11 beta-methoxyestradiol (MIVE2)— vinyl- and 16alpha-[125I]iodo-estradiols. Steroids
Part II. Preliminary results in patients with breast 2003;68(14):1189–200.
carcinoma. Int J Rad Appl Instrum B 1992; [89] Rijks LJ, van den Bos JC, van Doremalen PA, et
19(3):263–7. al. Synthesis, estrogen receptor binding, and
[77] Rijks LJ, Boer GJ, Endert E, et al. The stereo- tissue distribution of a new iodovinylestradiol
isomers of 17alpha-[123I]iodovinyloestradiol derivative (17alpha,20E)-21-[123I]Iodo-
and its 11beta-methoxy derivative evaluated 11beta-nitrato-19-norp regna-1,3,5 (10),20-tet-
for their oestrogen receptor binding in human raene-3,17-diol (E-[123I]NIVE). Nucl Med Biol
MCF-7 cells and rat uterus, and their distribu- 1998;25(4):411–21.
tion in immature rats. Eur J Nucl Med 1996; [90] Cummins CH. Radiolabeled steroidal estrogens
23(3):295–307. in cancer research. Steroids 1993;58(6):245–59.
[78] Rijks LJ, Boer GJ, Endert E, et al. The Z-isomer [91] McElvany KD, Katzenellenbogen JA, Shafer KE,
of 11 beta-methoxy-17 alpha-[123I]iodovinyl- et al. 16 alpha-[77Br]bromoestradiol: dosimetry
estradiol is a promising radioligand for estrogen and preliminary clinical studies. J Nucl Med
receptor imaging in human breast cancer. Nucl 1982;23(5):425–30.
Med Biol 1997;24(1):65–75. [92] McElvany KD, Carlson KE, Welch MJ, et al. In
[79] Rijks LJ, Bakker PJ, van Tienhoven G, et al. vivo comparison of 16 alpha[77Br]bromoestra-
Imaging of estrogen receptors in primary and diol-17 beta and 16 alpha-[125I]iodoestradiol-
metastatic breast cancer patients with iodine- 17 beta. J Nucl Med 1982;23(5):420–4.
123-labeled Z-MIVE. J Clin Oncol 1997;15(7): [93] Strickland LA, Ponce YZ, Hunter DH, et al.
2536–45. Amino and iodotamoxifens: synthesis, estrogen
[80] Nachar O, Rousseau JA, Lefebvre B, et al. Bio- receptor affinity and biodistribution. Drug Des
distribution, dosimetry and metabolism of Deliv 1990;6(3):195–212.
11beta-methoxy-(17alpha,20E/Z)-[123I]iodo- [94] Maaroufi Y, Quivy J, Trivedi S, et al.
vinylestradiol in healthy women and breast can- 4-iodotamoxifen aziridine, a new affinity label-
cer patients. J Nucl Med 1999;40(10):1728–36. ing agent for the rapid detection of estrogen
[81] Nachar O, Rousseau JA, Ouellet R, et al. receptor isoforms. J Steroid Biochem Mol Biol
Scintimammography with 11beta-methoxy- 1998;67(2):95–104.
(17alpha,20Z)-[123I]iodovinylestradiol: a com- [95] Hanson RN, Seitz DE. Tissue distribution of the
plementary role to 99mTc-methoxyisobutyl radiolabeled antiestrogen [125I]iodotamoxifen.
isonitrile in the characterization of breast tu- Int J Nucl Med Biol 1982;9(2):105–7.
mors. J Nucl Med 2000;41(8):1324–31. [96] Yang DJ, Li C, Kuang LR, et al. Imaging,
[82] Bennink ML, Leuba SH, Leno GH, et al. biodistribution and therapy potential of haloge-
Unfolding individual nucleosomes by stretch- nated tamoxifen analogues. Life Sci 1994;55(1):
ing single chromatin fibers with optical twee- 53–67.
zers. Nat Struct Biol 2001;8(7):606–10. [97] Van de Wiele C, De Vos F, De Sutter J, et al.
[83] Bennink RJ, van Tienhoven G, Rijks LJ, et al. In Biodistribution and dosimetry of (iodine-123)-
vivo prediction of response to antiestrogen treat- iodomethyl-N, N-diethyltamoxifen, an (anti)oestro-
ment in estrogen receptor-positive breast cancer. gen receptor radioligand. Eur J Nucl Med 1999;
J Nucl Med 2004;45(1):1–7. 26(10):1259–64.
[84] Ali H, Rousseau J, van Lier JE. Synthesis of [98] Van de Wiele C, Cocquyt V, VandenBroecke R,
A-ring fluorinated derivatives of (17 alpha,20E/Z)- et al. Iodine-labeled tamoxifen uptake in pri-
[125I]iodovinylestradiols: effect on receptor mary human breast carcinoma. J Nucl Med 2001;
binding and receptor-mediated target tissue up- 42(12):1818–20.
take. J Med Chem 1993;36(21):3061–72. [99] Skaddan MB, Wust FR, Jonson S, et al. Radio-
[85] Ali H, Rousseau J, van Lier JE. 7 alpha-Methyl- chemical synthesis and tissue distribution of
and 11 beta-ethoxy-substitution of [125I]-16 Tc-99m-labeled 7alpha-substituted estradiol
alpha-iodoestradiol: effect on estrogen receptor- complexes. Nucl Med Biol 2000;27(3):269–78.
mediated target tissue uptake. J Med Chem [100] Unak P, Enginar H, Biber FZ, et al. A correlative
1993;36(2):264–71. study between 99mTc-ESTCPTA and 99mTc-MIBI
[86] Ali H, Rousseau J, Gantchev TG, et al. 2- and in rats. Appl Radiat Isot 2002;57(5):733–42.
4-fluorinated 16 alpha(-)[125I]iodoestradiol de- [101] Hunter DH, Luyt LG. Single isomer technetium-
Steroid Receptor Imaging in Breast Cancer 69

99m tamoxifen conjugates. Bioconjug Chem agents for estrogen-receptor-positive breast tu-
2000;11(2):175–81. mors. J Med Chem 1993;36(11):1619–29.
[102] Ouellet R, Rousseau J, Brasseur N, et al. Syn- [115] VanBrocklin HF, Rocque PA, Lee HV, et al. 16
thesis, receptor binding, and target-tissue up- beta-[18F]fluoromoxestrol: a potent, metaboli-
take of carbon-11 labeled carbamate derivatives cally stable positron emission tomography imag-
of estradiol and hexestrol. J Med Chem 1984; ing agent for estrogen receptor positive human
27(4):509–13. breast tumors. Life Sci 1993;53(10):811–9.
[103] Dence CS, Napolitano E, Katzenellenbogen JA, [116] Jonson SD, Bonasera TA, Dehdashti F, et al.
et al. Carbon-11-labeled estrogens as potential Comparative breast tumor imaging and com-
imaging agents for breast tumors. Nucl Med parative in vitro metabolism of 16alpha-[18F]
Biol 1996;23(4):491–6. fluoroestradiol-17beta and 16beta-[18F]fluoro-
[104] Ali H, Rousseau J, Diksic M, et al. Synthesis, moxestrol in isolated hepatocytes. Nucl Med
receptor binding and target-tissue uptake of Biol 1999;26(1):123–30.
carbon-11-labeled carbamate derivatives of es- [117] Seimbille Y, Bénard F, van Lier JE. Synthesis of
tradiol and hexestrol. Int J Rad Appl Instrum B 16alpha-fluoro ICI 182,780 derivatives: power-
1992;19(2):175–82. ful antiestrogens to image estrogen receptor
[105] Ram S, Spicer LD. Direct Incorporation of densities in breast cancer by positron emission
[C-11] Carbon Dioxide for Labeling Bioactive tomography. J Chem Soc [Perkin 1] 2002;20:
Molecules - an Application to [C-11] Labeled 2275–81.
Tamoxifen. J Labelled Comp Radiopharm 1989; [118] Seimbille Y, Rousseau J, Benard F, et al. 18F-
27(6):661–8. labeled difluoroestradiols: preparation and pre-
[106] Kiesewetter D, Kilbourn M, Landvatter S, et al. clinical evaluation as estrogen receptor-binding
Preparation of four fluorine-18-labeled estro- radiopharmaceuticals. Steroids 2002;67(9):
gens and their selective uptake in target tissues 765–75.
of immature rats. J Nucl Med 1984;25(11): [119] Bénard F, Aliaga A, Ahmed N, et al. Biodis-
1212–21. tribution of fluorinated estradiol derivatives
[107] Mathias CJ, Welch MJ, Katzenellenbogen JA, in ER + tumor-bearing mice: impact of sub-
et al. Characterization of the uptake of 16 alpha- stituents, formulation and specific activity
([18F]fluoro)-17 beta-estradiol in DMBA- [abstract]. J Nucl Med 2004;45(5):329P.
induced mammary tumors. Int J Rad Appl [120] Inoue T, Kim EE, Wallace S, et al. Posi-
Instrum B 1987;14(1):15–25. tron emission tomography using [18F]fluoro-
[108] Mintun MA, Welch MJ, Siegel BA, et al. Breast tamoxifen to evaluate therapeutic responses
cancer: PET imaging of estrogen receptors. Ra- in patients with breast cancer: preliminary
diology 1988;169(1):45–8. study. Cancer Biother Radiopharm 1996;11(4):
[109] Mankoff DA, Peterson LM, Tewson TJ, et al. 235–45.
[18F]fluoroestradiol radiation dosimetry in [121] Landvatter SW, Kiesewetter DO, Kilbourn MR,
human PET studies. J Nucl Med 2001;42(4): et al. (2R*, 3S*)-1-[18F]fluoro-2,3-bis(4-hydro-
679–84. xyphenyl)pentane [(18F]fluoronor-hexestrol), a
[110] Mankoff DA, Tewson TJ, Eary JF. Analysis of positron-emitting estrogen that shows highly-
blood clearance and labeled metabolites for the selective, receptor-mediated uptake by target
estrogen receptor tracer [F-18]-16 alpha-fluo- tissues in vivo. Life Sci 1983;33(19):1933–8.
roestradiol (FES). Nucl Med Biol 1997;24(4): [122] Hostetler ED, Jonson SD, Welch PJ, et al.
341–8. Synthesis of 2-[F-18]fluoroestradiol, a potential
[111] Lim JL, Zheng L, Berridge MS, et al. The use of diagnostic imaging agent for breast cancer:
3-methoxymethyl-16 beta, 17 beta-epiestriol-O- Strategies to achieve nucleophilic substitution
cyclic sulfone as the precursor in the synthesis of an electron-rich aromatic ring with [F-18]F-.
of F-18 16 alpha-fluoroestradiol. Nucl Med Biol J Org Chem 1999;64(1):178–85.
1996;23(7):911–5. [123] Seimbille Y, Benard F, Rousseau J, et al. Impact
[112] Romer J, Fuchtner F, Steinbach J, et al. Auto- on estrogen receptor binding and target tissue
mated production of 16alpha-[18F]fluoroestra- uptake of [18F]fluorine substitution at the 16
diol for breast cancer imaging. Nucl Med Biol [alpha]-position of fulvestrant (faslodex; ICI
1999;26(4):473–9. 182,780). Nucl Med Biol 2004;31(6):691–8.
[113] Pomper MG, VanBrocklin H, Thieme AM, et al. [124] Pomper MG, Katzenellenbogen JA, Welch MJ,
11 beta-methoxy-, 11 beta-ethyl- and 17 alpha- et al. 21-[18F]fluoro-16 alpha-ethyl-19-norpro-
ethynyl-substituted 16 alpha-fluoroestradiols: gesterone: synthesis and target tissue selective
receptor-based imaging agents with enhanced uptake of a progestin receptor based radio-
uptake efficiency and selectivity. J Med Chem tracer for positron emission tomography.
1990;33(12):3143–55. J Med Chem 1988;31(7):1360–3.
[114] VanBrocklin HF, Carlson KE, Katzenellenbogen [125] Dehdashti F, McGuire AH, Van Brocklin HF,
JA, et al. 16 beta-([18F]fluoro)estrogens: sys- et al. Assessment of 21-[18F]fluoro-16 alpha-
tematic investigation of a new series of fluorine- ethyl-19-norprogesterone as a positron-emitting
18-labeled estrogens as potential imaging radiopharmaceutical for the detection of pro-
70 Beauregard et al

gestin receptors in human breast carcinomas. [18F] fluoro-17 beta-estradiol uptake in meta-
J Nucl Med 1991;32(8):1532–7. static breast carcinoma. J Nucl Med 1991;32(8):
[126] Jonson SD, Welch MJ. PET imaging of breast 1526–31.
cancer with fluorine-18 radiolabeled estro- [131] Dehdashti F, Mortimer JE, Siegel BA, et al. Posi-
gens and progestins. Q J Nucl Med 1998;42(1): tron tomographic assessment of estrogen recep-
8–17. tors in breast cancer: comparison with FDG-PET
[127] Ali H, Rousseau J, Ahmed N, et al. Synthesis and in vitro receptor assays. J Nucl Med 1995;
of the 7alpha-cyano-(17alpha,20E/Z)-[125I] 36(10):1766–74.
iodovinyl-19-nortestosterones: potential radio- [132] Mortimer JE, Dehdashti F, Siegel BA, et al.
ligands for androgen and progesterone recep- Metabolic flare: indicator of hormone respon-
tors. Steroids 2003;68(14):1163–71. siveness in advanced breast cancer. J Clin Oncol
[128] Rijks LJ, van den Bos JC, van Doremalen PA, 2001;19(11):2797–803.
et al. New iodinated progestins as potential [133] Mankoff DA, Peterson LM, Petra PH, et al.
ligands for progesterone receptor imaging in Factors affecting the level and heterogeneity
breast cancer. Part 2: In vivo pharmacological of uptake of [18F]Fluoroestradiol in patients
characterization. Nucl Med Biol 1998;25(8): with estrogen receptor positive breast cancer
791–8. [abstract]. J Nucl Med 2002;43(5):286P.
[129] Katzenellenbogen JA. Designing steroid receptor- [134] Linden HM, Stekhova S, Link JM, et al. HER2
based radiotracers to image breast and prostate expression and uptake of 18F-Fluoroestradiol
tumors. J Nucl Med 1995;36(Suppl 6):8S–13S. (FES) predict response of breast cancer to hor-
[130] McGuire AH, Dehdashti F, Siegel BA, et al. monal therapy [abstract]. J Nucl Med 2004;
Positron tomographic assessment of 16 alpha- 45(5):85P.
71

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 71–81

Changes in Glucose Metabolism


and Blood Flow Following
Chemotherapy for Breast Cancer
David A. Mankoff, MD, PhD*, Lisa K. Dunnwald, BS, CNMT

& Positron emission tomography & Other positron emission tomography


methodology radiopharmaceuticals
18
& F-fluordeoxyglucose positron emission & Summary
tomography and neoadjuvant & References
chemotherapy
& 18F-fluordeoxyglucose positron emission
tomography and systemic therapy of
metastatic disease

One of the important advances in breast cancer 40,000 women per year dying of breast cancer in
treatment over the past 2 decades is the recognition the United States, and their mortality is largely
that breast cancer is a systemic disease necessitating related to progressive metastatic disease [4]. The
systemic therapy, even when apparently localized ability to monitor response in patients who have
[1]. Although a variety of options for breast cancer macroscopic breast cancer treated by systemic ther-
systemic therapy have emerged, cytotoxic chemo- apy is therefore an important task for which posi-
therapy remains an essential and commonly used tron emission tomography (PET) is playing an
form of treatment. Most breast cancer chemo- emerging role. This article focuses on this applica-
therapy occurs in the adjuvant setting, following tion of PET to breast cancer.
definitive surgery, designed to eradicate systemic Many solid tumors respond poorly to systemic
micrometastases [2]. Imaging currently has a small therapy; however, breast cancer is one of the more
role in these patients, and is reserved for sur- chemotherapy-sensitive solid tumors [5]. Women
veillance or work-up of symptoms suggesting pos- who have locally advanced or metastatic breast can-
sible recurrence; however, a substantial fraction cer can have prolonged remissions [6–8]. Those
of women present with more advanced cancer, at that have failed first-line chemotherapy still have
Stage III or Stage IV, when resection may not be a number of reasonable choices for second-line
feasible nor appropriate. Furthermore, there is an therapy with substantial response rates [5,9]; how-
increasing trend toward presurgical or neoadjuvant ever, the ability to evaluate chemotherapy response,
chemotherapy, even for resectable disease [3]. For and thereby to make appropriate adjustments in
all these reasons, the systemic treatment of women systemic therapy, is limited. For primary breast tu-
who have one or more macroscopic tumor sites is mors, anatomic imaging and sized-based response
an important public health problem. There are evaluation can substantially over- or underestimate

This work was supported by NIH Grants R01CA72064, P01CA42045, R01CA90771, and S10RR17229; by the
Avon Cancer Crusader Fund; and by the British Columbia Cancer Fund.
Division of Nuclear Medicine, Department of Radiology, Box 356113, Room NN203, University of Washington,
1959 NE Pacific Street, Seattle, WA 98195, USA
* Corresponding author.
E-mail address: dam@u.washington.edu (D.A. Mankoff).

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.001
pet.theclinics.com
72 Mankoff & Dunnwald

response [6,10,11]. For certain sites of metastasis, period (μCi/g or μCi/mL), ID is the injected dose
for example bone metastasis, changes in imaging (mCi) and wt is the patient weight (kg). This mea-
for conventional methods such as CT and bone sure is simple to implement and clinically feasi-
scintigraphy can significantly lag response or pro- ble, but it is an approximation of more complex
gression [12]. Furthermore, it takes several weeks FDG kinetics, and therefore has a number of limi-
to months to evaluate efficacy using changes in tations, which are well-described in the litera-
tumor size to assess response. For some therapies ture [20]. Many of these problems arise from the
that are potentially cytostatic, such as hormonal fact that simple uptake measures include the FDG
therapy, it can be impossible to discern tumor that is present as unphosphorylated FDG in addi-
response from slow disease progression when rely- tion to phosphorylated FDG in the trapped com-
ing on anatomically based measures of response. partment [17,19]. Only the phosphorylated FDG
These are tasks to which a quantitative, functional carries specific information on glucose metabo-
imaging method such as PET is ideally suited. Better lism. For tissues with high metabolic rate of FDG
methods of predicting and evaluating response are (MRFDG), most of the label in the tumor region-
critical to the individualized therapy of patients of-interest late after injection will be in the form of
who have breast cancer, especially as advances in FDG-6-phosphate, and therefore SUV provides a
basic biology give rise to a wider variety of thera- reasonable measure for quantifying FDG uptake.
peutic options [2,9]. For less metabolically active tumors, a greater frac-
This article reviews the PET methodology used for tion of the label is nonphosphorylated FDG, and
breast cancer response assessment, with an empha- SUV measures FDG metabolism less well. This
sis on quantitative methods. This is followed by a has implications for tracking response to ther-
review of results to date for neoadjuvant chemo- apy in which tumors move from high glucose
therapy and therapy of metastatic breast cancer. metabolism pretreatment to lower glucose me-
Preliminary studies with tracers other than 18F- tabolism post-treatment, and may account for
fluordeoxyglucose (FDG) are reviewed, followed some of the differences in results between stud-
by a summary and discussion of future directions. ies using SUV versus those using more rigorous
methods. An important consideration is that SUV
changes quickly in metabolically active tumors
[21,22]; therefore maintaining a constant time be-
Positron emission tomography methodology tween injection and imaging is important in com-
Most of the studies using PET to monitor breast paring serial studies.
cancer chemotherapy response have used 18F- More rigorous quantitative analysis of dynamic
fluordeoxyglucose (FDG) [13]. Most applications FDG PET images is upon a model for FDG
have used fairly standard approaches, with patients kinetics [Fig. 1], based upon the original work
fasting at least 4 to 6 hours before imaging, and with of Sokoloff and colleagues [23], Reivich and col-
imaging times typically 45 to 60 minutes after injec- leagues [24], and Phelps and colleagues [25] for
tion, and attenuation-corrected imaging on whole- brain imaging. This model depicts the rate con-
body PET tomographs. Some studies [14,15] have stants for FDG as tracer moves from the blood to
also performed dynamic imaging over a particular tissue, where transport is largely reversible (K1 and
tumor site, typically the primary breast tumor, k2), and as it is phosphorylated to form FDG-
allowing for more rigorous quantitative analysis. 6-phosphate (k3) and trapped, with allowance
Because quantitative analysis of serial FDG PET for a small amount of dephosphorylation in
studies is key in application to response monitor- some tissues (k4). In this model, initial transport
ing, some aspects of quantitation are highlighted and retention in the reversible compartment are
here. More detailed discussion of quantitative nonspecific, in that FDG is not yet committed to
methods can be found in other studies [16–19]. the glucose metabolic pathway and may be influ-
The most commonly used measured of FDG uptake enced by factors that are unrelated to glucose
in published studies of breast cancer response is metabolism. These factors include blood flow, cap-
most frequently known as the standardized up- illary permeability, and the size of the local inter-
take value (SUV), defined by the following for- stitial fluid pool [17]. Glucose metabolism is more
mula [17]: specifically indicated by the FDG trapped as FDG-
6-phosphate, and the most relevant measure of
Ct glucose metabolism that can be obtained from
SUV ¼ FDG PET is the flux of FDG from the blood into
ID=wt
the trapped compartment—the phosphorylated
In this formula, Ct bar is the average tissue state. This measure is typically described in units
uptake during the course of the static imaging of μmoles/min/g of tissue and is related to the
PET & Breast Cancer Therapy 73

Blood Tissue Total Activity in PET Image

K1 k3
Exchangeable Trapped
Blood Tissue FDG k4 FDG-
k2
FDG 6P

Fig. 1. Compartmental model of FDG kinetics.

FDG compartmental model rate constants by the 18


F-fluordeoxyglucose positron emission
formula below:
tomography and neoadjuvant chemotherapy
K1 k 3 Presurgical or neoadjuvant therapy is increasingly
MRFDG ¼ ½Glucose ¼ ½GlucoseKi
k2 þ k3 used in patients presenting with nonmetastatic
breast cancer. The majority of studies performed
In this formula, [Glucose] is the plasma glucose evaluating PET to measure breast cancer response
concentration (μmole/mL). The combination of to systemic therapy have been done in this set-
rate constants in the fraction term, which describes ting. Neoadjuvant chemotherapy was originally
a rate constant for flux from the blood to the applied to locally advanced breast cancer (LABC),
phosphorylated state, is often termed the flux con- defined as primary tumors that are greater than
stant, Ki (mL/min/g). The equation differs from 5 cm, fixed axillary lymph nodes, or skin/chest
the classic work of Reivich and coworkers [24] wall invasion [32]. More recently, neoadjuvant che-
and Phelps and coworkers [25] for brain imaging, motherapy has been applied to smaller, resectable
in that there is no attempt to relate FDG kinetics breast cancers [3], often with the goal of shrinking
directly to glucose kinetics. Instead, the term the tumor and offering the possibility of breast-
MRFDG is used to indicate glucose flux estimated conserving surgery instead of mastectomy [33].
by FDG PET, and to reflect the fact that there are Neoadjuvant chemotherapy regimens are most
differences between FDG and glucose metabolism, commonly anthracylcine-based (doxorubicin or
especially in tumors [26]. Glucose metabolic rate or epirubicin), and in recent trends they are combined
MRFDG estimated from full compartmental mod- sequentially with a taxane [2,34]. Other chemo-
eling has been reported in several studies of neoad- therapy regimens have been successfully applied.
juvant chemotherapy of breast cancer [14,27,28]. Neoadjuvant hormonal therapy using tamoxifen or
An alternate and slightly simplified approach is aromatase inhibitors has been tested, but with
the graphic method devised by Patlak and Blasberg slower response rates and less complete response
[29] and Gjedde [30]. This method requires compared with chemotherapy [35].
dynamic tissue uptake curves, but transforms the Studies have shown that the extent of residual
problem of estimating the flux constant to linear breast and axillary tumor post-neoadjuvant che-
regression, or in other words, a line fit. In this motherapy is prognostic of disease-free survival
formulation, a plot of the ratio of tissue activity (DFS) and overall survival (OS) [6–8,36,37]. Pa-
to blood activity versus normalized time, which tients who have a pathologic complete response
takes the blood clearance curve into account, ap- (CR)—defined as disappearance of the primary tu-
proaches a straight line with slope equal to the flux mor on histopathologic analysis of the breast speci-
constant, Ki, under the condition that there is no men from definitive post-therapy surgery–have
loss from the trapped compartment (ie, k4 = 0). better disease-free survival than those patients
Graphic analysis is a simple and reasonably robust who have residual tumor [6,7,37]. This has there-
approach for quantifying FDG uptake kinetics. The fore become an important endpoint in evaluating
graphic approach does not yield all of the kinetic neoadjuvant therapy regimens. Earlier investiga-
insight that more rigorous quantitative analysis tions used a macroscopic complete response (mCR)
provides; however, it is simple and robust. Graphic as the endpoint, with mCR defined as the absence
analysis has been applied in some of the published of gross disease post-therapy, even if scattered mi-
literature on breast cancer neoadjuvant chemother- croscopic disease remained [6]. More recent studies
apy [15,31]. have used the microscopic resolution of all invasive
74 Mankoff & Dunnwald

Pre-Therapy Chemotherapy Surgery

8-12 wks 16 - 24 wks


Baseline

Early Therapy Mid-Therapy Post-Therapy

Fig. 2. Timeline for neoadjuvant treatment of breast cancer showing times of tumor assessment by FDG PET in
published studies.

cancer (pCR) as the preferred endpoint, following coworkers [41] found evidence of internal mam-
the lead of recent trials of neoadjuvant therapy of mary (IM) nodal involvement in approximately
resectable breast cancer [37]. The different defini- 20% of patients who had locally advanced breast
tion of complete response can cause confusion in cancer, and the presence of pretherapy FDG up-
comparing studies, and should be considered in take in IM lymph nodes predicted regional or sys-
evaluating studies of imaging to measure response temic failure.
to neoadjuvant therapy. The number of residual The majority of studies of FDG PET for neoadju-
lymph node metastases post-therapy is also prog- vant chemotherapy have measured FDG uptake
nostic. The studies of McCready and colleagues [8] midtherapy as a measure of response, and tested
showed that patients who had three or fewer lymph the ability of midtherapy measurements of FDG
nodes post-therapy were significantly more likely to uptake to predict post-therapy pathologic response
remain disease-free than those patients who had [14,15,42–44] [Fig. 4]. Results are summarized in
four or more positive lymph nodes post-therapy. Table 1. Almost all studies show a significant dif-
A number of investigators have tested the appli- ference in midtherapy FDG uptake for responders
cation of FDG PET to evaluate breast cancer re- versus nonresponders. Echoing the early studies of
sponse to neoadjuvant chemotherapy. In fact, Wahl and colleagues [14], all studies have shown
some of the earliest studies testing FDG PET to that in patients who go on to have a good response
measure chemotherapy response were performed to neoadjuvant chemotherapy, primary tumor FDG
in this clinical setting [14,38]. In these studies uptake declines by approximately 50% or more
of presurgical breast cancer treatment, FDG PET from pretherapy levels. Studies have shown that
was applied at two or more time points along nonresponding tumors also had a modest decline
the course of chemotherapy [Fig. 2]: pretherapy, in FDG uptake. This was as high as a 40% decline
early in the course of therapy (after a single cycle in nonresponders [15,43], but typically is less than
of chemotherapy), midtherapy, and post-therapy 50%. It therefore appears that a 50% or greater
(presurgery). These time points are discussed sepa- decline in FDG uptake midtherapy is a marker of
rately below. breast cancer response to neoadjuvant chemother-
The pretherapy scan serves as an important base- apy. So far, studies have not established the prog-
line in measuring changes in the extent and level nostic significance of this finding, although the
of FDG uptake post-therapy; however, it also pro- studies of Mankoff and coworkers [15] show a
vides information on the extent of disease that may
be important in determining postsurgical treat-
ment, especially regional radiotherapy. PET pro-
vides information on the presence or absence of
axillary lymph node metastasis. Because sentinel
lymph node biopsy can be misleading post-therapy
if there was macroscopic disease pretherapy [39],
this can be very helpful in directing the approach
to surgical staging or lymph node dissection. In
addition, PET accurately depicted the extent of
macroscopic axillary disease (reviewed in [40])
[Fig. 3]. This can be especially important in defin-
ing radiotherapy fields post-therapy when there has Fig. 3. Coronal FDG PET images from a patient with
been substantial resolution of macroscopic bulky multifocal left breast cancer (thin arrows) and exten-
disease by systemic treatment. Finally, FDG PET can sive axillary disease (thick arrow), occupying all three
identify spread to extra-axillary nodes. Bellon and axillary levels.
PET & Breast Cancer Therapy 75

Fig. 4. Axial FDG PET images through the thorax and breasts in a patient with inflammatory breast cancer treated
with neoadjuvant chemotherapy and imaged after 2 and 4 months of treatment. The pretherapy scan shows a
large tumor with uptake in the skin consistent with dermal invasion of the cancer. Abnormalities progressively
decline over the course of treatment. The patient was found to have no residual invasive disease, and only
noninvasive disease ductal carcinoma in situ (DCIS) at surgery post-therapy. The effect of marrow-stimulating
cytokines granulocyte colony stimulating factor (GCSF), given as part of the chemotherapy regimen, is seen on the
2- and 4-month scans as increased vertebral marrow uptake (dashed arrows).

surements [14,43,44] [Fig. 5]. These exciting results


trend for patients who had lower midtherapy FDG
suggest that FDG PET may be an early predictor of
uptake, measured as MRFDG, to have improved
chemotherapy response and merit further study.
disease-free survival. Demonstration that serial
Some studies have looked at post-therapy FDG
FDG PET predicts survival in the neoadjuvant set-
PET as a measure of the extent of residual disease
ting is an important area of future investigation that
[Table 3] [42,45,46]. This information can help
could establish FDG PET as a quantitative surrogate
direct post-therapy definitive surgery and possibly
in future drug therapy trials.
adjuvant radiotherapy. Studies have shown that
A smaller number of studies have looked at the
although residual FDG uptake post-therapy is a
change in FDG uptake early in the course of ther-
good indicator of residual macroscopic disease,
apy as a predictor of pathologic response. Wahl
the absence of FDG uptake post-therapy is not a
and colleagues [14], Smith and colleagues [43],
reliable indicator of a complete pathologic re-
and Schelling and colleagues [44] all measured
sponse. This is especially true for axillary nodal
FDG uptake after the first cycle of every-3-weeks
metastases, in which the sensitivity of post-therapy
anthracycline-based chemotherapy [Table 2]. All
FDG for residual nodal disease appears to be low
three of these studies showed that early FDG PET
[42,45]. This is likely due to several factors, includ-
predicted subsequent response to chemotherapy,
ing the observation that chemotherapy lowers
in some cases with greater separation between re-
FDG uptake even in patients who have a modest
sponders and nonresponders than midtherapy mea-
response, and that incompletely treated lymph
nodes often harbor small-volume disease for
Table 1: FDG PET results for midtherapy which PET sensitivity is limited even in the absence
evaluation of treatment [47]. These results limit the utility of
Reference N Therapy Results post-therapy FDG PET to determine the extent of
residual disease in locally advanced breast cancer. A
Wahl 11 AC R: −48% SUV possible role for FDG PET in this setting, however,
et al [14] NR: −19% SUV was highlighted in a preliminary study by Chen
Bassa 15 FAC All: −51% SUV
and coworkers [48], who used FDG PET in
et al [42]
Schelling 24 EC or mCR: −46% SUV conjunction with breast MRI. In this study, FDG
et al [44] ET not mCR: −8% SUV PET was complementary to the detailed anatomic
Smith 30 CVAP mCR: −86% SUV information provided by MRI on the extent of re-
et al [43] not mCR: −40% SUV sidual disease. FDG PET added specificity to the
Mankoff 35 FAC mCR: −65%
et al [15] or AC MRFDG
PR: −49% Table 2: FDG PET results for early therapy
MRFDG evaluation
NR: −40% Reference N Therapy Results
MRFDG
Wahl et al 11 AC R: −22% SUV
Abbreviations: AC, epirubicin/cyclophosphamide; CVAP, [14] NR: no change
epirubicin/cyclophosphamide/vincristine/prednisolone; Schelling 24 EC or mCR: −54% SUV
EC, epirubicin/cyclophosphamide; ET, epirubicin/pacli- et al [44] ET not mCR: −19% SUV
taxel; FAC, 5-fluorouracil/doxorubicin/cyclophosphamide; Smith 30 CVAP mCR: −77% SUV
mCR, macroscopic complete response; NR, no response;
et al [43] not mCR: +1% SUV
PR, partial response; R, response.
76 Mankoff & Dunnwald

A 140%
B 0.12
120% 0.1
% Change Mean

100%
0.08
80% mCR

DUR
GRD
SUV

0.06
60% MRD Other
0.04
40%

20% 0.02

0% 0
0 1 2 3 0 1 2 3 4 5
Cycle # Cycle #
Fig. 5. Time course of FDG uptake taken from [44] (A) and [43] (B). Both plots show an early decline in tumor
uptake after the first course of chemotherapy for minimal residual disease (MRD) versus gross residual disease
(GRD) (A) and for mCR versus other response (B). There is no further separation of the different response curves at
the midtherapy time point. In the Smith et al data [43] (B), there is less separation of mCR versus other at
midtherapy versus early therapy, due to a decline in uptake in patients with other than mCR at midtherapy not
seen early. DUR, differential uptake ratio (equivalent to the SUV).

MRI examinations and was accurate in identifying and therefore FDG PET may be particularly help-
resistant cancer. These encouraging results warrant ful in this clinical setting. Unfortunately, there
further multimodality studies in the setting of neo- have been relatively few studies of Stage IV breast
adjuvant chemotherapy. cancer response monitoring using FDG PET
[50,51], complicated by small patient numbers, a
variety of disease presentations, and a number
18F-fluordeoxyglucose positron emission of different types of therapy including chemo-
tomography and systemic therapy of therapy, hormonal therapy, and biologic therapy
(eg, trastuzumab).
metastatic disease
Gennari and colleagues [50] studied 13 patients
Unlike many other solid tumors, breast cancer is who had metastatic breast cancer undergoing che-
often responsive to systemic therapy even at ad- motherapy of Stage IV breast cancer using one of
vanced stages [49]. Although Stage IV (metastatic) two different epirubicin-based regimens. The study
breast cancer is rarely ‘‘cured,’’ patients who have authors found that after a single cycle of che-
Stage IV disease often have indolent disease, and motherapy (day 8), those patients who went on
there is a wide and expanding range of systemic to a clinical response had an average 25% decline
therapy options. These patients may therefore in FDG SUV, whereas nonresponders had no
have prolonged survival with good quality of life. change. By the completion of therapy, responders
Evaluation of metastatic breast cancer response had a greater than 80% average decline in FDG
by conventional imaging, looking for change in SUV, similar to the neoadjuvant setting, whereas
tumor size [12], can be particularly challenging, nonresponders had essentially no change. These
preliminary results suggest that, as in neoadjuvant
chemotherapy, objective response is accompanied
Table 3: FDG PET results for post-therapy by a large decline in FDG uptake (50%–75% or
Results (for more) after at least 2 or 3 months of treatment, and
Reference N Therapy residual disease) by a smaller decline early in the course of che-
Bassa 11 FAC Primary: Sens 75% motherapy. Further studies are needed to better
et al [42] Axilla: Sens 42%, define endpoints for a variety of treatment regi-
Spec 100% mens in the metastatic setting.
Burcombe 9 FEC Primary: Sens 0% (0/9) Bone metastases present a particularly troubling
et al [45] Axilla: Sens 0% (0/3) site of disease for evaluating response. Although
Kim et al 50 ACT Primary: bone scan and MRI are highly accurate in detecting
[46] or XT SUV: CR, −97%; PR, breast cancer bone metastasis, they are ineffective
−88%; NR, −56%; for measuring response [52,53]. Changes in bone
Sens, 86%; Spec, 83% metastasis appearance on bone scan and MRI can
(−79% threshold)
lag metastasis response, and lesions may appear to
Abbreviations: Sens, sensitivity; Spec, specificity; XT, ca- worsen early in the course of successful treatment
pecitabine/docetaxol. [54,55]. Given the possibility of prolonged sur-
PET & Breast Cancer Therapy 77

vival, the wide array of therapeutic choices, and the present, in the form of aromatase inhibitors, which
inadequacy of current conventional imaging for do not have an agonist effect and therefore likely
evaluating bone metastasis response, better tools do not have an associated flare. The authors have
for evaluating response are clearly needed. Taking observed substantial declines in FDG uptake as
advantage of the fact that breast cancer bone metas- little as 6 weeks after initiation of aromatase in-
tases appear to indicate the tumor itself, rather than hibitors for metastatic breast cancer in our pre-
the bony reaction to the tumor [56,57], Stafford liminary experience [Fig. 6].
and colleagues [51] tested the use of serial FDG PET
to measure breast cancer bone metastasis response.
They found that the change in FDG PET correlated Other positron emission tomography
with clinical response, based upon a combination radiopharmaceuticals
of conventional imaging, clinical symptoms, and Several studies have used PET radiopharmaceuticals
tumor markers. Furthermore, there was a quantita- other than FDG to examine other aspects of breast
tive correlation between the fractional change in cancer biology in patients undergoing systemic
FDG SUV and fractional change in tumor markers, therapy. Based on the work of Beaney and col-
two measurements often used to assess bone metas- leagues [60] and Wilson and colleagues [61], sev-
tasis response [53]. Preliminary follow-up of these eral studies have used 15O-water to measure blood
results suggests that the change in FDG uptake in flow in patients who had LABC treated by neo-
treated bone metastases is not only an indicator of adjuvant chemotherapy. Simultaneous studies of
response, but also appears to predict progression- blood flow and metabolism using 15O-water and
free survival [58], in that patients who have a larger FDG PET have yielded some interesting results.
decline in FDG SUV over the course of therapy have Zasadny and coworkers [28] found some correla-
a longer time to progression. These encouraging tion between blood flow and metabolism, but bet-
preliminary results merit further prospective study. ter correlation between blood flow and early FDG
Some caution must be taken in interpreting the delivery (FDG K1). Tseng and coworkers [27] found
results of serial FDG PET studies in evaluating the similar results; in addition, their data suggested that
response of metastatic breast cancer to systemic blood flow and early delivery of glucose were not
therapy other than chemotherapy. For example, the rate-limiting step in the LABC glucose metabolic
Mortimer and coworkers [59] found that FDG up- rate in most cases. Interestingly, Mankoff and co-
take increased early in the course of successful workers [31] found that a metabolism-blood flow
tamoxifen therapy. A ‘‘flare’’ of disease in the first mismatch, in the form of aberrantly high FDG
7 to 10 days after starting tamoxifen accurately metabolism relative to blood flow, was an indicator
predicted good response to subsequent treatment, of poorer response to neoadjuvant chemotherapy
likely a result of the early agonist effect of tamoxi- and poorer prognosis.
fen in hormone-sensitive breast cancer. First-line In serial 15O-water PET studies over the course of
hormonal therapy is more commonly given, at neoadjuvant chemotherapy of locally advanced
breast cancer, Mankoff and colleagues [15] found
that the change in tumor blood flow midtherapy
was an excellent predictor of response and of
patient outcome, better than FDG uptake in both
regards. Patients who responded to neoadjuvant
chemotherapy had an average decline in tumor
blood flow, whereas nonresponders had an aver-
age increase in blood flow. Furthermore, residual
blood flow midtherapy predicted both disease-free
and overall survival; lower midtherapy blood flow
was associated with better disease-free and over-
all survival. This echoes results with other imag-
ing modalities strongly influenced by tumor blood
flow, such as contrast-enhanced MRI [62–65],
Doppler ultrasound [66,67], and technetium-99m
Fig. 6. Response to aromatase inhibitors. Coronal FDG
(99mTc)-sestamibi [68–72]. A comparison of changes
PET images of a patient with recurrent breast cancer
presenting as isolated sternal disease (arrows). FDG in blood flow and FDG metabolism over the course
PET shows a substantial decline in uptake after only of therapy by Tseng and coworkers [27] suggested
6 weeks of treatment. The patient went on to have a that the pattern of glucose metabolism changed.
prolonged response to treatment with minimal resi- Blood flow and metabolism were better matched
dual disease. after chemotherapy, with a ratio closer to the nor-
78 Mankoff & Dunnwald

mal breast than the untreated tumor, even in non- which conventional imaging is inadequate for mea-
responders. This provides an interesting insight into suring response. Importantly, future use will likely
the biology of chemotherapy-treated tumors, and include use of PET radiopharmaceuticals beyond
may help explain the decline in FDG uptake seen FDG to help guide and monitor breast cancer sys-
after chemotherapy, even in nonresponders. temic therapy by helping to identify therapeutic
PET can be used to measure the pharmacologic targets, and by determining response early in the
effect of drugs directly. Dehdashti and colleagues course of therapy.
[73] and Mortimer and colleagues [59] performed
serial 18F-fluoroestradiol (FES) PET studies to docu-
ment tamoxifen blockade of the estrogen receptor References
(ER). These studies showed that high pretherapy
FES uptake, as an indicator of estrogen receptor [1] Fisher B, Wickerham DL, Redmond C. Recent
expression, and a decline in FES uptake, as an in- developments in the use of systemic adjuvant
dicator of estrogen receptor blockade, were pre- therapy for the treatment of breast cancer. Semin
Oncol 1992;19(3):263–77.
dictive of response to primary tamoxifen therapy.
[2] Thomas E, Hortobagyi GN. New paradigms in
The studies set forth an important paradigm for the adjuvant systemic therapy of breast cancer.
use of PET imaging in early drug trials, and this Endocr Relat Cancer 2003;10(1):75–89.
merits further study in a variety of clinical settings [3] Bonadonna G, Valagussa P, Zucali R, et al.
and for a variety of treatments. Primary chemotherapy in surgically resectable
Finally, some studies have investigated the use of breast cancer. CA Cancer J Clin 1995;45:227–43.
other PET radiopharmaceuticals to measure breast [4] Jemal A, Murray T, Samuels A, et al. Cancer Sta-
cancer response. Energy metabolism fuels tumor tistics, 2003. CA Cancer J Clin 2003;53(1):5–26.
growth, but it also fuels a variety of other biologic [5] Hortobagyi GN. Progress in systemic chemother-
processes, including cellular repair. Therefore, it is apy of primary breast cancer: an overview. J Natl
Cancer Inst Monogr 2001;30:72–9.
likely that radiopharmaceuticals measuring pro-
[6] Feldman LD, Hortobagyi GN, Buzdar AU, et al.
cesses more specific to cellular growth may be Pathological assessment of response to induction
advantageous for response evaluation. In early chemotherapy in breast cancer. Cancer Res
and pioneering studies, Huovinen and coworkers 1986;46:2578–81.
[74] and Jansson and coworkers [75] measured [7] Machiavelli MR, Romero AO, Perez JE, et al.
breast cancer response to chemotherapy using la- Prognostic significance of pathological response
beled amino acids as a measure of cellular bio- of primary tumor and metastatic axillary lymph
synthesis with promising initial results, In more nodes after neoadjuvant chemotherapy for lo-
recent studies paralleling work with cellular prolif- cally advanced breast carcinoma. Cancer J Sci Am
eration tracers in other tumors [76], Pio and col- 1998;4(2):125–31.
[8] McCready DR, Hortobagyi GN, Kau SW, et al.
leagues [77] measured early breast cancer response
The prognostic significance of lymph node
using the thymidine analog, 18F-fluorothymidine metastases after preoperative chemotherapy for
(FLT) with promising early results. Imaging cellular locally advanced breast cancer. Arch Surg 1989;
death or apoptosis as an early measure of therapeu- 124:21–5.
tic efficacy also holds promise, and has been tested [9] Kaklamani V, O’Regan RM. New targeted therapies
in preliminary studies of 99mTc-annexin V [78]. in breast cancer. Semin Oncol 2004;31(2)(Suppl 4):
18 20–5.
F-labeled versions of annexin have been tested
[79–81], and may offer advantages for quantitative [10] Moscovic EC, Mansi JL, King DM, et al. Mam-
serial imaging. mography in the assessment of response to
medical treatment of large primary breast tumor.
Clin Radiol 1993;47:339–44.
Summary [11] Segel MC, Paulus DD, Hortobagyi GN. Advanced
primary breast cancer: assessment at mammog-
The evaluation of breast cancer response to sys- raphy of response to chemotherapy. Radiology
temic therapy is a clinical task to which quantitative 1988;169:49–54.
PET imaging is ideally suited. FDG PET already [12] Husband JE. Monitoring tumor response. Eur
plays a role in response evaluation for neoadjuvant Radiol 1996;6:775–85.
therapy of primary breast cancer and systemic ther- [13] Hoekstra CJ, Paglianiti I, Hoekstra OS, et al.
Monitoring response to therapy in cancer using
apy of metastatic breast cancer. Breast cancer is one
[18F]-2-fluoro-2-deoxy-D-glucose and positron
of the few cancers for which FDG PET is approved emission tomography: an overview of different
by US health care insurances for therapy response analytical methods. Eur J Nucl Med 2000;27:
evaluation. Increasing use of FDG PET to measure 731–43.
breast cancer response is likely, especially in appli- [14] Wahl RL, Zasadny K, Helvie M, et al. Metabolic
cations such as bone-dominant breast cancer, in monitoring of breast cancer chemohormono-
PET & Breast Cancer Therapy 79

therapy using positron emission tomography: [29] Patlak CS, Blasberg RG. Graphical evaluation of
initial evaluation. J Clin Oncol 1993;11:2101–11. blood-to-brain transfer constants from multiple-
[15] Mankoff DA, Dunnwald LK, Gralow JR, et al. time uptake data. Generalizations. J Cereb Blood
Changes in blood flow and metabolism in lo- Flow Metab 1985;5(4):584–90.
cally advanced breast cancer treated with neoad- [30] Gjedde A. Calculation of cerebral glucose phos-
juvant chemotherapy. J Nucl Med 2003;44(11): phorylation from brain uptake of glucose ana-
1806–14. logs in vivo: a re-examination. Brain Res 1982;
[16] Graham MM, Peterson LM, Hayward RM. Com- 257(2):237–74.
parison of simplified quantitative analyses of [31] Mankoff DA, Dunnwald LK, Gralow JR, et al.
FDG uptake. Nucl Med Biol 2000;27(7):647–55. Blood flow and metabolism in locally advanced
[17] Huang S-C. Anatomy of SUV. Nucl Med Biol breast cancer: relationship to response to ther-
2000;27:643–6. apy. J Nucl Med 2002;43(4):500–9.
[18] Lammertsma AA. Measurement of tumor re- [32] Beahrs OH. Staging of cancer. CA Cancer J Clin
sponse using [18F]-2-fluoro-2-deoxy-D-glucose 1991;41(2):121–5.
and positron-emission tomography. J Clin Phar- [33] Schwartz GF, Birchansky CA, Komarnicky LT,
macol 2001;41:104–6. et al. Induction chemotherapy followed by breast
[19] Mankoff DA, Muzi M, Krohn KA. Quantita- conservation for locally advanced carcinoma of
tive positron emission tomography imaging to the breast. Cancer 1994;73:362–9.
measure tumor response to therapy: what is [34] Schwartz GF, Hortobagyi GN. Proceedings of the
the best method? Mol Imaging Biol 2003;5(5): Consensus Conference on Neoadjuvant Che-
281–5. motherapy in Carcinoma of the Breast, April
[20] Keyes JWJ. SUV: standard uptake or silly useless 26–28, 2003, Philadelphia, Pennsylvania. Cancer
value? J Nucl Med 1995;36:1836–9. 2004;100(12):2512–32.
[21] Beaulieu S, Kinahan P, Tseng J, et al. SUV varies [35] Wong ZW, Ellis MJ. Neoadjuvant endocrine
with time after injection in (18)F-FDG PET of therapy for breast cancer: an overlooked option?
breast cancer: characterization and method to Oncology (Williston Park) 2004;18(4):411–20.
adjust for time differences. J Nucl Med 2003; [36] Valagussa P, Zambetti M, Bonadonna G, et al.
44(7):1044–50. Prognostic factors in locally advanced nonin-
[22] Thie JA, Hubner KF, Smith GT. Optimizing imag- flammatory breast cancer. Long-term results
ing time for improved performance in oncology following primary chemotherapy. Breast Ca Res
PET studies. Mol Imaging Biol 2002;4(3):238–44. Treat 1990;15:137–47.
[23] Sokoloff L, Reivich M, Kennedy C, et al. The [37] Wolmark N, Wang J, Mamounas E, et al.
[14C]deoxyglucose method for the measurement Preoperative chemotherapy in patients with
of local cerebral glucose utilization: theory, operable breast cancer: nine-year results from
procedure, and normal values in the conscious National Surgical Adjuvant Breast and Bowel
and anesthetized albino rat. J Neurochem 1977; Project B-18. J Natl Cancer Inst Monogr 2001;
28:897–916. 30:96–102.
[24] Reivich M, Alavi A, Wolf A, et al. Glucose [38] Minn H, Soini I. [18F]fluorodeoxyglucose scintig-
metabolic rate kinetic model parameter determi- raphy in diagnosis and follow up of treatment
nation in humans: the lumped constant and rate in advanced breast cancer. Eur J Nucl Med 1989;
constants for [18F]fluorodeoxyglucose and [11C] 15:61–6.
deoxyglucose. J Cereb Blood Flow Metab 1985;5: [39] Nason KS, Anderson BO, Byrd DR, et al.
179–92. Increased false negative sentinel node biopsy
[25] Phelps M, Huang S, Hoffman E. Tomographic rates after preoperative chemotherapy for inva-
measurement of local cerebral glucose metabolic sive breast carcinoma. Cancer 2000;89(11):
rate in humans with (18F)2-fluoro-2-deoxy-D- 2187–94.
glucose: validation of method. Ann Neurol 1979; [40] Eubank WB, Mankoff DA. Current and future
6(5):371–88. uses of positron emission tomography in breast
[26] Spence A, Muzi M, Graham M, et al. Glucose cancer imaging. Semin Nucl Med 2004;34(3):
metabolism in human malignant gliomas mea- 224–40.
sured quantitatively with PET, 1-[C-11]glucose [41] Bellon JR, Livingston RB, Eubank WB, et al.
and FDG: analysis of the FDG lumped constant. Evaluation of the internal mammary lymph
J Nucl Med 1998;39:440–8. nodes by FDG-PET in locally advanced breast
[27] Tseng J, Dunnwald LK, Schubert EK, et al. 18F- cancer (LABC). Am J Clin Oncol 2004;27(4):
FDG kinetics in locally advanced breast can- 407–10.
cer: correlation with tumor blood flow and [42] Bassa P, Kim EE, Inoue T, et al. Evaluation of
changes in response to neoadjuvant chemother- preoperative chemotherapy using PET with
apy. J Nucl Med 2004;45(11):1829–37. fluorine-18-fluorodeoxyglucose in breast cancer.
[28] Zasadny KR, Tatsumi M, Wahl RL. FDG metabo- J Nucl Med 1996;37:931–8.
lism and uptake versus blood flow in women [43] Smith IC, Welch AE, Hutcheon AW, et al.
with untreated primary breast cancers. Eur J Nucl Positron emission tomography using [18F]-
Med Mol Imaging 2003;30(2):274–80. fluorodeoxy-D-glucose to predict the pathologic
80 Mankoff & Dunnwald

response of breast cancer to primary chemother- and osteolytic lesions. J Clin Oncol 1998;16(10):
apy. J Clin Oncol 2000;18:1676–88. 3375–9.
[44] Schelling M, Avril N, Nahrig J, et al. Positron [58] Tam SL, Gralow JR, Livingston RB, et al.
emission tomography using [18F] fluorodeoxy- Serial FDG-PET to monitor treatment of bone-
glucose for monitoring primary chemotherapy in dominant metastatic breast cancer predicts time
breast cancer. J Clin Oncol 2000;18:1689–95. to progression (TTP). Proceedings of ASCO 2005;
[45] Burcombe RJ, Makris A, Pittam M, et al. Evalua- 23:36s.
tion of good clinical response to neoadjuvant [59] Mortimer JE, Dehdashti F, Siegel BA, et al.
chemotherapy in primary breast cancer using Metabolic flare: indicator of hormone respon-
[18F]-fluorodeoxyglucose positron emission to- siveness in advanced breast cancer. J Clin Oncol
mography. Eur J Cancer 2002;38(3):375–9. 2001;19(11):2797–803.
[46] Kim SJ, Kim SK, Lee ES, et al. Predictive value of [60] Beaney R, Jones T, Lammertsma A, et al. Positron
[18F]FDG PET for pathological response of breast emission tomography for in-vivo measurement
cancer to neoadjuvant chemotherapy. Ann Oncol of regional blood flow, oxygen utilisation, and
2004;15(9):1352–7. blood volume in patients with breast carcinoma.
[47] Barranger E, Grahek D, Antoine M, et al. Evalua- Lancet 1984;1(8369):131–4.
tion of fluorodeoxyglucose positron emission [61] Wilson CBJH, Lammertsma AA, McKenzie CG,
tomography in the detection of axillary lymph et al. Measurements of blood flow and exchang-
node metastases in patients with early-stage breast ing water space in breast tumors using positron
cancer. Ann Surg Oncol 2003;10(6):622–7. emission tomography: a rapid and non-invasive
[48] Chen X, Moore MO, Lehman CD, et al. dynamic method. Cancer Res 1992;52:1592–7.
Combined use of MRI and PET to monitor [62] Abraham DC, Jones RC, Jones SE, et al. Evalua-
response and assess residual disease for locally tion of neoadjuvant chemotherapeutic response
advanced breast cancer treated with neoadju- of locally advanced breast cancer by magnetic
vant chemotherapy. Acad Radiol 2004;11(10): resonance imaging. Cancer 1996;78:91–100.
1115–24. [63] Esserman L, Kaplan E, Partridge S, et al. MRI
[49] Seidman A. Introduction. Single-agent or com- phenotype is associated with response to doxo-
bination chemotherapy in metastatic breast can- rubicin and cyclophosphamide neoadjuvant che-
cer. Oncology (Williston Park) 2003;17(Suppl 14): motherapy in stage III breast cancer. Ann Surg
9–14. Oncol 2001;8(6):549–59.
[50] Gennari A, Donati S, Salvadori B, et al. Role of [64] Gilles R, Guinebretiere JM, Toussaint C, et al. Lo-
2-[18F]-fluorodeoxyglucose (FDG) positron emis- cally advanced breast cancer: contrast-enhanced
sion tomography (PET) in the early assessment subtraction MR imaging of response to pre-
of response to chemotherapy in metastatic breast operative chemotherapy. Radiology 1994;191:
cancer patients. Clin Breast Cancer 2000;1(2): 633–8.
156–61. [65] Knopp MV, Brix G, Junkerman HJ, et al. MR
[51] Stafford SE, Gralow JR, Schubert EK, et al. Use of mammography with pharmacokinetic mapping
serial FDG PET to measure the response of bone- for monitoring breast cancer treatment during
dominant breast cancer to therapy. Acad Radiol neoadjuvant chemotherapy. Magn Reson Imag-
2002;9(8):913–21. ing Clin N Am 1994;2(4):633–58.
[52] Nielsen OS, Munro AJ, Tannock IF. Bone [66] Kedar RP, Cosgrove DO, Smith IE, et al. Breast
metastases: pathophysiology and management carcinoma: measurement of tumor response to
policy. J Clin Oncol 1991;9(3):509–24. primary medical therapy with color flow Dopp-
[53] Bast Jr RC, Ravdin P, Hayes DF, et al. 2000 up- ler imaging. Radiology 1994;190:825–30.
date of recommendations for the use of tumor [67] Peters-Engl C, Frank W, Leodolter S, et al. Tumor
markers in breast and colorectal cancer: clini- flow in malignant breast tumors measured by
cal practice guidelines of the American Society Doppler ultrasound: an independent predictor
of Clinical Oncology. J Clin Oncol 2001;19(6): of survival. Breast Cancer Res Treat 1999;54(1):
1865–78. 65–71.
[54] Vogel CL, Schoenfelder J, Shemano I, et al. Wors- [68] Maini CL, Tofani A, Sciuto R, et al. Technetium-
ening bone scan in the evaluation of antitumor 99m-MIBI scintigraphy in the assessment of
response during hormonal therapy of breast neoadjuvant chemotherapy in breast carcinoma.
cancer. J Clin Oncol 1995;13(5):1123–8. J Nucl Med 1997;38:1546–50.
[55] Schneider JA, Divgi CR, Scott AM, et al. Flare on [69] Mankoff DA, Dunnwald LK, Gralow JR, et al.
bone scintigraphy following Taxol chemotherapy Monitoring the response of patients with locally
for metastatic breast cancer. J Nucl Med 1994; advanced breast carcinoma to neoadjuvant che-
35(11):1748–52. motherapy using [technetium-99m]-sestamibi
[56] Mundy GR. Mechanisms of bone metastasis. scintimammography. Cancer 1999;85:2410–23.
Cancer 1997;80(Suppl):1546–56. [70] Tiling R, Kessler M, Untch M, et al. Breast cancer:
[57] Cook GJ, Houston S, Rubens R, et al. Detection monitoring response to neoadjuvant chemother-
of bone metastases in breast cancer by 18FDG apy using Tc-99m sestamibi scintimammogra-
PET: differing metabolic activity in osteoblastic phy. Onkologie 2003;26(1):27–31.
PET & Breast Cancer Therapy 81

[71] Mankoff DA, Dunnwald LK, Gralow JR, et al. [Tc- [76] Shields AF, Mankoff DA, Link JM, et al. Carbon-
99m]-sestamibi uptake and washout in locally 11-thymidine and FDG to measure therapy
advanced breast cancer are correlated with tumor response. J Nucl Med 1998;39(10):1757–62.
blood flow. Nucl Med Biol 2002;29(7):719–27. [77] Pio BS, Park CK, Satyamurthy N, et al. PET with
[72] Dunnwald LK, Gralow JR, Ellis GK, et al. fluoro-L-thmyidine allows early prediction of
Residual tumor uptake of [99mTc]-sestamibi after breast cancer response to chemotherapy. J Nucl
neoadjuvant chemotherapy for locally advanced Med 2003;44(5):76P.
breast carcinoma predicts survival. Cancer 2005; [78] Blankenberg F, Ohtsuki K, Strauss HW. Dying a
103(4):680–8. thousand deaths. Radionuclide imaging of apop-
[73] Dehdashti F, Flanagan FL, Mortimer JE, et al. tosis. Q J Nucl Med 1999;43(2):170–6.
Positron emission tomographic assessment of [79] Grierson JR, Yagle KJ, Eary JF, et al. Production
‘‘metabolic flare’’ to predict response of meta- of [F-18]fluoroannexin for imaging apoptosis
bolic breast cancer to antiestrogen therapy. Eur J with PET. Bioconjug Chem 2004;15(2):373–9.
Nucl Med 1999;26:51–6. [80] Yagle KJ, Eary JF, Tait JF, et al. Evaluation of
18
[74] Huovinen R, Leskinen-Kallio S, Någren K, et al. F-Annexin V as a PET imaging agent in an
Carbon-11-methionine and PET in evaluation of animal model of apoptosis. J Nucl Med 2005;
treatment response of breast cancer. Br J Cancer 46(4):658–66.
1993;67:787–91. [81] Zijlstra S, Gunawan J, Burchert W. Synthesis
[75] Jansson T, Westlin J, Ahlstrom H, et al. Positron and evaluation of a 18F-labelled recombinant
emission tomography studies in patients with annexin-V derivative, for identification and quan-
locally advanced and/or metastatic breast cancer: tification of apoptotic cells with PET. Appl Radiat
a method for early therapy evaluation? J Clin Isot 2003;58(2):201–7.
Oncol 1995;13(6):1470–7.
83

POSITRON
EMISSION
TOMOGRAPHY
PET Clin 1 (2006) 83–94

Histopathologic and Metabolic


Criteria for Assessment of
Treatment Response in
Breast Cancer
a,* a b
Stefanie Sassen, MD , Falko Fend, MD , Norbert Avril, MD

& Neoadjuvant treatment of breast cancer & Limitation of F-18


& Assessment of treatment response by fluorodeoxyglucose-positron emission
anatomical imaging tomography for prediction of treatment
& Assessment of treatment response by response
histopathology & Comparison between histopathologic and
Predictive markers of pathologic metabolic response criteria
response & References
Limitations and future perspectives of
histopathologic response criteria
& Prediction of response to chemotherapy by
F-18 fluorodeoxyglucose-positron
emission tomography

Most breast cancers develop from the terminal phoplasmacytic infiltrate, and frequent lack of estro-
ducto-lobular unit, which comprises the glandular gen receptor expression [1].
component of normal breast parenchyma. There- Grading of breast cancer is usually performed
fore, virtually all breast cancers are adenocarcino- according to Bloom and Richardson, modified by
mas. Invasive ductal carcinoma is the most common Elston and Ellis [2]. It takes into account the fre-
histological type (70%–80%; Fig. 1), followed by quency of mitotic cells (rate of cell division), the
invasive lobular carcinoma (~10%; Fig. 2). A com- architecture, namely tubule formation (percentage
bination of lobular and ductal features is not of cancer composed of tubular structures), and nu-
uncommon [1]. The remaining cases fall into a clear pleomorphism (variation in size and shape
variety of histological types that are generally asso- of nuclei). Each of these features is assigned a score
ciated with less aggressive biologic behavior and a ranging from 1 to 3 (3 indicating higher prolifera-
better prognosis. These include mucinous (mucoid tive activity, nuclear atypia, and less differentiated
or colloid) carcinoma, consisting of neoplastic cells architecture). After adding the scores, breast can-
floating in pools of extravasated mucin; and med- cer is graded as well-differentiated Grade I (score 3,
ullary carcinoma, which is characterized by large 4 or 5), moderately-differentiated Grade II (score 6
pleomorphic cells, a prominent intratumoral lym- or 7), or poorly-differentiated Grade III (score 8

a
Department of Pathology, Technical University Munich, Ismaningerstrasse 22, 81675 Munich, Germany
b
Department of Nuclear Medicine, Barts and the London School of Medicine, Queen Mary, University of
London, West Smithfield (QEII), London, EC1A 7BE, UK
* Corresponding author.
E-mail address: stefanie.sassen@lrz.tum.de (S. Sassen).

1556-8598/06/$ – see front matter © 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.cpet.2005.09.007
pet.theclinics.com
84 Sassen et al

risk for tumor recurrence, especially in patients who


have positive lymph nodes, tumors of more than
1 cm in size, or other unfavorable prognostic fea-
tures. Doxorubicin and cyclophosphamide, often
combined with taxanes, is the most common adju-
vant therapy [10,11]. Anti-estrogen treatment with
tamoxifen or other estrogen antagonists is often ap-
plied to postmenopausal patients who have estro-
gen receptor-positive tumors.
Neoadjuvant (preoperative) chemotherapy is in-
creasingly used for managing patients who have
locally advanced breast cancer. Disease-free and
overall survival and loco-regional control are simi-
Fig. 1. Invasive ductal carcinoma, moderately differ- lar to conventional adjuvant chemotherapy; how-
entiated (grade 2), with features of glandular dif- ever, neoadjuvant chemotherapy reduces the tumor
ferentiation, fibrosis, and diffuse infiltration of the volume, which enables more patients to be treated
surrounding fat. with breast-conserving surgery [12]. In addition,
this approach frequently produces better cosmetic
or 9). The histologic grade is of prognostic rele- results and allows for observation of response
vance, especially in smaller-sized tumors that show to chemotherapy. Approximately 70% of patients
a strong correlation between higher grade and demonstrate clinical response to neoadjuvant che-
shorter disease-free and overall survival [3]. motherapy, but only 20% to 40% have partial or
The prognostic factors in locally advanced breast complete pathologic response [12–14]. The assess-
cancer are similar to those for earlier tumor stages, ment of response to neoadjuvant chemotherapy
with lymph node status and tumor size having the provides important prognostic information, and
strongest effect on survival. The prognostic informa- patients who had complete pathological response
tion of the hormone receptor status is less apparent, were found to have significantly longer disease-free
although earlier studies found a better prognosis for and overall survival than nonresponders [15,16].
patients who had estrogen or progesterone recep- An important aspect of neoadjuvant treatment is
tor positive breast cancer [4,5]. Independent of its that response reflects an individual in-vivo assay of
rather complex prognostic relevance, the assessment tumor chemosensitivity, which ultimately deter-
of hormone receptor status, nowadays usually by mines survival [17].
immunohistochemistry, is very useful to predict re- Monitoring response to treatment is highly rele-
sponse to hormonal treatment. Human epidermal vant in the current clinical setting. Chemotherapy
growth factor receptor 2 (HER-2/ErbB2), a transmem- for breast cancer is still associated with significant
brane protein receptor, is a key component in regu- side effects, such as nausea, vomiting, alopecia, vari-
lating cell growth. Alterations in the HER-2 gene with ous grades of hematological toxicity with the risk
subsequent overexpression of HER-2/neu protein of subsequent (life-threatening) infections, and po-
affect approximately 25% to 30% of breast cancer tential neurotoxicity (eg, taxanes) and cardiotoxicity
patients, and are associated with poor clinical out-
come [6,7]. Other extensively studied prognostic
markers, including measurement of cell prolifera-
tion and mutations in p53, have not consistently
emerged as independent predictors of survival [8,9].

Neoadjuvant treatment of breast cancer


The treatment of newly diagnosed breast cancer
is primarily determined by the extent of disease,
and generally consists of surgery—either breast-
conserving surgery, usually combined with post-
operative radiation therapy, or total mastectomy.
Axillary lymph node dissection has now been re-
placed by sentinel lymph node biopsy in many Fig. 2. Invasive lobular carcinoma, moderately differ-
patients who have clinically negative axillary find- entiated (grade 2), with a typical Indian-file pattern
ings. In addition, adjuvant systemic therapy is fre- and concentrically surrounding non-tumor–involved
quently recommended after surgery to reduce the ducts (arrow).
Histopathologic and Metabolic Response Criteria 85

(eg, anthracyclines). Early identification of tumor end point for the evaluation of new anticancer
resistance allows one to avoid ineffective therapies drugs [24]. Furthermore, dissolving and shrinkage
and unnecessary side effects, and to change to of a tumor mass is the final step in a complex cas-
potentially more effective treatments. The rate of cade of cellular and subcellular changes after initia-
complete pathologic response after primary che- tion of treatment. Frequently, several cycles of
motherapy varies between 3% and 27% [13–16, chemotherapy or other treatment modalities need
18–21]. The largest series of neoadjuvant chemo- to be applied before treatment response can be as-
therapy [14], with 608 breast cancer patients, found sessed by current anatomical imaging. Even the eval-
a complete pathologic response rate of 20%. Of uation of response after completion of treatment
note, in most cases, no definite correlation was can pose a challenge. If a residual mass is present,
found between clinical response and pathologic re- it is difficult to differentiate viable tumor tissue from
sponse. Only about one third of patients with a post-treatment changes such as scarring and fibrosis.
complete clinical response also had a complete
pathologic response [14,18]. Conversely, a com-
plete pathologic response can be observed in some
patients who have partial or no clinical response, Assessment of treatment response by
mainly because of the persistence of collagenous histopathology
scar tissue despite disappearance of tumor cells. Histopathologic tumor regression is the current
gold standard for assessment of treatment response
in several types of tumors. Most criteria have been
Assessment of treatment response by
established for response evaluation of surgical
anatomical imaging
specimens after neoadjuvant chemotherapy. Re-
The ultimate goal of breast cancer treatment is to sponse criteria were initially developed for osteosar-
achieve cure, or at least a complete remission. The coma, with six response grades describing the range
current standard end point for assessing response of histopathological treatment changes based on
to therapy in solid tumors is by measuring the the percentage of residual viable tumor [25].
change in tumor size [22]. Anatomical imaging, Similar histopathologic response criteria have been
predominantly CT, MRI, and ultrasound (US) is defined for gastric cancer [26], esophageal cancer
used to obtain unidimensional or bidimensional [27], pancreatic cancer [28], and non-small–cell lung
measurements of reference tumor lesions from cancer [29]. Generally, the surgical specimens are
pretreatment scans relative to follow-up; however, analyzed for residual viable tumor relative to the
there are several limitations of anatomic imaging- macroscopically identifiable tumor bed. Two thresh-
based assessment of treatment response. According olds have been identified to provide prognostic in-
to the World Health Organization criteria, a tumor formation following neoadjuvant chemotherapy:
is classified as responding when the product of two response defined by less than 10% viable tumor,
perpendicular diameters has decreased by at least and minimal or no response defined by more than
50%. In cases that have multiple lesions, the sum- 50% residual tumor.
mation of the products should decrease by more For breast cancer, histopathological response to
than 50% [23]. More recently, the response evalua- neoadjuvant chemotherapy is based on the pres-
tion criteria in solid tumors (RECIST) define tumor ence or absence of residual tumor and on regressive
response as a decrease of the maximum tumor di- changes within the remaining tumor tissue [20,30].
ameter by at least 30% [22]. For a spherical tumor, Regressive changes are observed in approximately
a decrease of its diameter by 30% corresponds to a 50% of breast cancers after chemotherapy [14,31].
decrease of the product of two perpendicular di- Regression is commonly defined as replacement of
ameters by 50%. Therefore, the RECIST criteria invasive tumor by fibrous or hyaline stroma, along
have been primarily designed to simplify and stan- with a reduction in the relative number of tumor
dardize the tumor size measurement, and have not cells [Figs. 3, 4]. Regressive changes also include
changed the threshold values for definition of a cellular changes, such as cytoplasmic vacuolization
tumor response. Despite the widely accepted prac- of tumor cells [Fig. 5] [32,33], an increase in cell
tice of using these criteria, it is important to note size, and the presence of giant tumor cells [Fig. 6]
that a 30% (or 50%) decrease in tumor size is a [14,20,34,35]. Regression in the tumoral stroma is
more or less arbitrary convention, which is not characterized by lymphoplasmacytic or histiocytic
based on results of outcome studies. In fact, the infiltrates, iron-storing macrophages, or calcifica-
correlation between radiographic tumor response tions [14,36]. Common changes within the normal
and patient outcome is frequently weak. Therefore, breast tissue after chemotherapy include lobular
tumor response as defined by anatomical imaging atrophy, sclerosis of ductal and ductolobular base-
techniques is frequently not accepted as a surrogate ment membranes, and mild epithelial atypia [34].
86 Sassen et al

Fig. 3. Highly pleomorphic tumor cells scattered within


Fig. 5. Cytoplasmic vacuolization of tumor cells (arrows).
extensive hyaline fibrosis.
compared with the whole patient group [37]. A
Chevallier and colleagues [30] and Sinn and co-
significantly longer overall survival in breast cancer
workers [20] defined various response grades fol-
patients who had pathologic complete response
lowing neoadjuvant chemotherapy of breast cancer
after neoadjuvant chemotherapy has also been re-
using opposite scaling directions [Table1]. Patho-
ported by others [13,14,16,19,38].
logic complete response after neoadjuvant che-
Honkoop and colleagues [38,39] suggested a
motherapy is generally defined by the absence of
modified classification for pathologic response. In
residual invasive tumor. Consequently, patients
42 breast cancer patients, they found no difference
who have residual ductal carcinoma in situ are
in disease-free and overall survival for patients who
still classified as pathologic complete response in
had scattered microscopic foci of tumor cells and
most studies [see Fig. 4]. The largest study of
no macroscopically visible residual tumor, com-
neoadjuvant chemotherapy in breast cancer, the
pared with patients who had complete pathologic
National Surgical Adjuvant Breast and Bowel Pro-
response. These two groups were combined in a re-
ject (NSABP) B-18 trial [37], showed a significant
sponse category ‘‘minimal residual disease’’ (MRD),
correlation between primary tumor response and
and all other patients were classified as ‘‘gross re-
patient outcome. At 9 years of follow-up, the over-
sidual disease’’ (GRD). Patients who had MRD after
all survival rate for patients who had a pathologic
chemotherapy had a 2-year overall survival rate of
complete response was 85%, compared with 73%
94%, compared with 68% for patients who had
in patients who had residual invasive tumor [37].
GRD. The respective rates for disease-free survival
The respective rates for disease-free survival were
were 80% and 45% [38].
75% and 58%. After adjustment for other prognos-
The presence of axillary lymph node metastasis is
tic factors, patients who had a pathologic complete
generally associated with a poor prognosis. The
response had a 50% reduction in risk of death
lymph node status remains an important inde-
pendent prognostic factor, even after neoadjuvant
chemotherapy. Disease-free and overall survival
correlated significantly with the number of resid-

Fig. 4. Residual intraductal carcinoma within fibrotic


scar tissue (arrow). Intraductal residual tumor is com-
monly classified as complete pathologic response
(response Grade III [20].). Fig. 6. The appearance of giant tumor cells (arrow).
Histopathologic and Metabolic Response Criteria 87

Table 1: Histopathologic response classifications [42,47]. A measurable increase in apoptosis was


for breast cancer observed within 1 to 2 days after initiation of che-
motherapy in breast cancer, but this does not allow
Chevallier Sinn consistently predicting response to chemother-
Response et al [30] et al [20]
apy so far [43,48]. Currently, there are no reliable
No residual tumor Grade I Grade IV markers available to predict treatment response for
No residual invasion Grade II Grade III individual patients based on the pretreatment
tumor breast cancer specimens [11].
Minimal residual — Grade II In-vitro chemosensitivity testing in breast cancer
tumor with
was introduced over 2 decades ago [49]; however,
extensive fibrosis
the need for adequate tissue samples and low yield
Residual tumor Grade III Grade I
with fibrosis and of tumor cells from breast biopsies has limited
regressive changes the implementation in clinical routine use [50]. It
No apparent Grade IV Grade 0 remains uncertain if chemosensitivity of cancer
histopathological cells in two-dimensional (2D) culture assays is
changes comparable to the chemoresponsiveness in vivo,
which is also determined by local tissue-specific
factors, the three-dimensional (3D) microenviron-
ual tumor-involved axillary lymph nodes [14,31,
ment, and individual metabolism and clearance
38,40]. In 135 patients who had locally advanced
of the chemotherapeutic agents. Using an extreme
breast cancer, patients who had tumor-free axillary
drug resistance assay, Mehta and colleagues [51]
lymph nodes after neoadjuvant chemotherapy had
demonstrated in a study of 103 newly diagnosed
an overall 5-year survival rate of 71%, compared
breast cancer patients that extreme drug resistance
with 54% in patients who had one to nine tumor-
scores were significantly associated with time to
involved lymph nodes [31]. The overall 5-year sur-
tumor progression and overall survival. Newer as-
vival rate was only 35% in patients who had 10 or
says are able to test smaller tumors.
more positive lymph nodes after neoadjuvant che-
motherapy [31]. The NSABP B-18 trial also noted a
Limitations and future perspectives of
significantly longer overall and disease-free survival
histopathologic response criteria
for patients who had negative axillary lymph nodes
compared with either one to three or more than It is important to note that regressive changes such
three tumor-involved lymph nodes after neoadju- as stromal hyalinization and fibrosis, necrosis, or
vant chemotherapy [14]. There was a significant reactive inflammation may be a priori present in
correlation between the pathologic response of the breast cancer specimens. The NSABP B-18 trial [14]
primary tumors and the axillary lymph nodes noted a higher frequency of cytoplasmic vacuoliza-
[14,16,31]. Kuerer and coworkers [16] studied the tion in tumor cells, the presence of giant tumor
histopathologic response of lymph nodes in 356 pa- cells, and stromal hyalinization in patients who
tients, and noted residual tumor involvement of had received preoperative chemotherapy. Most of
axillary lymph nodes in 28% of patients who these histologic changes have a moderate-to-high
achieved a complete pathologic response in their specificity (48%–99%), but relatively low sensitivity
primary tumor, compared with 74% of patients (7%–67%), when compared with nontreated tumor
who did not have a complete pathologic response. specimens. Thus, there is no single morphologic
marker that identifies treatment-related changes
with high accuracy in the majority of patients [14].
Predictive markers of pathologic response In contrast to complete pathological response,
A complete pathologic response of the primary the presence of chemotherapy-induced regressive
tumor and of axillary lymph nodes after che- changes was not related to better patient outcome
motherapy is an important predictive factor for [14,52]. The clinical relevance and potential prog-
survival. Pathologic response was consistently cor- nostic information of histomorphologic and cyto-
related with smaller tumor size at initial diagnosis pathic changes after chemotherapy still remains to
[16,18,31] and a lack of estrogen-receptor expres- be defined. The histopathologic assessment of sur-
sion [16,41,42]. In several studies, including the gical specimens may also be affected by sampling
NSABP B-18 trial [14,16,43], a high nuclear grade error, depending on the technique used to examine
at initial diagnosis was predictive of a subsequent the initial tumor bed. Currently, pathologic response
pathologic response to chemotherapy. Mutations is evaluated after several cycles of chemotherapy
in the p53 gene did not predict response to che- (three to six cycles) or several months of endocrine
motherapy in breast cancer [44–46]. Results regard- therapy; however, this approach does not address
ing the role of HER-2/neu are still conflicting the early identification of nonresponders, which is
88 Sassen et al

of critical importance because these patients may tion in tissue, which is particularly important for
benefit from alternative treatment regimens [53]. monitoring therapeutic response. Frequently, stan-
Recently, various molecular markers, including dardized uptake values (SUV) are being calculated,
large-scale gene expression profiling, and the pres- providing a semiquantitative measure of FDG ac-
ence of tumor cells in the blood have been sug- cumulation in tissue by normalizing the tissue
gested to be helpful in predicting response to radioactivity concentration measured with PET to
treatment [54,55]. Gene expression profiling with injected dose and patient’s body weight [62]. Pre-
microarrays allows simultaneous assessment of vious studies have demonstrated that SUV provide
thousands of genes potentially involved in the sen- highly reproducible parameters of tumor glucose
sitivity or resistance of cancer cells to chemotherapy use [63,64]. Weber and coworkers [64] studied
[56]. In breast cancer, recent studies identified gene 16 patients twice within 10 days with FDG-PET
expression profiles with independent prognos- while they were receiving no therapy. None of vari-
tic information in early disease stages [57,58]. A ous semiquantitative parameters of FDG uptake
70-gene profile predicted the outcome in young showed a significant increase or decrease between
patients who had Stage I or II breast cancer more the two PET scans. The study authors found that
accurately than standard clinical and histological changes of a parameter of more than 20% are out-
criteria [58]. A poor-prognosis gene profile was side the 95% range for spontaneous fluctuations,
associated with a fivefold increased relative risk of and can be considered to reflect true changes in
distant metastases as compared with patients who tumor glucose metabolism. At least two sequential
had a good-prognosis profile. Recent studies also FDG-PET scans are currently necessary for predic-
focused on the potential use of gene expression tion of treatment response: one PET scan before
profiles for prediction of response to neoadjuvant treatment serving as baseline, and one PET scan
chemotherapy. A gene expression signature com- after initiation of chemotherapy (eg, after the first
prising of 92 genes predicted clinical response to or second cycle). Comparing the percentage change
a taxane-based neoadjuvant chemotherapy with of FDG uptake in tumors provides specific infor-
positive and negative predictive values of 92% mation about the response to a given treatment.
and 83%, respectively [59]. Another group also This characteristic behavior of tumor metabolism is
identified a gene expression signature comprising based on the observation that the majority of
of 74 genes that predicted pathologic response with changes in FDG uptake occur early after initiation
an accuracy of 78% and a positive predictive value of treatment. In successfully treated non-Hodgkin’s
of 100% [60]. Principal limitations of this promis- lymphoma for example, the FDG uptake decreased
ing approach currently include the need for ade- by 60% to 67% from baseline to day 7 [65]. Simi-
quate tissue samples, tumor heterogeneity, and lar changes were also observed in breast cancer
host factors such as drug delivery and metabolism [66,67].
that may not be reflected by gene expression pro- In 1993, Wahl and colleagues [68] studied
files of tumor cells. 11 women who had newly diagnosed, locally ad-
vanced primary breast cancers and who were un-
dergoing chemo-hormonotherapy with sequential
FDG-PET imaging. Patients had a baseline and four
Prediction of response to chemotherapy by
follow-up PET scans during the first three cycles of
F-18 fluorodeoxyglucose-positron emission
treatment, and final tumor response was deter-
tomography
mined by histopathology after nine cycles of treat-
The uptake mechanism and biochemical pathway ment. The FDG uptake in 8 patients who had
of the glucose analog [F-18] fluorodeoxyglucose partial or complete pathologic responses decreased
(FDG) has been extensively studied in vitro and promptly with treatment. On day 8 of therapy,
in vivo. The transport of the radiotracer through mean tumor FDG uptake was 78% of baseline,
the cell membrane via glucose transport proteins and decreased further to 68% at day 21, 60% at
(GLUT) and subsequent intracellular phosphoryla- day 42, and 52% at day 63. In contrast, 3 patients
tion by hexokinase (HK) have been identified as who had nonresponding tumors did not show a
key steps for subsequent tissue accumulation [61]. significant decrease in FDG uptake. In 30 breast
Because FDG-6-phosphate is not a suitable substrate cancer patients who received eight doses of primary
for glucose-6-phosphate isomerase, and the enzyme chemotherapy, the mean reduction in FDG uptake
levels of glucose-6-phosphatase are generally low in after the first cycle of chemotherapy was signifi-
tumors, the intracellular accumulation of FDG-6- cantly higher in lesions with partial, complete mac-
phosphate reflects tissue glucose metabolism. roscopic, or complete microscopic response [67].
Attenuation-corrected PET images provide quan- After a single cycle of chemotherapy, PET predicted
titative information about the tracer concentra- complete pathologic response with a sensitivity of
Histopathologic and Metabolic Response Criteria 89

90% and specificity of 74%. Schelling and cowork- at later time points during and after chemotherapy
ers [66] compared results from PET imaging with [Fig. 7] [69–71].
pathological response using distinct histopatho- It is important to note that a transient increase
logical criteria, namely MRD and GRD, previously in glucose use has been found in responding tu-
identified to provide prognostic information [38]. mors treated with hormonotherapy. In a first series,
FDG uptake in breast cancer after the first and Dehdashti and colleagues [72] performed FDG-PET
second cycle of chemotherapy was compared with in 11 women before and approximately 1 week
baseline PET. Patients classified as responders after initiation of tamoxifen therapy. In all patients,
by histopathology had a significantly more pro- clinical and radiological follow-up was performed
nounced decrease in FDG uptake than nonre- with a median interval of 12 months. Seven pa-
sponding patients. Therefore, responding and tients responded, and all showed an initial increase
nonresponding tumors could be differentiated by of FDG uptake 1 week after therapy. This ‘‘meta-
PET as early as after the first cycle of chemotherapy. bolic flare’’ effect is a recognized side effect of anti-
By a threshold defined as an SUV decrease below estrogen therapy, which is clinically characterized
55% compared with the baseline, all responders by pain and erythema in soft-tissue lesions and
were correctly identified after the first cycle (sensi- increased pain in osseous metastases. An expla-
tivity 100%, specificity 85%). The accuracy in pre- nation for this metabolic flare effect is that anti-
dicting histopathological response was 88% after estrogen therapy first has an agonist effect before
the first cycle of therapy and 91% after the second. the antagonist effect overrules. This agonist effect
Other studies also addressed the role of FDG-PET occurs within the first 1 to 2 weeks after the begin-

Fig. 7. Sixty-eight-year-old breast cancer patient, postmastectomy of the left breast. Baseline FDG-PET/CT before
chemotherapy (A) demonstrates recurrence in the right axilla. FDG-PET/CT after chemotherapy (B) shows a
decrease in the size of axillary lymph nodes, but mild increased FDG uptake indicates viable residual tumor.
90 Sassen et al

ning of a treatment, and is usually followed by FDG-PET appears to be the identification of non-
disease remission. Recently, the same group con- responders who are characterized by virtually no
firmed their findings in a larger series including change in FDG uptake after initiation of che-
40 patients [73]. In responders, the tumor FDG up- motherapy. The overall survival in patients who
take increased after tamoxifen by 28.4%, with only did not respond according to FDG-PET was 8.8 ±
5 of these patients having evidence of a clinical flare 6.7 months, compared with 19.2 ± 13.6 months in
reaction. In nonresponders, there was no signifi- responding patients. An important finding of the
cant change in tumor FDG uptake from baseline. study from Dose Schwarz and coworkers [77] is
There are no reports so far about metabolic flare that by using a PET-based strategy, 14 cycles of
in more aggressive chemotherapeutic regimens. chemotherapy could have been avoided in 5 non-
There are significant differences between the neo- responding patients, and they could have earlier
adjuvant setting and the treatment of metastatic received second-line chemotherapy.
breast cancer. Generally, more aggressive chemo-
therapeutic regimens are used in the neoadjuvant
Limitation of F-18
setting compared with a large variety of treatment
fluorodeoxyglucose-positron emission
options available for metastatic disease. Modern
tomography for prediction of treatment
treatment regimens are developing toward individual-
response
ized treatment tailored to the need of each patient,
emphasizing the importance of methods for predic- Although numerous studies indicate that SUV may
tion of treatment response. There is little informa- be used to determine quantitative changes in tumor
tion available on the clinical utility of sequential glucose use over time, it is important to note that
FDG-PET imaging in patients who have metastatic adherence to a strict PET protocol is required to
disease. A decrease in FDG uptake was observed 6 minimize the variation between FDG-PET studies.
to 13 days after the first cycle of chemotherapy in 8 The radioactivity in the syringe should be measured
out of 12 patients with metastatic disease who had before and after injection, and corrected for radio-
responded to chemotherapy [74]. Gennari and co- active decay to accurately determine the dose of
workers [75] also observed a rapid and significant FDG injected. Extravasation of the radiotracer dur-
decrease in tumor glucose metabolism after the first ing injection will influence quantification of PET
course of chemotherapy in 6 out of 9 responding imaging. The radioactivity concentration in the
patients, and no significant decrease in nonrespond- body measured by PET is affected by the time
ing patients. A close correlation was found between interval between tracer administration and PET
changes in FDG uptake and the overall clinical imaging. Many institutions use a 60-minute uptake
assessment of response in breast cancer patients period in which the patient is resting before ac-
who had bone metastases [76]. quiring the PET scan. The uptake period needs to
Dose Schwarz and colleagues [77] recently con- be standardized, and should vary less than 5 min-
firmed previous reports on the predictive informa- utes between baseline and follow-up PET. Beaulieu
tion of early changes in glucose metabolism after and colleagues [78] recently described a method to
initiation of chemotherapy in patients who had adjust for time differences. There is an inverse rela-
metastatic breast cancer. Changes in tumor FDG tionship between FDG uptake and blood glucose
uptake were statistically significantly different be- level that has to taken into account for the compari-
tween responding and nonresponding metastatic son of serial PET scans. The patient’s blood glucose
lesions after the first and second cycle of chemo- levels should be comparable between the PET
therapy. When compared with the baseline FDG- scans, and ideally less than 150 mg/dl at the time
PET, the FDG uptake in responding metastatic of tracer injection. The specific method for calcula-
lesions decreased to 72 ± 21% after the first cycle, tion of SUV, for example, normalization to body
and to 54 ± 16% after the second cycle of che- weight or the lean body mass, is less important for
motherapy. In contrast, the FDG uptake in metas- the purpose of therapy monitoring because intra-
tases not responding to chemotherapy declined individual comparisons of SUV are being made.
only to 94 ± 19% after the first cycle, and to 79 ± When FDG-PET is used for early assessment of ther-
9% after the second cycle of chemotherapy. FDG- apy response, significant changes in body weight
PET whole-body imaging correctly predicted the influencing the results of the SUV calculation are
response in all patients as early as after the first unlikely. In contrast, normalization to body surface
cycle (3 weeks) of chemotherapy. FDG-PET was area or lean body mass is preferable for interindi-
more accurate than conventional imaging proce- vidual comparison of SUV, especially if the study
dures obtained after the third cycle (9 weeks) of population includes obese patients [62].
chemotherapy, which misclassified three patients The SUV measurements are also affected by the
(27%) falsely as responders. A specific strength of size of a lesion. Partial volume effects cause signifi-
Histopathologic and Metabolic Response Criteria 91

cant underestimation of isotope concentration in of tissue specimens, and therapeutic changes are
structures smaller than two times the scanner reso- assessed in vitro on a microscopic, cellular level.
lution at full-width, half-maximum (FWHM). Ap- In contrast, FDG-PET imaging provides informa-
propriate recovery coefficients, which represent the tion about tumor metabolism in vivo, although
ratio of apparent to true isotope concentration in on a macroscopic level. Whereas histopathological
the structure of interest, can be used to correct for response assessment requires invasive procedures
partial volume effects. SUV are obtained by placing to obtain surgical specimens and is routinely only
a region of interest (ROI) on the PET images. Of performed after completion of therapy, PET imag-
note, the size of the ROI affects the radioactivity ing may provide early response evaluation. FDG-
measurements—the larger the ROI, the lower the PET imaging allows monitoring of lesions in soft
mean SUV. In the clinical setting, the maximum tissue, lymph nodes, the liver, the lungs, and in the
SUV within the ROI, which represents the highest bone in one imaging procedure, in contrast to
radioactivity concentration in one voxel within the histopathology. Histopathology is affected by the
tumor, is frequently being used for comparison be- experience of the pathologist, with some unavoid-
tween PET studies. Of note, quantitative measure- able observer variability. PET provides quantitative
ments with PET require appropriate and regularly measurements that are less observer-dependent. On
quality control, including scanner cross calibration. the other hand, histopathology is widely available,
In the Unites States, health insurance providers whereas PET imaging is generally less accessible,
reimburse monitoring treatment response in breast although it is becoming standard of care in major
cancer, as an adjunct to standard imaging modali- hospitals throughout the United States. Histopa-
ties for women who have locally advanced and thology is currently the gold standard for assess-
metastatic breast cancer, when a change in ther- ment of treatment response, with promising new
apy is anticipated. Specific criteria to determine re- developments in genetic and proteomic profiling.
sponse by FDG-PET have yet to be established, Sequential PET imaging, although promising, needs
however. The threshold to differentiate between to be validated and its criteria standardized in
responders and nonresponders based on changes appropriately sized prospective clinical trials before
in FDG uptake is of critical importance. Chemo- it can and should be used in the clinical setting. The
therapy should not be discontinued in respond- metabolic information from PET requires thorough
ing patients, even at the cost of not identifying evaluation regarding differences in disease-free and
all nonresponding patients. Weber and coworkers overall survival of metabolic responders versus
[64,79] recently proposed to define metabolic re- nonresponders, as well as evaluation of the cost-
sponse as an SUV decrease larger than two times effectiveness of this approach.
the standard deviation (20%) of spontaneous
changes in FDG uptake. In breast cancer, a decrease
in FDG uptake of more than 20% compared with
baseline correctly identified 5 out of 7 (71.4%)
References
nonresponding lesions, and 12 out of 14 (85.7%) [1] Rosai J, editor. Rosai and Ackermann’s surgical
responding lesions after the first cycle of che- pathology. 9th edition. Philadelphia: Mosby,
motherapy [77]. The proposed approach is also Elsevier; 2004. p. 1787–827.
supported by a recent study in non-small–cell [2] Elston CW, Ellis IO. Pathological prognostic fac-
lung cancer, in which this criterion was prospec- tors in breast cancer. I. The value of histological
grade in breast cancer: experience from a large
tively applied to 57 patients who had advanced
study with long-term follow-up. Histopathology
stage of disease [79]. Only 1 out of 27 metabolic 1991;19:403–10.
nonresponders achieved a partial response after [3] Genestie C, Zafrani B, Asselain B, et al. Compari-
completion of chemotherapy, which resulted in a son of the prognostic value of Scarff-Bloom-
negative predictive value of 96%; however, this Richardson and Nottingham histological grades
strategy needs to be validated in a separate prospec- in a series of 825 cases of breast cancer: major
tive study for metastatic breast cancer patients. importance of the mitotic count as a component
of both grading systems. Anticancer Res 1998;18:
571–6.
[4] Aamdal S, Bormer O, Jorgensen O, et al. Estrogen
Comparison between histopathologic and receptors and long-term prognosis in breast
metabolic response criteria cancer. Cancer 1984;53:2525–9.
[5] Butler JA, Bretsky S, Menendez-Botet C, et al. Es-
Important differences between histopathology and trogen receptor protein of breast cancer as a pre-
FDG-PET imaging for assessment of treatment dictor of recurrence. Cancer 1985;55:1178–81.
response in breast cancer should be considered. [6] Slamon DJ, Godolphin W, Jones LA, et al. Stud-
Histopathology is based on morphologic features ies of the HER-2/neu proto-oncogene in human
92 Sassen et al

breast and ovarian cancer. Science 1989;244: biomarkers in predicting pathologic response to
707–12. anthracycline-based neoadjuvant chemotherapy
[7] Menard S, Fortis S, Castiglioni F, et al. HER2 as in patients with breast carcinoma. Cancer 2002;
a prognostic factor in breast cancer. Oncology 94:3107–14.
2001;61(Suppl 2):67–72. [22] Therasse P, Arbuck SG, Eisenhauer EA, et al. New
[8] Barnes DM, Dublin EA, Fisher CJ, et al. Im- guidelines to evaluate the response to treatment
munohistochemical detection of p53 protein in in solid tumors. European Organization for Re-
mammary carcinoma: an important new in- search and Treatment of Cancer, National Cancer
dependent indicator of prognosis? Hum Pathol Institute of the United States, National Cancer
1993;24:469–76. Institute of Canada. J Natl Cancer Inst 2000;92:
[9] Rosen PP, Lesser ML, Arroyo CD, et al. p53 in 205–16.
node-negative breast carcinoma: an immuno- [23] Miller AB, Hoogstraten B, Staquet M, et al. Re-
histochemical study of epidemiologic risk factors, porting results of cancer treatment. Cancer 1981;
histologic features, and prognosis. J Clin Oncol 47:207–14.
1995;13:821–30. [24] Buyse M, Thirion P, Carlson RW, et al. Relation
[10] Cocconi G. Chemotherapy with and without an- between tumour response to first-line chemo-
thracycline. J Clin Oncol 2000;18:1392–3. therapy and survival in advanced colorectal can-
[11] Hamilton A, Hortobagyi G. Chemotherapy: what cer: a meta-analysis. Meta-Analysis Group in
progress in the last 5 years? J Clin Oncol 2005; Cancer. Lancet 2000;356:373–8.
23:1760–75. [25] Salzer-Kuntschik M, Brand G, Delling G. [Deter-
[12] van der Hage JA, van de Velde CJ, Julien JP, et al. mination of the degree of morphological regres-
Preoperative chemotherapy in primary opera- sion following chemotherapy in malignant bone
ble breast cancer: results from the European tumors]. Pathologe 1983;4:135–41 [in German].
Organization for Research and Treatment of Can- [26] Becker K, Mueller JD, Schulmacher C, et al.
cer trial 10902. J Clin Oncol 2001;19:4224–37. Histomorphology and grading of regression in
[13] Bonadonna G, Veronesi U, Brambilla C, et al. gastric carcinoma treated with neoadjuvant che-
Primary chemotherapy to avoid mastectomy in motherapy. Cancer 2003;98:1521–30.
tumors with diameters of three centimeters or [27] Mandard AM, Dalibard F, Mandard JC, et al.
more. J Natl Cancer Inst 1990;82:1539–45. Pathologic assessment of tumor regression after
[14] Fisher ER, Wang J, Bryant J, et al. Pathobiology preoperative chemoradiotherapy of esophageal
of preoperative chemotherapy: findings from the carcinoma. Clinicopathologic correlations. Can-
National Surgical Adjuvant Breast and Bowel cer 1994;73:2680–6.
(NSABP) protocol B-18. Cancer 2002;95:681–95. [28] Evans DB, Rich TA, Byrd DR, et al. Preoperative
[15] Feldman LD, Hortobagyi GN, Buzdar AU, et al. chemoradiation and pancreaticoduodenectomy
Pathological assessment of response to induction for adenocarcinoma of the pancreas. Arch Surg
chemotherapy in breast cancer. Cancer Res 1986; 1992;127:1335–9.
46:2578–81. [29] Junker K, Thomas M, Schulmann K, et al.
[16] Kuerer HM, Newman LA, Smith TL, et al. Clinical [Regression grading of neoadjuvant non-small-
course of breast cancer patients with complete cell lung carcinoma treatment]. Pathologe 1997;
pathologic primary tumor and axillary lymph 18:131–40 [in German].
node response to doxorubicin-based neoadjuvant [30] Chevallier B, Roche H, Olivier JP, et al. Inflam-
chemotherapy. J Clin Oncol 1999;17:460–9. matory breast cancer. Pilot study of intensive
[17] Fisher B, Bryant J, Wolmark N, et al. Effect of induction chemotherapy (FEC-HD) results in a
preoperative chemotherapy on the outcome of high histologic response rate. Am J Clin Oncol
women with operable breast cancer. J Clin Oncol 1993;16:223–8.
1998;16:2672–85. [31] Gajdos C, Tartter PI, Estabrook A, et al. Relation-
[18] Chollet P, Amat S, Cure H, et al. Prognostic sig- ship of clinical and pathologic response to neo-
nificance of a complete pathological response adjuvant chemotherapy and outcome of locally
after induction chemotherapy in operable breast advanced breast cancer. J Surg Oncol 2002;80:
cancer. Br J Cancer 2002;86:1041–6. 4–11.
[19] Machiavelli MR, Romero AO, Perez JE, et al. [32] Aktepe F, Kapucuoglu N, Pak I. The effects of
Prognostic significance of pathological response chemotherapy on breast cancer tissue in locally
of primary tumor and metastatic axillary lymph advanced breast cancer. Histopathology 1996;29:
nodes after neoadjuvant chemotherapy for lo- 63–7.
cally advanced breast carcinoma. Cancer J Sci Am [33] Seno R, Sparano JA, Fineberg SA. Gross and
1998;4:125–31. histologic features of locally advanced breast
[20] Sinn HP, Schmid H, Junkermann H, et al. [His- cancer after neoadjuvant chemotherapy. Anat
tologic regression of breast cancer after pri- Pathol 1998;3:169–80.
mary (neoadjuvant) chemotherapy]. Geburtshilfe [34] Kennedy S, Merino MJ, Swain SM, et al. The ef-
Frauenheilkd 1994;54:552–8 [in German]. fects of hormonal and chemotherapy on tumoral
[21] Wang J, Buchholz TA, Middleton LP, et al. As- and nonneoplastic breast tissue. Hum Pathol
sessment of histologic features and expression of 1990;21:192–8.
Histopathologic and Metabolic Response Criteria 93

[35] Rasbridge SA, Gillett CE, Seymour AM, et al. The neoadjuvant chemotherapy for breast cancer.
effects of chemotherapy on morphology, cellular Endocr Relat Cancer 2002;9:183–95.
proliferation, apoptosis and oncoprotein expres- [48] Buchholz TA, Davis DW, McConkey DJ, et al.
sion in primary breast carcinoma. Br J Cancer Chemotherapy-induced apoptosis and Bcl-2 lev-
1994;70:335–41. els correlate with breast cancer response to che-
[36] Moreno A, Escobedo A, Benito E, et al. Patho- motherapy. Cancer J 2003;9:33–41.
logic changes related to CMF primary chemo- [49] Oude Elferink RF, Goldschmidt HM, Majoor HJ,
therapy in breast cancer. Pathological evaluation et al. Rapid assay for the in vitro chemosensitivity
of response predicts clinical outcome. Breast testing of human breast tumours. Control of
Cancer Res Treat 2002;75:119–25. assay conditions and quality assurance. Pharm
[37] Wolmark N, Wang J, Mamounas E, et al. Pre- Weekbl Sci 1989;11:155–60.
operative chemotherapy in patients with op- [50] Krasna L, Netikova I, Chaloupkova A, et al. As-
erable breast cancer: nine-year results from sessment of in vitro drug resistance of human
National Surgical Adjuvant Breast and Bowel breast cancer cells subcultured from biopsy speci-
Project B-18. J Natl Cancer Inst Monogr 2001; mens. Anticancer Res 2003;23:2593–9.
30:96–102. [51] Mehta RS, Bornstein R, Yu IR, et al. Breast cancer
[38] Honkoop AH, van Diest PJ, de Jong JS, et al. survival and in vitro tumor response in the ex-
Prognostic role of clinical, pathological and treme drug resistance assay. Breast Cancer Res
biological characteristics in patients with locally Treat 2001;66:225–37.
advanced breast cancer. Br J Cancer 1998;77: [52] Ziegler LD, Connelly JH, Frye D, et al. Lack of
621–6. correlation between histologic findings and re-
[39] Honkoop AH, Pinedo HM, De Jong JS, et al. sponse to chemotherapy in metastatic breast
Effects of chemotherapy on pathologic and bio- cancer. Cancer 1991;68:628–33.
logic characteristics of locally advanced breast [53] Thomas E, Holmes FA, Smith TL, et al. The use of
cancer. Am J Clin Pathol 1997;107:211–8. alternate, non-cross-resistant adjuvant chemo-
[40] Pierga JY, Mouret E, Dieras V, et al. Prognostic therapy on the basis of pathologic response to
value of persistent node involvement after neo- a neoadjuvant doxorubicin-based regimen in
adjuvant chemotherapy in patients with opera- women with operable breast cancer: long-term
ble breast cancer. Br J Cancer 2000;83:1480–7. results from a prospective randomized trial.
[41] Buzdar AU, Singletary SE, Theriault RL, et al. J Clin Oncol 2004;22:2294–302.
Prospective evaluation of paclitaxel versus com- [54] Cristofanilli M, Budd GT, Ellis MJ, et al. Cir-
bination chemotherapy with fluorouracil, doxo- culating tumor cells, disease progression, and
rubicin, and cyclophosphamide as neoadjuvant survival in metastatic breast cancer. N Engl J
therapy in patients with operable breast cancer. Med 2004;351:781–91.
J Clin Oncol 1999;17:3412–7. [55] Shimizu D, Ishikawa T, Ichikawa Y, et al. Cur-
[42] Colleoni M, Minchella I, Mazzarol G, et al. Re- rent progress in the prediction of chemosensi-
sponse to primary chemotherapy in breast can- tivity for breast cancer. Breast Cancer 2004;11:
cer patients with tumors not expressing estrogen 42–8.
and progesterone receptors. Ann Oncol 2000;11: [56] Perou CM, Sorlie T, Eisen MB, et al. Molecular
1057–9. portraits of human breast tumours. Nature 2000;
[43] Stearns V, Singh B, Tsangaris T, et al. A pro- 406:747–52.
spective randomized pilot study to evaluate pre- [57] Sorlie T, Perou CM, Tibshirani R, et al. Gene
dictors of response in serial core biopsies to expression patterns of breast carcinomas distin-
single agent neoadjuvant doxorubicin or pacli- guish tumor subclasses with clinical implications.
taxel for patients with locally advanced breast Proc Natl Acad Sci USA 2001;98:10869–74.
cancer. Clin Cancer Res 2003;9:124–33. [58] van’t Veer LJ, Dai H, van de Vijver MJ, et al. Gene
[44] MacGrogan G, Mauriac L, Durand M, et al. expression profiling predicts clinical outcome of
Primary chemotherapy in breast invasive carci- breast cancer. Nature 2002;415:530–6.
noma: predictive value of the immunohisto- [59] Chang JC, Wooten EC, Tsimelzon A, et al. Gene
chemical detection of hormonal receptors, p53, expression profiling for the prediction of thera-
c-erbB-2, MiB1, pS2 and GST pi. Br J Cancer peutic response to docetaxel in patients with
1996;74:1458–65. breast cancer. Lancet 2003;362:362–9.
[45] Niskanen E, Blomqvist C, Franssila K, et al. Pre- [60] Ayers M, Symmans WF, Stec J, et al. Gene ex-
dictive value of c-erbB-2, p53, cathepsin-D and pression profiles predict complete pathologic
histology of the primary tumour in metastatic response to neoadjuvant paclitaxel and fluoroura-
breast cancer. Br J Cancer 1997;76:917–22. cil, doxorubicin, and cyclophosphamide chemo-
[46] Rozan S, Vincent-Salomon A, Zafrani B, et al. No therapy in breast cancer. J Clin Oncol 2004;22:
significant predictive value of c-erbB-2 or p53 2284–93.
expression regarding sensitivity to primary che- [61] Avril N. GLUT1 expression in tissue and (18)F-
motherapy or radiotherapy in breast cancer. Int J FDG uptake. J Nucl Med 2004;45:930–2.
Cancer 1998;79:27–33. [62] Zasadny KR, Wahl RL. Standardized uptake val-
[47] Cleator S, Parton M, Dowsett M. The biology of ues of normal tissues at PET with 2-[fluorine-18]-
94 Sassen et al

fluoro-2-deoxy-D-glucose: variations with body in response to neoadjuvant chemotherapy.


weight and a method for correction. Radiology J Nucl Med 2004;45:1829–37.
1993;189:847–50. [72] Dehdashti F, Flanagan FL, Mortimer JE, et al.
[63] Minn H, Zasadny KR, Quint LE, et al. Lung can- Positron emission tomographic assessment of
cer: reproducibility of quantitative measurements ‘‘metabolic flare’’ to predict response of meta-
for evaluating 2-[F-18]-fluoro-2-deoxy-D-glucose static breast cancer to antiestrogen therapy. Eur J
uptake at PET. Radiology 1995;196:167–73. Nucl Med 1999;26:51–6.
[64] Weber WA, Ziegler SI, Thodtmann R, et al. Re- [73] Mortimer JE, Dehdashti F, Siegel BA, et al.
producibility of metabolic measurements in ma- Metabolic flare: indicator of hormone respon-
lignant tumors using FDG PET. J Nucl Med 1999; siveness in advanced breast cancer. J Clin Oncol
40:1771–7. 2001;19:2797–803.
[65] Romer W, Hanauske AR, Ziegler S, et al. Positron [74] Jansson T, Westlin JE, Ahlstrom H, et al. Positron
emission tomography in non-Hodgkin’s lym- emission tomography studies in patients with
phoma: assessment of chemotherapy with fluoro- locally advanced and/or metastatic breast cancer:
deoxyglucose. Blood 1998;91:4464–71. a method for early therapy evaluation? J Clin
[66] Schelling M, Avril N, Nahrig J, et al. Positron Oncol 1995;13:1470–7.
emission tomography using [(18)F]fluorodeoxy- [75] Gennari A, Donati S, Salvadori B, et al. Role
glucose for monitoring primary chemotherapy in of 2-[18F]-fluorodeoxyglucose (FDG) positron
breast cancer. J Clin Oncol 2000;18:1689–95. emission tomography (PET) in the early assess-
[67] Smith IC, Welch AE, Hutcheon AW, et al. ment of response to chemotherapy in metastatic
Positron emission tomography using [(18)F]- breast cancer patients. Clin Breast Cancer 2000;
fluorodeoxy-D-glucose to predict the pathologic 1:156–61 [discussion: 162–3].
response of breast cancer to primary chemother- [76] Stafford SE, Gralow JR, Schubert EK, et al. Use of
apy. J Clin Oncol 2000;18:1676–88. serial FDG PET to measure the response of bone-
[68] Wahl RL, Zasadny K, Helvie M, et al. Metabolic dominant breast cancer to therapy. Acad Radiol
monitoring of breast cancer chemohormono- 2002;9:913–21.
therapy using positron emission tomography: [77] Dose Schwarz J, Bader M, Jenicke L, et al. Early
initial evaluation. J Clin Oncol 1993;11:2101–11. prediction of response to chemotherapy in meta-
[69] Mankoff DA, Dunnwald LK, Gralow JR, et al. static breast cancer using sequential 18F-FDG-PET
Blood flow and metabolism in locally advanced imaging. J Nucl Med 2005;46(7):1144–50.
breast cancer: relationship to response to ther- [78] Beaulieu S, Kinahan P, Tseng J, et al. SUV varies
apy. J Nucl Med 2002;43:500–9. with time after injection in (18)F-FDG PET of
[70] Mankoff DA, Dunnwald LK, Gralow JR, et al. breast cancer: characterization and method to
Changes in blood flow and metabolism in lo- adjust for time differences. J Nucl Med 2003;44:
cally advanced breast cancer treated with neo- 1044–50.
adjuvant chemotherapy. J Nucl Med 2003;44: [79] Weber WA, Petersen V, Schmidt B, et al. Positron
1806–14. emission tomography in non-small-cell lung
[71] Tseng J, Dunnwald LK, Schubert EK, et al. 18F- cancer: prediction of response to chemotherapy
FDG kinetics in locally advanced breast cancer: by quantitative assessment of glucose use. J Clin
correlation with tumor blood flow and changes Oncol 2003;21:2651–7.

Potrebbero piacerti anche