ArticlePDF AvailableLiterature Review

Endocrine scintigraphy with hybrid SPECT/CT

Authors:
  • Azienda ULSS 5 Polesana

Abstract and Figures

Nuclear medicine imaging of endocrine disorders takes advantage of unique cellular properties of endocrine organs and tissues that can be depicted by targeted radiopharmaceuticals. Detailed functional maps of bio-distributions of radiopharmaceutical uptake can be displayed in 3D-tomographic formats, using single photon emission computed tomography (SPECT), that can now be directly combined with simultaneously acquired cross-sectional anatomic maps derived from computed tomography (CT). The integration of function depicted by scintigraphy and anatomy with CT has synergistically improved the efficacy of nuclear medicine imaging across a broad spectrum of clinical applications, that include some of the oldest imaging studies of endocrine dysfunction.
Content may be subject to copyright.
Endocrine Scintigraphy with Hybrid SPECT/CT
Ka Kit Wong, Lorraine M. Fig, Ehab Youssef, Alice Ferretti, Domenico Rubello,
and Milton D. Gross
Nuclear Medicine/Radiology Department (K.K.W., E.Y., M.D.G.), University of Michigan Hospital, Ann Arbor, Michigan
48109; Nuclear Medicine Service (K.K.W., L.M.F., M.D.G.), Department of Veterans Affairs Health System, Ann Arbor,
Michigan 48105; and Department of Nuclear Medicine (A.F., D.R.), Radiology, Medical Physics, Santa Maria della
Misericordia Hospital, 45100 Rovigo, Italy
Nuclear medicine imaging of endocrine disorders takes advantage of unique cellular properties of endocrine
organs and tissues that can be depicted by targeted radiopharmaceuticals. Detailed functional maps of bio-
distributions of radiopharmaceutical uptake can be displayed in three-dimensional tomographic formats, using
single photon emission computed tomography (CT) that can now be directly combined with simultaneously
acquired cross-sectional anatomic maps derived from CT. The integration of function depicted by scintigraphy
and anatomy with CT has synergistically improved the efficacy of nuclear medicine imaging across a broad
spectrum of clinical applications, which include some of the oldest imaging studies of endocrine dysfunction.
(Endocrine Reviews 35: 717–746, 2014)
I.
Introduction
II. Hybrid SPECT/CT Technology
A. History of SPECT and SPECT/CT systems
B. Technological characteristics of latest generation
SPECT/CT hybrid scanners
C. Quantitative SPECT/CT for radiometabolic
dosimetry
D. Patient radiation dose exposure
E. Radioisotopes commonly used for investigation of
endocrine dysfunction
III. Radioiodine SPECT/CT Imaging of Thyroid Cancer
A.
123
I/
131
I thyroid cancer scans
B. Staging, management, prognosis, and tumor
dosimetry
IV. Somatostatin Receptor Scintigraphy With SPECT/CT
for Staging of Neuroendocrine Tumors
A.
111
In-DTPA-octreotide imaging of neuroendo-
crine tumors
B.
111
In- and
99m
Tc- labeled radionuclide imaging of
medullary thyroid cancer
V. Adrenal Cortical Imaging With
131
I-NP59 SPECT/CT
VI. Sympathetic Adrenomedullary Imaging With
SPECT/CT
A.
123
I/
131
I-MIBG imaging of pheochromocytoma
and paraganglioma
B.
123
I/
131
I-MIBG imaging of neuroblastoma
VII. Parathyroid and Thyroid Scintigraphy With
SPECT/CT
A.
99m
Tc-sestamibi parathyroid scintigraphy
B.
99m
Tc-pertechnetate thyroid scans
C. Miscellaneous endocrine applications of
SPECT/CT
VIII. Current Clinical Role of SPECT/CT in the Diagnostic
Workup of Endocrine Disorders
A. Protocols for SPECT/CT use
B. Economic evaluation and cost-effectiveness
C. Guidelines for indications and role
D. Strategies for reducing radiation exposure
IX. Summary and Future Directions
I. Introduction
The histories of nuclear medicine and endocrinology
have long been linked by radionuclide assessment of
thyroid gland function using iodine-131 (
131
I), a radio-
isotope of stable iodine and one of the earliest examples of
a molecular probe, initially believed to reflect thyroid io-
dine trapping and organification and subsequently found
to be mediated by Na
/I
symporter (NIS) expression (1–
3). Radioiodine was first used to treat hyperthyroidism in
the early 1940s (4), with commercial production and dis-
tribution of
131
I for medical uses by the US Atomic Energy
ISSN Print 0163-769X ISSN Online 1945-7189
Printed in U.S.A.
Copyright © 2014 by the Endocrine Society
Received March 18, 2013. Accepted June 16, 2014.
First Published Online June 30, 2014
Abbreviations: CT, computed tomography; 3D, three-dimensional; 4D, four-dimensional;
DMSA, dimercaptosuccinic acid; DTPA, diethylene triamine penta-acetic acid; EDDA, eth-
ylenediamine diacetic acid; FDG,
18
F-fluorodeoxyglucose;
18
F-DOPA,
18
F-dihydroxyphe-
nylalanine; HYNIC, 6-hydrazinopyridine-3-carboxylic acid;
123
I, radioiodine-123;
131
I, ra-
dioiodine-131;
131
I-NP59,
131
I-6-
-iodomethyl-19-norcholesterol;
111
In, indium-111;
MEN, multiple endocrine neoplasia; MIBG, metaiodobenzylguanidine; MRI, magnetic res-
onance imaging; MTC, medullary thyroid cancer; NET, neuroendocrine tumor; NIS, sodi-
um-iodide symporter; PET, positron emission tomography; PGL, paraganglioma; PHEO,
pheochromocytoma; SPECT, single photon emission CT; SRS, somatostatin receptor scin-
tigraphy; SSTR, somatostatin receptor;
99m
Tc, technetium-99m; TOC, Tyr3-octreotide;
WDTC, well-differentiated thyroid cancer.
REVIEW
doi: 10.1210/er.2013-1030 Endocrine Reviews, October 2014, 35(5):717–746 edrv.endojournals.org 717
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
Laboratory at Oak Ridge, Tennessee, beginning in 1946
(5). From those early beginnings to the present,
131
I con-
tinues as an integral part of the management of well-dif-
ferentiated thyroid cancer (WDTC), used for radioiodine
scintigraphy to detect thyroid cancer metastases and as a
radiotherapeutic agent with
-particle emission for thy-
roid remnant radioablation and treatment of WDTC me-
tastases (6).
In much the same manner as the clinical deployment of
131
I, development of radiopharmaceuticals that target
unique cellular properties of normal and neoplastic endo-
crine tissues has greatly expanded the clinical applications
of radionuclide scintigraphy for the study of endocrine
disorders. Somatostatin receptor scintigraphy (SRS) and
metaiodobenzylguanidine (MIBG) imaging are two prime
examples of successful radiotracers in current clinical use
for staging of neuroendocrine tumors (NETs) and deter-
mining the likely efficacy of long-acting somatostatin an-
alog and radionuclide-based therapy options. Although
providing unique information regarding endocrine phys-
iology and dysfunction, scintigraphy inherently suffers
from low spatial resolution and a paucity of anatomic
information, rendering diagnostic interpretation depen-
dent on pattern-recognition of radiopharmaceutical bio-
distributions. Hybrid single photon emission computed
tomography (SPECT)/computed tomography (CT) imag-
ing devices, capable of combining simultaneously ac-
quired functional maps with cross-sectional anatomic CT
data, have created a powerful new approach to diagnostic
imaging, improving the efficacy of nuclear medicine stud-
ies of endocrine disorders (7–10).
II. Hybrid SPECT/CT Technology
A. History of SPECT and SPECT/CT systems
1. Photon detection and imaging
The radioisotopes used in conventional nuclear medi-
cine emit monoenergetic photons, ie, quanta with a spe-
cific and known spectrum of energy. Many types of de-
tectors are available to identify ionizing radiation, but for
imaging
-photons, scintillation crystals coupled with an
appropriate focusing collimator are used for their high
count radiation detection efficiency. Commercial scintil-
lation cameras (hence the name “scintigraphy”) intro-
duced for
-imaging are based on the original design pro-
posed by Anger in 1958 where incident radiation emitted
by a radiopharmaceutical is converted into light energy,
collected through light guides, and converted into an elec-
tric signal by a matrix of photo-multiplier tubes (11, 12).
In the energy range typical of nuclear medicine radiophar-
maceuticals (71–365 keV; Table 1), the primary detector
material used is a monocrystal of sodium iodide activated
with thallium, NaI(Tl). This fundamental approach al-
lows accurate measurement and reconstruction of the spa-
tial distribution of
-photons. An important aspect affect-
ing image quality is selection of the unique direction of
incident
-photons, achieved through the use of a colli-
mator placed directly in front of the crystal, which serves
to decrease scattered photons. Collimator hole diameter
and thickness of the intervening septae affect count effi-
ciency and spatial resolution. These collimator character-
istics are used to advantage to select between high-reso-
lution vs high-sensitivity or more general all-purpose
collimation, depending upon the characteristic energy
spectrum of the isotope employed and the imaging pro-
cedure to be performed.
2. Single photon emission CT
In 1963, Kuhl and Edwards (13) introduced the first
tomographic device based on a series of scans obtained at
different angles relative to the organ/tissue of interest with
collimated scintillation detectors that allowed acquisition
of transaxial brain “scintiscans” (14). However, it wasn’t
until the 1980s that a concentrated effort was made to
develop rotating SPECT systems for clinical applications.
During SPECT acquisition, the
-camera rotates around
the patient, with multiple two-dimensional projections
(views) acquired at equally distributed angles, covering
arcs of 180° or 360°. These projections are used for three-
dimensional (3D) reconstruction of the image volume and,
like other tomographic modalities, are presented as cross-
sectional “slices” reconstructed by computer-based ana-
lytical or iterative algorithms (15). Filtered back-projec-
Table 1. The Main Isotopes and Corresponding Collimators Used for Planar and SPECT Scintigraphy
Radionuclide T
1/2
E
, keV (% Emissions) Collimator Type
99m
Tc 6.0 h 140.5 (89) Low energy
123
I13.2 h 159.0 (83) Low energy
201
Tl 3.0 d 70.8 (46) Low energy
111
In 2.8 d 171.3 (90), 245.4 (94) Medium energy
67
Ga 3.26 d 93.3 (39), 184.6 (21), 300.2 (17) Medium energy
131
I8.0 d 364.5 (82) High energy
Abbreviation: T
1/2
, half-life.
718 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
tion, an analytical method initially applied for SPECT, is
a mathematical process that uses Fourier transforms to
superimpose and normalize imaging projections, generat-
ing an estimate of the radioactivity contained within each
volume element (voxel) (15). Conversely, the statistical
image reconstruction algorithms, so-called iterative meth-
ods, create reconstructed images by successive estimates
and comparisons with the measured data. The result of
each comparison is used to modify the current estimate in
repeating iterations until the difference between estimated
and measured data is small. Despite their early devel-
opment in the 1970s, iterative reconstructive methods
had to wait for computer technology to “catch up” de-
cades later (13, 16). Iterative methods produce images
of higher quality than filtered back-projection, with the
ability to perform attenuation, scatter, and partial vol-
ume corrections (15).
3. First hybrid SPECT/CT systems
One of the main sources of image degradation with
SPECT is attenuation of photons by overlying body tis-
sues. The amount of attenuation depends on path length
(distance to the detector), the types of tissue, and emitted
photon energy. An “attenuation map” is necessary to cor-
rect for proportionally decreased detected radiation from
targets located deeper within the body. One of the first
methods developed for attenuation correction was the
Chang method, a geometric model applied primarily to
brain scans (17). Subsequently, transmission source-based
attenuation correction was introduced in the mid-1980s,
where emission-transmission scanning was performed us-
ing an external radiation source for acquiring transmis-
sion images (18). More recently, high-resolution struc-
tural imaging with CT in conjunction with functional
emission tomographic data was proposed by Lang et al
(19), who developed the first prototype of a dual-modality
SPECT/CT system. The first commercial system with hy-
brid SPECT/CT capability combined a dual-detector
-camera with an independent low-dose x-ray tube, co-
positioned in the same scanner gantry in an “in-line” con-
figuration. Although this device was not a diagnostic-
quality CT scanner, its usefulness in improving SPECT
interpretation was reported in several studies (20, 21).
Early steps in the development of commercial SPECT/CT
scanners required accurate conversion of x-ray attenua-
tion maps to the
-photon energy of the SPECT study (22,
23) and to correct coregistration of SPECT and CT images.
Hybrid scanners acquire SPECT and CT data sequentially
with the patient in an optimally stationary, fixed position.
Once the appropriate calibrations for SPECT and CT
alignment are executed, the two studies are acquired
coregistered and can be easily superimposed (fused).
B. Technological characteristics of latest generation
SPECT/CT hybrid scanners
1. Technological innovations
Recent dual-modality SPECT/CT systems include inte-
grated dual-head SPECT and multislice CT. Multislice CT
scanners adopt third-generation slip-ring geometry (tube
and detector bank linked and rotating together) along
with the multislice technology, consisting of multiple, par-
allel arcs of detectors that collect data from more slices at
the same time (up to 64 slices or rows in the latest gener-
ation SPECT/CT systems) (24).
2. Reconstruction algorithm innovations
The current iterative reconstruction algorithms use CT
information and system-response models to perform
state-of-the-art quantification methods (25) (Figure 1). In-
novations include: 1) CT-based photon attenuation cor-
rection—CT data in Hounsfield units from the transmis-
sion scan is converted to a map of attenuation coefficients
matching the effective energy and the resolution of the
emission SPECT (22); 2) scatter correction—scattered
photons, which have lost their original direction and also
a part of their energy, are detected using dual, triple, or
multiple energy windows (26, 27) to estimate the amount
of scattered photons in the image and subtract this value
from the reconstructed images (28, 29); 3) partial volume
effect correction—compensates for an apparent loss of
counts in small structures using either recovery coefficient
methods (25, 30) or methods incorporating anatomic cor-
rection information (30–32); and 4) motion correction—
patient motion occurring between emission (SPECT) and
transmission (CT) scans leads to incorrect superimposi-
tion of the attenuation map and SPECT datasets. For in-
voluntary motion (breathing or heartbeat), physiological
gating of image acquisition can reduce these artifacts (33).
Nonphysiological patient motion can be corrected by a
postacquisition manual shifting of images technique, with
cross-correlation methods (34), or with correction meth-
ods implemented within the iterative reconstruction algo-
rithm (35–37).
C. Quantitative SPECT/CT for radiometabolic dosimetry
The ability to estimate internal dosimetry is often useful
in planning and managing radionuclide therapy. Conju-
gated views with planar imaging or stand-alone SPECT
acquisition allow rough dosimetric approximation, with
variability ranging from 30 to 100% (7, 38). Hybrid
SPECT/CT systems provide a more accurate approach to
dosimetry because CT data can be used to create patient-
specific, anatomically corrected, 3D target organ/tissue
attenuation and density maps. SPECT/CT-based 3D do-
simetry can thus be used to calculate radiation dose to both
doi: 10.1210/er.2013-1030 edrv.endojournals.org 719
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
target and nontarget tissues with the therapeutic goals of
maximizing radiation dose to an endocrine malignancy
and, at the same time, limiting radiation exposure to other
sensitive tissues (eg, lung, kidney, bone marrow) (38).
D. Patient radiation dose exposure
The effective dose attributable to SPECT acquisition
from the radiopharmaceutical biodistribution ranges be-
tween 3 and 12 mSv, depending on the radiopharmaceu-
tical type and activity used (39, 40). The effective dose
from typical multislice CT scans performed during dual-
modality SPECT/CT examinations calculated for low-
dose protocols (20 –40 mA, 120 –130 kV) is an additional
2- to 4-mSv exposure from the CT scan with 40-cm length
(ie, single SPECT/CT bed position) (7). In cases of
SPECT/CT multiple bed positions, the effective dose val-
ues for each bed should be added together. The CT
component of SPECT/CT is typically “nondiagnostic,”
performed for anatomic correlation and attenuation cor-
rection, with lower tube energies and without iv contrast
enhancement, and as such, it does not replace conven-
tional diagnostic contrast-enhanced CT protocols.
E. Radioisotopes commonly
used for investigation of
endocrine dysfunction
As discussed earlier,
131
I has a
364-keV
-emission and half-life (8
d) suitable for thyroid cancer imag-
ing, and its
-particle emission al-
lows radionuclide therapy of thyroid
cancer metastases. Another radioio-
dine isotope, iodine-123 (
123
I), can
be used for diagnostic imaging.
123
I
has the advantage of a lower 159-
keV
-emission better suited for
modern
-cameras, providing higher
quality imaging. Also, because
123
I
lacks particulate emissions, it has not
been implicated in the so-called
“stunning” of thyroid tissues re-
ported after diagnostic doses of
131
I
(41). Drawbacks to the use of
123
I are
the higher cost for the isotope and
the shorter half-life (13 h), preclud-
ing multiday imaging and leading to
greater complexity of dosimetry
calculations.
SRS is a whole-body imaging
technique that depicts biodistribu-
tions of radiolabeled somatostatin
analogs (43). Somatostatin is a reg-
ulatory peptide with affinity for G
protein-coupled membrane-bound somatostatin receptor
(SSTR) subtypes 1–5, including 2A and 2B, which are
overexpressed in NETs (42–44). Peptide-based imaging
with somatostatin analogs has advantages of high speci-
ficity, good tissue penetration, rapid clearance, and low
antigenicity (42). The most common radiopharmaceutical
used for SRS is indium-111 (
111
In)-diethylene triamine
penta-acetic acid (DTPA)-octreotide (
111
In-pentetreotide)
with affinity for SSTR2 and SSTR5. Normal physiological
distribution occurs in liver, spleen, kidneys and bladder,
gallbladder, and bowel. Mild uptake is seen in the pitu-
itary, thyroid, adrenal glands, and head of the pancreas.
SRS provides evidence for targeting of tumor tissue be-
cause octreotide uptake in NETs can be used to predict
therapeutic success with
90
Y- or
177
Lu-labeled somatosta-
tin analogs (45).
123
I- and
131
I-labeled MIBG is a “false neurotransmit-
ter” analog of norepinephrine, used to image NETs.
MIBG accumulation shares norepinephrine transporter
uptake with affinity for vesicular monoamine transporter
and incorporation into neurosecretory vesicles (44, 46,
47). Physiological MIBG uptake is seen in the salivary
Figure 1.
Figure 1. Illustration of the image formation chain. During SPECT reconstruction, corrections are
made for the four main confounding effects, producing a 3D measured count distribution. With
a calibration step, this count distribution can also be translated into an activity distribution (25).
720 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
glands, thyroid (unless blocked with potassium iodide),
heart, liver, spleen, kidneys, and bladder, with variable
uptake in the adrenal medulla and in brown fat. Thera-
peutic doses of
131
I-MIBG may be employed for the treat-
ment of metastatic NETs (48).
Technetium-99m (
99m
Tc)-sestamibi, a lipophilic cation
of the isonitrile family, is accumulated and retained in the
mitochondria of oxyphil-rich parathyroid adenomas.
99m
Tc-sestamibi has been used to perform parathyroid
scintigraphy in conjunction with
99m
Tc-pertechnetate or
123
I for thyroid tissue subtraction (49). In 1992, Taillefer
introduced a dual-phase protocol with
99m
Tc-sestamibi
alone to exploit its differential kinetic properties in thyroid
and parathyroid tissues (50). Parathyroid adenomas dis-
play focal radiotracer retention and delayed washout com-
pared to normal thyroid tissues. This dual-phase single
isotope technique (early 10- to 20-min and delayed 2-h
time-points) has similar diagnostic accuracy to dual-iso-
tope subtraction methods but is simpler to perform. Table
2 lists the commonly used radiopharmaceuticals in endo-
crine scintigraphy.
III. Radioiodine SPECT/CT Imaging of
Thyroid Cancer
Thyroid cancer is the most common endocrine malignancy
in adults, with 60 220 (14 910 males, 45 310 females)
newly diagnosed cases reported in the United States in
2013 (51). The incidence of WDTC has continued to in-
crease over the last three decades, in part due to earlier
detection of so-called papillary thyroid “microcarcino-
mas” (size 1.0 cm), although other factors (eg, dietary,
environmental) may also be involved (52, 53). The prog-
nosis for most patients with WDTC is excellent, with a
10-year survival of approximately 93% for papillary thy-
roid cancer and approximately 85% for follicular thyroid
cancer. However, mortality increases to 25–45% at 10
years in higher-risk patients (TNM stages III/IV), and re-
currences may occur in one-third of patients (54–56). Tu-
mor staging using systems such as the American Joint
Committee on Cancer (AJCC) TNM seventh edition or
MACIS (Metastasis, Age at presentation, Completeness of
surgical resection, Invasion [extrathyroidal], Size) pro-
vides important prognostic information. Contemporary
guidelines for diagnosis and management of WDTC em-
phasize schema for risk stratification based on risk of re-
currence, rather than older staging systems designed to
predict survival (6, 57).
After diagnosis, total or near-total thyroidectomy, and
in some cases cervical lymph node resection, is performed
to remove the primary tumor and establish histopathol-
ogy. Furthermore, removal of the normal thyroid tissues,
which accumulate radioiodine more avidly than tumor,
facilitates subsequent radioiodine imaging and therapy.
For patients with locoregional disease or distant metasta-
ses, treatment with
131
I has been shown to improve out-
comes (54, 55). Previously, radioiodine ablation of rem-
nant thyroid tissue was performed in most cases to
facilitate clinical surveillance; however, because statistical
evidence indicates that
131
I neither decreases recurrence
nor increases survival in low-risk patients (stage I disease),
radioiodine ablation is no longer routinely performed in
Table 2. Commonly Used Radiopharmaceuticals for Endocrine Scintigraphy
Radiopharmaceutical Molecular Target/Mechanism of Uptake
131
I (radioiodine isotope) Affinity for the NIS expressed by differentiated thyroid cancer,
-particle emission allows
radionuclide therapy
123
I (radioiodine isotope) Affinity for the NIS expressed by differentiated thyroid cancer
111
In-DTPA-octreotide
(
111
In-pentetreotide)
Somatostatin analog with affinity for somatostatin receptors expressed by endocrine
neoplasms
111
In-DOTA-lanreotide Somatostatin analog with affinity for somatostatin receptors expressed by endocrine
neoplasms
99m
Tc-EDDA/HYNIC-TOC Somatostatin analog with affinity for somatostatin receptors expressed by endocrine
neoplasms
123
I-MIBG Catecholamine analog with uptake by the norepinephrine transporter expressed by neural
crest tumors
131
I-MIBG Catecholamine analog with uptake by the norepinephrine transporter expressed by neural
crest tumors,
-particle emission allows radionuclide therapy
131
I-NP59 Cholesterol analog that enters the cortisol synthesis pathway of adrenocortical tumors
99m
Tc-sestamibi (methoxyisobutylisonitrile) Uptake in benign and malignant tumor cells dependent on cell membrane and
mitochondrial potentials
99m
Tc-tetrofosmin (lipophilic diphosphine) Uptake in benign and malignant tumor cells dependent on cell membrane and
mitochondrial potentials
99m
Tc-pertechnetate (
99m
Tc-0
4
)Trapping in the thyroid gland via the NIS and Na
/Cl
channels
99m
Tc(V)-DMSA Uptake in benign and malignant tumors dependent on metal complex equilibrium and pH
Abbreviation: DOTA, 1,4,7,10–tetra-azacyclododecane–1,4,7,10–tetra-acetic acid.
doi: 10.1210/er.2013-1030 edrv.endojournals.org 721
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
this group (54 –56, 58– 60). Risk stratification and careful
patient selection are key to this approach.
A.
123
I/
131
I thyroid cancer scans
131
I imaging has poor spatial resolution, and collimator
septal penetration of the high-energy 364 keV
-emissions
produces artifacts that degrade image quality. The paucity
of anatomical information provided by planar or SPECT
imaging often leads to difficulties in distinguishing benign
from malignant etiologies (61–63). Diagnostic CT has
played a limited role in initial WDTC workup due to the
need to avoid iodinated contrast when contemplating ra-
dioiodine treatment and the high frequency of nodal me-
tastases in otherwise anatomically normal neck lymph
nodes. Coregistration of tomographic functional and an-
atomic information with hybrid SPECT/CT has been the
subject of several reviews (6469) which conclude that
radioiodine
131
I/
123
I SPECT/CT is a powerful diagnostic
tool that overcomes many of the limitations of planar im-
aging (70–99) (Table 3).
1. Strategies for SPECT/CT
131
I SPECT/CT scans have used diagnostic doses be-
tween 37 and 430 MBq (1–11 mCi) (89, 90, 94–96, 100)
and postradioiodine therapy doses of 1.1 to 9.7GBq (30
to 260mCi) (70, 74, 75, 77– 80, 84– 88, 90–92, 97, 99)
with patients prepared under endogenous hypothyroid-
ism or exogenous recombinant human TSH stimulation.
Investigators have found radioiodine SPECT/CT useful in
the immediate postsurgical setting, either with preablation
diagnostic (94, 95, 100) or post-therapy scanning (70, 74,
75, 77–81, 83, 85, 86, 91, 92, 98, 99), and also for long-
term surveillance (72, 73, 77, 89, 90).
131
I SPECT/CT was
first reported in 2001 by Even-Sapir et al (20) in a sub-
group of four patients with thyroid cancer studied with
SPECT/CT. In a larger group of 71 patients, the addition
of anatomical information from SPECT/CT allowed bet-
ter characterization of neck uptake and more accurate dif-
ferentiation of foci outside the neck as benign vs metastatic
disease. Incremental diagnostic value of SPECT/CT over
Table 3. Utility of
131
I/
123
I SPECT/CT for Diagnostic and Post-Therapy Thyroid Cancer Scans
Advantages of SPECT/CT (Refs) Comments and Findings
Localization and characterization of radioactivity
foci (74, 77, 78, 80, 84, 92, 95, 97)
SPECT/CT depicts benign vs malignant uptake in the neck (ie, thyroid
remnant vs cervical nodal disease) with incremental diagnostic value
compared to planar imaging in 21–64% of patients.
Clarification of equivocal findings (70, 72, 77,
78, 99)
SPECT/CT clarified findings otherwise equivocal (ie, inconclusive,
indeterminate, unclear) on planar imaging, as benign uptake or
metastatic disease in 15–30% of patients.
Confirmation of cryptic findings (73, 120, 121) SPECT/CT is able to confirm cryptic findings, physiological variants, and
benign mimics of disease in the neck, thorax, abdomen, and pelvis.
Evaluation of distant metastatic disease (70, 72,
75, 77–79, 84, 89, 91, 92, 95, 97, 99)
SPECT/CT is useful for evaluating distant sites of disease and it may obviate
the need for additional diagnostic CT when pulmonary and bone
metastases are confirmed. Incremental diagnostic value of SPECT/CT over
planar imaging for distant metastases is reported in 40–100% of
patients.
Identification of non-iodine-avid disease
(72, 79, 99)
SPECT/CT provides anatomical information from the CT component and
detects non-iodine-avid disease in between 3 and 21% of patients.
Improved diagnostic performance due to
increased specificity (72, 79)
Diagnostic
123
I SPECT/CT has sensitivity of 50% and specificity of 100%,
compared to planar scans with sensitivity of 41% and specificity of 68%.
Diagnostic 131I SPECT/CT has sensitivity of 62% and specificity of 98%,
compared to planar scans with sensitivity of 62% and specificity of 78%.
Improved interobserver agreement and reader
confidence (91, 95)
Interobserver disagreement for neck foci occurred in 24 of 42 (57%)
patients on planar imaging, decreasing to 9 of 42 (21%) on SPECT/CT.
SPECT/CT increases reader confidence.
Staging (TNM) and risk stratification (10, 77, 78,
86, 99, 100)
Preablation diagnostic
131
I SPECT/CT detected regional metastases (35%)
and distant metastases (8%); changing TNM stage in 4% of younger and
25% of older patients. Post-therapy
131
I SPECT/CT changed stage in 6.1–
26.4% patients
Change in management (74, 77, 78, 84, 89, 90,
92, 94)
SPECT/CT led to change in management in between 2 and 58% of patients;
SPECT/CT can affect decisions regarding surgical referral, to treat or not
treat with
131
I, selection of
131
I dose, external-beam radiation therapy
referral, ordering FDG PET.
Prognosis (75, 85) SPECT/CT: sensitivity of 78%, specificity of 100% for predicting disease-free
status at 29 mo median. Lymph nodes with size 0.9 mL on SPECT/CT
respond to
131
I treatment.
Tumor dosimetry (139) Feasible when considering radioiodine treatment of macroscopic iodine-avid
disease.
722 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
planar imaging was reported in 41 of 71 (57%)
patients (90).
2. Diagnostic interpretation
Common to all published studies to date is the ability of
SPECT/CT to provide more precise anatomic localization
of radioactivity, aiding the characterization of radioiodine
uptake as benign (in remnant thyroid tissues or due to
physiological biodistribution) or malignant (in cervical
lymph nodes or distant metastases) (Figure 2). A major
strength of
131
I SPECT/CT is to substantially reduce equiv-
ocal scan findings frequently encountered on planar im-
aging. In 147 patients, post-therapy
131
I SPECT/CT clar-
ified all 25 equivocal interpretations in the neck as either
regional lymph nodes or remnant thyroid tissues. For dis-
tant foci, SPECT/CT removed uncertainty on planar im-
aging in 21 of 52 (40%) foci of
131
I uptake by distinguish-
ing metastases from benign physiological “mimics” of
disease (78). Similar findings have been reported with
preablation diagnostic
131
I SPECT/CT (95, 96).
Diagnostic radioiodine planar imaging for surveillance
of thyroid cancer has a sensitivity that varies between 40
and 75% (in part, dependent upon the
131
I dose admin-
istered) and high specificity of 90–100% (61, 89, 90).
SPECT/CT using
123
I was found to have superior accu-
racy, with sensitivity and specificity of 50% and 100%,
respectively, compared to SPECT (45% and 89%) and
planar imaging (41% and 61%) (72). Another study of
diagnostic
131
I SPECT/CT in 123 pa-
tients found that sensitivity of both
planar imaging and SPECT/CT for
the detection of iodine-avid disease
was 62%, although SPECT/CT had
significantly higher specificity than
the planar imaging (94 vs 74%),
which is the major strength of its
use (79).
Non-iodine-avid disease due to
the lack or subsequent loss of NIS
expression occurs in 20–30% of
WDTC at the time of diagnosis and is
seen more frequently with Hürthle
cell cancer, with papillary subtypes
with unfavorable features (eg, tall
cell, columnar, cribiform), and
with poorly differentiated (insular)
cancer (101). Radioiodine
131
I
SPECT/CT can localize additional
non-iodine-avid disease on the CT
component in as many as 32 of 148
(21.6%) patients (99). Detection of
non-iodine-avid disease signifi-
cantly impacts clinical manage-
ment by directing treatment to additional surgical in-
tervention or external beam radiation therapy and also
identifies the need for alternative imaging strategies
such as
18
F-fluorodeoxyglucose (FDG) positron emis-
sion tomography (PET).
3. SPECT/CT identification of metastatic disease
Interpretation of
131
I uptake in the central neck can
be challenging, depending on patients’ body habitus.
SPECT/CT accurately identifies cervical nodal metastases,
often in lymph nodes of normal size. SPECT/CT has de-
picted rare parapharyngeal metastases (occurring in 2.5%
of 561 patients) that, owing to their location, may not be
apparent with preoperative neck ultrasound and thus are
not resected during neck compartment dissection (82).
WDTC has a predilection to metastasize distantly to
lung and bone and more rarely to soft tissues: liver, brain,
skin, and other sites. Pulmonary metastases on planar
scintigraphy are characterized as either micronodular or
macronodular disease, and miliary lung metastases may
escape detection on chest x-ray or even diagnostic chest
CT.
131
I SPECT/CT is useful for evaluation of distant met-
astatic disease (72, 74, 77–79, 89, 92, 94, 95), and after
confirmation of pulmonary or bone metastases with
SPECT/CT, further diagnostic CT may be omitted (99). In
the thorax, SPECT/CT can precisely localize malignancy
to lung, bone (ribs, sternum, or spine), or mediastinal
Figure 2.
Figure 2. Diagnostic preablation 37 MBq (1 mCi)
131
I scan in a 23-year-old female status after
total thyroidectomy for multifocal papillary thyroid carcinoma. A and B, Planar anterior and
posterior images at 24 hours demonstrate that several foci increased radiotracer activity in the
neck, including right superior neck (arrow) and multiple foci in the thorax (arrowheads). C–F,
Axial CT and SPECT/CT fusion images confirm that the activity in the neck represents a cervical
lymph node metastasis (arrows), and the activity in the thorax represents bilateral micronodular
lung metastases (arrowheads).
doi: 10.1210/er.2013-1030 edrv.endojournals.org 723
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
lymph nodes (Figure 3). A spectrum of rare metastatic sites
depicted on
131
I SPECT/CT has been described (87, 88),
with reports of metastatic disease in unanticipated tumor
locations; eg, liver, kidney, muscle, and trachea (93,
102–110).
4. SPECT/CT evaluation of benign uptake
Radioiodine scintigraphy has numerous physiological
variants and potential disease “mimics” that must be rec-
ognized for accurate image interpretation (111–113). The
NIS is an intrinsic thyrocyte-based plasma membrane gly-
coprotein that allows an increase of cytoplasmic iodine
concentration 20 to 40 times above plasma levels (1, 114,
115). NIS is expressed in thyroid, salivary glands, gastric
mucosa, lactating mammary glands (114), and nonlactat-
ing breasts in young women (116, 117) and has also been
detected in the lacrimal glands, choroid plexus, ciliary
body of the eye, skin, placenta, and thymus (1, 114–116,
118, 119). Knowledge of these biodistributions will pre-
vent false-positive interpretation.
131
I SPECT/CT was used for evaluation of a wide range
of cryptic findings occurring in 40 of 184 (22%) scans,
including a spectrum of physiological benign etiologies
(73). Although these variants are usually recognized on
planar imaging, SPECT/CT resolves diagnostic uncer-
tainty with greater confidence and improved accuracy
(120, 121). Numerous case reports have demonstrated the
utility of SPECT/CT for confirmation of benign “mimics”
of disease; eg, blocked nasolacrimal
duct, lingual thyroid, thymus, struma
ovarii, ovarian teratoma, menstruat-
ing uterus, and radioiodine accumula-
tion in simple renal cysts (93, 102–
110, 122–126).
5. Comparison of
131
I SPECT/CT to
other imaging modalities
The established role of FDG PET
in WDTC is to search for occult me-
tastases in patients with elevated thy-
roglobulin levels suggesting persis-
tent or recurrent disease, despite
nonlocalizing
131
I scintigraphy.
FDG PET has particular value when
thyroglobulin levels are high and/or
when distant metastases are sus-
pected (63, 127–131).
131
I SPECT/
CT, FDG PET, and
99m
Tc-methylene
diphosphonate bone scans were
compared in patients with suspected
thyroid cancer bone metastases.
131
I
SPECT/CT had sensitivity of 92%
and specificity of 98%, which was
similar to FDG PET sensitivity of 85% and specificity of
88%, with both modalities superior to
99m
Tc-methylene
diphosphonate bone scintigraphy (83). In another study,
131
I SPECT/CT and FDG PET had similar diagnostic per-
formance, although
131
I SPECT/CT was observed to out-
perform FDG PET when there was a history of a single
131
I
therapy, whereas FDG PET had better sensitivity in pa-
tients after multiple radioiodine treatments (81).
B. Staging, management, prognosis, and
tumor dosimetry
1. SPECT/CT for staging of WDTC
Accurate staging and risk stratification are important
for the management of thyroid cancer to guide therapeutic
decisions and intensity of surveillance. Post-therapy
131
I
SPECT/CT imaging shows improved characterization of
nodal (N) and distant metastatic (M) status for TNM stag-
ing, compared to planar imaging (70, 75, 80, 86, 90, 92,
99). At the time of the first radioablation, SPECT/CT led
to reclassification of the cervical nodal (N) status in 20 of
57 (35%) patients (86). In another study, SPECT/CT
changed the N score in 12 of 33 (36.4%) patients and the
M score in four of 19 (21.4%) directly, leading to man-
agement changes in 10 (24.4%) patients (77).
Preablation diagnostic SPECT/CT changed TNM stag-
ing in 10 of 48 (21%) patients by identification of regional
and distant metastases before the first radioiodine abla-
Figure 3.
Figure 3. False-positive uptake depicted on preablation diagnostic 37 MBq (1 mCi)
131
I scan in a
57-year-old female status after total thyroidectomy for resection of invasive, multifocal follicular
thyroid carcinoma. A and B, Planar anterior and posterior images demonstrate focal uptake in
the central neck (arrowhead) suspicious for residual disease, and in the right thorax (arrows)
suspicious for metastasis. C–F, Axial CT and SPECT/CT fusion images localize the activity to the
patient’s tracheostomy (arrows), due to retained secretions (arrows) and the thoracic uptake to
osseous metastasis of the right sixth rib (arrows).
724 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
tion, thereby allowing patient-specific selection of
131
Iac
-
tivities; eg, lower activities, 1.1 GBq (30 mCi) for remnant
ablation in low-risk patients; and higher activities, 3.7 to
7.7GBq (100 to 200mCi) for treatment of thyroid
cancer (94). In 320 patients, the impact of preablation
diagnostic
131
I SPECT/CT on TNM stage was significant,
resulting in a change in staging in 4% of younger patients
(45 y) and 25% of older patients (45 y) (100). In
younger patients, distant metastases were detected in five
of 138 (4%) cases and nodal metastases in 61 of 138
(44%) cases. In the older group, distant metastases were
detected in 18 of 182 (10%) patients and nodal metastases
in 51 of 182 (28%).
Information obtained from SPECT/CT at the first ra-
dioablation has extended our knowledge regarding the
incidence of nodal metastases in T1 tumors. Using a com-
bination of pN1 (surgical neck dissection) and sN1
(SPECT/CT) in a bicentric study of 151 patients, lymph
node metastases occurred in 26% of T1 (2.0 cm) tumors
and 22% of microcarcinomas (1.0 cm) (80). In another
study in which SPECT/CT contributed to N staging, T1a
tumors occurred in 49 of 320 (15%) patients and were
associated with nodal metastases in 28 of 49 (57%) pa-
tients, and unsuspected distant metastases were detected
in two of 49 (4%) (100).
2. Changes in management and prognostic indicators
Radioiodine SPECT/CT has been reported to change
clinical management in 11–58% of patients (74, 79, 94,
99). Changes include decisions to use or withhold radio-
iodine treatment, guidance on surgical planning, selection
of patients for external beam radiation therapy, and the
use of alternative imaging strategies such as FDG PET.
Actual changes to management strategies depend upon the
specific clinical context and treatment protocols at a given
institution. Avoidance of unnecessary
131
I therapy is im-
portant when residual and/or metastatic disease has been
excluded and the use of empiric
131
I treatment would re-
sult in unnecessary radiation exposure (133, 134). For
example, in 13 children with WDTC, SPECT/CT (
123
Ior
131
I) precisely localized all neck foci of radioiodine, in-
cluding benign etiologies, and supported a decision not to
use therapeutic
131
I in two children (76).
131
I SPECT/CT imaging for follow-up and surveillance
also provides prognostic information regarding the suc-
cess of radioiodine treatment. A neck nodal volume of
0.9 mL as assessed on SPECT/CT was highly likely to
respond to
131
I therapy alone without surgical resection
(85). In a study of 170 patients to identify prognostic fac-
tors predicting treatment failure at 2 years, TNM staging,
macroscopic cervical lymph node disease, and positivity of
planar and SPECT/CT imaging were identified as significant
predictive factors, with only SPECT/CT as an independent
prognostic indicator after multivariate analysis (75).
3. Tumor dosimetry
Diagnostic radioiodine SPECT/CT imaging provides
the opportunity to identify patients with unsuspected me-
tastases, defines the target(s) of
131
I therapy, and permits
adjustment of prescribed therapeutic radioactivity and do-
simetry calculations when high-dose
131
I therapy is con-
sidered (94, 135). The aim of the initial
131
I treatment is to
destroy as much malignant mass as possible, with the tu-
mor radiation-absorbed dose as the best predictor of suc-
cess. Absorbed doses of 7800 cGy have been estimated
as necessary to destroy residual carcinomas (137, 138). An
example of the utility of SPECT/CT to measure volumes of
regional metastases, perform tumor dosimetry, and
thereby determine the optimal radioiodine therapy dose
has been reported (139).
IV. Somatostatin Receptor Scintigraphy
With SPECT/CT for Staging of
Neuroendocrine Tumors
NETs comprise a rare, heterogeneous group of neoplasms
derived from endocrine stem cells of the amine precursor
uptake and decarboxylation system. They have a widely
variable natural history and prognosis (140, 141), with the
potential to cause clinical syndromes due to hypersecre-
tion of biogenic amines and polypeptides (142–144). The
annual incidence of these neoplasms is increasing due to
greater awareness and earlier detection; however, despite
earlier diagnosis and sensitive biochemical markers, met-
astatic disease is often present at diagnosis (44, 141, 142).
Enterochromaffin NETs derived from enteroendocrine
cells within the epithelial lining of the digestive and respi-
ratory tracts and from pancreatic islet cells were previ-
ously classified based on secretory activity as carcinoid
tumors (50% of NETs) with the classical triad of flushing,
hypotension, and diarrhea, or noncarcinoid tumors (eg,
gastrinomas, glucagonomas, insulinomas, and others).
Recent revisions to the classification system propose that
NETs be grouped according to their differentiation, how
closely they resemble their non-neoplastic counterparts,
and grade, a measure of their biological aggressiveness and
malignant potential based upon mitotic rates and Ki-67
index (140, 142). These features, combined with staging
systems such as the AJCC TNM seventh edition, more
appropriately reflect the prognosis of NETs and guide sub-
sequent management and therapy.
111
In-DTPA-octreotide scintigraphy is considered a
first-line imaging modality for staging of NETs, with an
doi: 10.1210/er.2013-1030 edrv.endojournals.org 725
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
overall sensitivity of 80–90% for carcinoid and 50–70%
for pancreatic NET (with especially low sensitivity for in-
sulinoma due to the lack of SSTR2 expression). Specificity
is high at 88–97% (46, 47, 140), although false-positives
occur due to uptake in physiological
structures or due to inflammation or
infection.
A.
111
In-DTPA-octreotide imaging of
neuroendocrine tumors
SPECT/CT for neuroendocrine
imaging was first evaluated in mixed
patient groups undergoing
123
I-
MIBG,
111
In-DTPA-octreotide, and
123/131
I scintigraphy (20, 21, 145,
146). Improved diagnostic accuracy
of SPECT/CT over planar and
SPECT imaging has been consis-
tently reported for SRS, frequently
resulting in changes to management
plans (147–157) (Figure 4 and Table
4). The precise anatomic localization
of radioactivity using SPECT/CT
provides insight into the etiology of
uptake as benign or malignant, re-
ducing the number of equivocal find-
ings encountered on planar imaging
(148, 156, 157). Furthermore, the
CT component of SPECT/CT can
provide valuable additional ana-
tomic information that aids in image
interpretation. In one study, SPECT/CT
more accurately characterized 163
of 169 (96%) tumor foci compared
to only 138 of 169 (81%) foci on
planar and SPECT (152). In another study, SPECT/CT
had incremental diagnostic value compared to planar im-
aging, with superior localization and/or detection of ad-
ditional lesions in 23 of 44 (52%) cases, although no ad-
Figure 4.
Figure 4. Bronchial carcinoid in a 32-year-old man demonstrated with
111
In-pentetreotide SPECT/
CT. A and B, Whole-body anterior and posterior images at 24 hours show focal activity in the left
thorax (arrows). There is otherwise physiological distribution of radioactivity in the remainder of
the body. C–E, Coronal SPECT, CT, and fused SPECT/CT localize thoracic activity to a calcified
mass adjacent to the descending aorta (arrows). Axial CT (not shown) demonstrated an
endobronchial location compatible with endobronchial carcinoid.
Table 4. Utility of SPECT/CT for Somatostatin Receptor and Sympathoadrenomedullary Scintigraphy
Advantages of SPECT/CT (Refs) Comments and Findings
Localization and characterization of radioactivity foci
(95, 149–151, 202, 203)
SPECT/CT has superior localization, characterization and detection rate of
lesions compared to planar imaging in 37–73% of patients.
Clarification of equivocal findings (156) SPECT/CT clarifies findings otherwise equivocal on planar imaging, as
benign uptake or metastatic disease in 27/98 (28%) foci.
Improved diagnostic performance (147, 152, 155–157,
192, 202–206, 208)
99m
Tc/
111
In-labelled octreotide SPECT/CT for staging NETs has sensitivity
92–97% and specificity 88–100% compared to SPECT with sensitivity
of 79–83% and specificity of 64 –100%;
131
I/
123
I-MIBG SPECT/CT for
PHEO/PGL has sensitivity of 87–100% and specificity of 94–100%
compared to SPECT with sensitivity of 90% and specificity of 68%;
111
In-DTPA-octreotide SPECT/CT has higher sensitivity than side-by-side
visual analysis or software fusion of separate SPECT and CT data.
Improved interobserver agreement (148, 155) Agreement
: Planar
0.59, SPECT
0.74, SPECT/CT
0.86.
Change in management (150, 151) SPECT/CT changes management in 13.9–40% of patients.
Attenuation-correction (153) Attenuation-corrected SPECT/CT leads to increased uptake in 30% of
lesions with no change to diagnostic performance.
Additional benefits of SPECT/CT (95, 155, 159, 160) Improved reader confidence, allows single time-point imaging at 24 h, and
can perform patient-specific and tumor-specific dosimetry.
726 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
ditional benefit occurred when planar images were
negative (151).
99m
Tc-ethylenediamine diacetic acid
(EDDA)/6-hydrazinopyridine-3-carboxylic acid (HYNIC)-
Tyr3-octreotide (TOC) SPECT/CT has sensitivity of 92% and
specificity of 88% for NET staging (158).
99m
Tc-EDDA/
HYNIC-octreotide and
111
In-DTPA-octreotide SPECT/CT
have been reported to have higher sensitivity and specificity than
SPECT or diagnostic CT imaging (156, 157). Hybrid
SPECT/CT somatostatin receptor imaging is easier to perform
and more accurate than previous techniques for performing ret-
rospective software fusion or side-by-side analysis of SPECT
and CT (147). Attenuation-corrected images using CT-based
maps increase lesion contrast and/or intensity in the abdomen
(153).
SPECT/CT SRS increases interobserver agreement, as
reported in two studies: SPECT/CT
was reported as 0.86
in both, compared to planar imaging
of 0.56 and 0.59,
respectively (148, 155). Furthermore, SPECT/CT allows
imaging to be compressed into a single time-point at 24
hours, improving the efficiency of scheduling (155).
SPECT/CT has been used to perform individualized do-
simetry in patients receiving
177
Lu-DOTA-octreotate
treatment of metastatic NETs (159). Using whole-body
imaging and blood and urine measurements at 24-, 96-,
and 168-hour time-points, the absorbed dose to the kidney
was calculated with SPECT/CT and this represented the
organ receiving the highest radiation dose for most pa-
tients. This method was also adapted for purposes of cal-
culating doses, with the goal of limiting bone marrow
radiation dose. In a study of 12 patients undergoing
99m
Tc-
EDDA/HYNIC-TOC imaging of NETs, SPECT/CT afforded
the calculation of radiation dosimetry identifying the
highest absorbed radiation doses to liver, spleen, kidneys,
and bladder (160).
B.
111
In- and
99m
Tc- labeled radionuclide imaging of
medullary thyroid cancer
Medullary thyroid cancers (MTCs) are derived from
parafollicular (C cells) of neural crest origin, comprising
3–12% of all thyroid cancers. MTC is often an indolent
disease where, after surgery, up to 40% of patients will
have residual or recurrent neoplasm identified by elevated
serum calcitonin or carcinoembryonic antigen tumor bio-
markers (161). All molecular imaging techniques for MTC
have limited sensitivities:
99m
Tc(V) dimercaptosuccinic
acid (DMSA) (5080%),
201
Tl-thallium (19%),
99m
Tc-
sestamibi (47%),
123/131
I-MIBG (25–30%),
111
In-oc-
treotide (50–75%) (162), and recently,
99m
Tc-EDDA/
HYNIC-TOC (80%) (163).
111
In-octreotide SPECT/CT
was able to depict a paratracheal MTC recurrence (164),
whereas another report described a multiple endocrine
neoplasia (MEN) 2B patient with
99m
Tc-(V)DMSA
SPECT/CT imaging of bone metastases (165). To our
knowledge, due to the rarity of MTC, no dedicated co-
horts have been studied with SPECT/CT, although initial
utility has been reported in small case series (166, 167).
V. Adrenal Cortical Imaging With
131
I-NP59
SPECT/CT
131
I-6-
-iodomethyl-19-norcholesterol (
131
I-NP59) is a
cholesterol precursor analog developed in the early 1970s
that is incorporated into the cholesterol synthesis pathway
via packaging into lipoproteins in the adrenal cortex
(168). Adrenal cortical imaging with
131
I-NP59 has been
used in the workup of hormonally active adrenal nodules
(169). Unilateral
131
I-NP59 uptake has been shown to
correlate with hormone hypersecretion by adrenocortical
adenomas (169) and selects patients suitable for surgical
intervention with high efficacy by lateralizing radiotracer
uptake to the side of hypersecretion (170–173).
Traditionally,
131
I-NP59 scans have been interpreted
on posterior planar images of the upper abdomen acquired
48 days after iv injection of 37 MBq (1 mCi)
131
I-NP59.
Patients with suspected hyperaldosteronism receive phar-
macological adrenosuppressive preparation with dexa-
methasone (174). A large study found that planar
131
I-
NP59 accuracy was 71% in primary aldosteronism and
100% in Cushing’s syndrome (175). The addition of 3D
SPECT images improves contrast resolution and the abil-
ity to detect smaller lesions (176) and allows the use of
semiquantitative parameters such as adrenal-to-liver up-
take ratios to aid diagnosis of pre- and subclinical Cush-
ing’s disease (177, 178).
131
I-NP59 SPECT/CT has proven
useful for primary hyperaldosteronism related to smaller
adenomas (in one report able to detect the culprit 0.8 mm
adrenocortical microadenoma not apparent on CT) (179)
and to assist in the diagnosis of subclinical or atypical
hyperaldosteronism in chronic renal failure (180, 181). In
29 patients with inconclusive adrenal venous sampling,
131
I-NP59 SPECT/CT had sensitivity of 81.8% compared
to SPECT of 68.2% and planar imaging of 40.9% (182).
The increased sensitivity of SPECT/CT was attributed to
the ability to detect functioning adenomas 1.5 cm or
smaller, and as small as 6 mm in one patient (182). One of
the strengths of
131
I-NP59 is for lateralization of hyper-
functioning cortical adenomas predicting biochemical
cure after surgical resection of the adenoma (170–173).
131
I-NP59 imaging is superior to CT for this indication.
SPECT/CT may further refine the utility of the
131
I-NP59
scan for lateralization by allowing direct assessment of
function within adrenal nodules (both for Cushing’s and
Conn’s syndromes) and minimizing false-positive inter-
doi: 10.1210/er.2013-1030 edrv.endojournals.org 727
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
pretations (318). Identification of bilateral adrenal uptake
is important to exclude a process of macrohyperplasia, for
which surgical approaches will be ineffective.
VI. Sympathetic Adrenomedullary Imaging
With SPECT/CT
Chromaffin NETs are derived from sympathomedullary
cells of postganglionic sympathetic neurons (183), the ma-
jority being located in the adrenal medulla or in an extra-
adrenal location near the celiac axis or aortic bifurcation
(organ of Zuckerkandl) (143, 144). Those arising in the
adrenal gland are called pheochromocytomas (PHEOs),
whereas those arising from extra-adrenal tissues are para-
gangliomas (PGLs). Patients may present with symptoms
of biogenic amine excess (141, 183), with PHEO occur-
ring in 0.5% of hypertensive patients and 4% of inciden-
tally discovered adrenal masses (143). PGLs are generally
intra-abdominal in 85%, thoracic in 15%, and cervical in
1–3% of patients (143). Cervical PGLs are usually of para-
sympathetic origin, do not contain chromaffin cells, and
are frequently nonsecretory.
Conventional imaging techniques, CT and magnetic
resonance imaging (MRI), have excellent sensitivities of
93–100% for adrenal PHEO and 90% for extra-adrenal
PGL (141, 183). The specificity is lower, at around 80%,
and another limitation is that cross-sectional, site-targeted
imaging may not readily detect a multifocal process. Func-
tional
123
I-MIBG is a widely used modality that is highly
specific to characterize suspicious adrenal lesions and pro-
vides whole-body screening for metastatic disease (143,
184).
123
I-MIBG has sensitivity of 83–100% and speci-
ficity of 95–100%, preferable to earlier
131
I-MIBG imag-
ing with a lower sensitivity of 77–90% (44, 46, 47). A
recent meta-analysis confirmed high
123
I-MIBG sensitiv-
ity of 94% and specificity of 95% for PHEO imaging
(185). SRS imaging is generally considered a second-line
test to MIBG for PHEO or PGLs; however, SRS is pre-
ferred to MIBG for head and neck PGLs (186–189). Rou-
tine performance of
123
I-MIBG imaging in sporadic, uni-
lateral adrenal masses fully characterized on CT or MRI
has been questioned, particularly because MIBG sensitiv-
ity is lower for detection of extra-adrenal or metastatic
disease (190, 191). However, some authors have sug-
gested a role for MIBG in the presence of genetic mutations
with increased malignant potential (192).
A.
123
I/
131
I-MIBG imaging of pheochromocytoma
and paraganglioma
123/131
I-MIBG SPECT/CT is valuable for evaluation of
PHEO and PGL (193–201). One study found that
SPECT/CT had incremental diagnostic value over planar
scintigraphy in up to 53% of cases (202) (Figure 5). In
another study,
123
I-MIBG SPECT/CT had sensitivity and
specificity of 100% in 22 patients, changing the diagnosis
in three of 14 scan-positive cases and three of eight scan-
negative cases (192).
123/131
I-MIBG SPECT/CT improved
localization and interpretation in 12 of 16 (75%) diag-
nostic cases and nine of 12 (75%) therapeutic cases, es-
pecially when radioactive foci were near or overlapping
regions of physiological MIBG uptake, such as adjacent to
liver, spleen, or bladder (203).
123
I-MIBG SPECT/CT was
compared with MRI in 22 patients with suspected PHEO.
Sensitivity was 87.5% for both modalities, and specificity
of SPECT/CT was 93% as compared to MRI of 97%.
After SPECT/MRI software fusion, there was 100% sen-
sitivity and specificity on a per patient and per lesion anal-
ysis (204). When CT or MRI appearances are indetermi-
nate, the high specificity of
123
I-MIBG imaging has
traditionally been used to confirm that an adrenal mass
represents a PHEO, lateralizing the disease process before
operation to ensure surgical cure (190, 191). In the setting
of known or suspected genetic predisposition, in which
PGL and metastatic disease occur more commonly,
123
I-
MIBG SPECT/CT improves evaluation for staging before
surgery and is of use for depicting bilateral adrenal in-
volvement, which when discovered, would modify the sur-
gical approach.
123
I-MIBG SPECT/CT was compared with
18
F-dihy-
droxyphenylalanine PET/CT in 12 patients with meta-
static PGL.
123
I-MIBG SPECT/CT had a sensitivity of
75% (38% per lesion), compared with
18
F-dihydroxy-
phenylalanine sensitivity of 100% (98% per lesion)
(205). In a comparison study between
123
I-MIBG
SPECT/CT and
11
C-hydroxyephedrine PET/CT,
123
I-
MIBG SPECT/CT had sensitivity of 93% compared to
11
C-hydroxyephedrine PET/CT sensitivity of 99%;
both had specificity of 100% (206). Despite lower sen-
sitivity in these studies,
123
I-MIBG scans retain a unique
role to inform on potential efficacy of
131
I-MIBG
therapy.
B.
123
I/
131
I-MIBG imaging of neuroblastoma
Neuroblastoma is the most common tumor of infants,
accounting for 8–10% of all childhood malignancies
(207). This neoplasm is derived from sympathetic neuro-
blast cells located in the adrenal medulla (4060%), ret-
roperitoneum (20%), and mediastinum (10%) (207).
Functional
123
I-MIBG imaging in neuroblastoma has an
overall sensitivity of 97% and specificity of 100% (185).
The feasibility of software fusion of
123
I-MIBG
SPECT/CT images was first reported in pediatric patients
in 2001 (208). In three children evaluated for neuroblas-
728 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
toma,
123
I-MIBG SPECT/CT confirmed true positive pla-
nar scan findings and clarified the clinical significance of
nonspecific foci of radiotracer uptake. The CT added an
estimated 1.5 mSv radiation dose but provided the advan-
tage of a single imaging session, important for children
who may require sedation for imaging (209). A review of
the utility of pediatric SPECT/CT recommended the com-
bination of contrast-enhanced CT with SPECT to reduce
the number of imaging visits (210).
VII. Parathyroid and Thyroid
Scintigraphy With SPECT/CT
Primary hyperparathyroidism is the
most common cause of hypercalce-
mia in the outpatient setting, affect-
ing two to three of 500 women and
one of 2000 men (211, 212), with a
peak incidence in the fourth and fifth
decades of life (213). Diagnosis is
based on elevated serum and ionized
calcium levels with elevated or nor-
mal intact PTH levels in the presence
of normal renal function (211). Cur-
rently, most patients diagnosed with
hyperparathyroidism are asymp-
tomatic as a result of widespread
screening biochemical laboratory
tests (211). The etiology is due to a
single parathyroid adenoma in 85–
96% of cases, chief cell hyperplasia
in 10%, and a parathyroid cancer in
1–4% (211, 213–215).
Parathyroid adenomas range in size
between 100 mg and 100 g, with a cor-
relation between size and serum calcium
levels (211, 213). The majority are spo-
radic, although associated genetic disor-
ders such as MEN1 and MEN2A and
familial hyperparathyroidism are de-
scribed (211). Secondary hyperpara-
thyroidism arises in the setting of
chronic renal failure, gastrointesti-
nal malabsorption, or osteomala-
cia, with compensation for hy-
pocalcemia (49). So-called “tertiary
hyperparathyroidism” occurs when
compensatory mechanisms result
in autonomous function. Surgical
extirpation of hyperfunctioning
gland(s) is the treatment for primary hy-
perparathyroidism, and for tertiary hy-
perparathyroidism when medical treat-
ment fails (216).
The traditional surgical procedure of bilateral four-
gland exploration without preoperative imaging is cura-
tive in 90% of cases in the hands of an experienced
surgeon. Because similar success rates are achieved with
unilateral exploration via a minimally invasive approach
(217), there has been a shift to preoperative localization of
the “culprit” gland, with or without intraoperative
-probe assistance (49), and intraoperative PTH monitor-
Figure 5.
Figure 5. Pheochromocytoma on
123
I-MIBG scanning in a 54-year-old male with hypertension
and recurrent symptoms of catecholamine hypersecretion. A and B,
123
I-MIBG 24-hour static
planar anterior and posterior images show focal activity in the region of the right adrenal gland
(arrowhead). C–H, Axial and coronal SPECT, CT, and fused SPECT/CT clearly depict a right
adrenal mass with intense focal MIBG accumulation (arrows) compatible with
pheochromocytoma. No additional foci of uptake were seen elsewhere in the body. Note that
the z-axis field-of-view of the SPECT/CT has been limited to reduce radiation exposure because
surgical planning relies mainly on the axial views.
doi: 10.1210/er.2013-1030 edrv.endojournals.org 729
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
ing (success based on a 50% fall in PTH at 10 min after
gland excision) (211). Advantages of the minimally inva-
sive approach include smaller incisions (2- to 4-cm lower
neck incision) with improved cosmetic results, shorter op-
erating times, and faster patient recovery (211, 218). Sur-
gical failure is more common in multiglandular disease
(double adenoma or multiglandular hyperplasia) reported
in 2–5% of patients (211, 216) with the rate of ectopic and
multiglandular disease estimated at 44–70% in reopera-
tive cohorts (216). Furthermore, reoperation for hyper-
parathyroidism has higher failure rates.
The parathyroid glands are usually located in the per-
ithyroid sheath, with the superior glands lying posterior to
the upper third of the thyroid, whereas the inferior glands
are located posterior to the lower third of the thyroid.
Normal glands weigh 2045 mg and measure 6 3mm
and are typically not seen on any imaging modalities. Para-
thyroid tissue is composed of parenchymal chief and mi-
tochondria-rich oxyphilic cells, both of which secrete
PTH. Anatomic variants include accessory parathyroid
glands (13%), three parathyroid glands (3%), and con-
genital absence such as in the Di-George syndrome (211,
213). Ectopic adenomas occur in 10–20% of cohorts; su-
perior parathyroid glands have a 1% ectopic rate, com-
pared with the inferior parathyroid glands exhibiting ec-
topia in 20–35% (211, 219). Ectopic locations include
paraesophageal (28%), mediastinal (26%), intrathymic
(24%), intrathyroidal (11%), carotid sheath (9%), and
high cervical sites (2%) (213).
Imaging modalities for preoperative localization of
parathyroid adenoma include ultrasound, parathyroid
scintigraphy, CT, and MRI (211, 214, 215). Dual-phase
99m
Tc-sestamibi is the most widely used protocol for para-
thyroid scintigraphy (49, 216). Planar images are acquired
with static views of the neck as well as the thorax to include
the mediastinum. Pinhole collimation results in superior
spatial resolution (49, 220), and oblique angle views in-
crease the separation of the posteriorly positioned para-
thyroid adenoma from overlying thyroid tissue. SPECT
has superior sensitivity to planar imaging (221, 222), im-
proving object contrast, removing overlying tissues, and
increasing depth information from 3D images (221, 223,
224). The reported sensitivity of planar parathyroid scin-
tigraphy ranges from 54 to 96% (216). In a meta-analysis
of 20 225 patients with primary hyperparathyroidism,
parathyroid scintigraphy had a sensitivity of 88% for de-
tection of single adenomas; however, sensitivity fell to
30% for multiple adenomas and to 45% for multigland
hyperplasia (225, 226). Detection of rare intrathyroidal
adenomas is challenging (227). In a small study of patients
with tertiary hyperparathyroidism, parathyroid imaging
had a sensitivity of 76% (228).
False-negative studies occur with small adenomas (217,
229); larger adenomas measuring 1.9–3.5 cm are more
likely to be true positive than smaller 0.3- to 1.8-cm lesions
(74 vs 40%; P.001) (224). A prior history of parathy-
roid surgery affects detection rates, with the sensitivity of
parathyroid SPECT reported to decrease from 61% in pa-
tients with no prior surgery to 33% for reoperative ex-
ploration (226).
A.
99m
Tc-sestamibi parathyroid scintigraphy
1. SPECT/CT for parathyroid scintigraphy
Minimally invasive surgical procedures rely upon ac-
curate preoperative localization of hyperfunctioning
parathyroid adenomas. As early as 2002, Rubello et al
(230) reported a patient in whom SPECT/CT precisely
identified an ectopic parathyroid adenoma. The following
year, SPECT/CT depicted ectopic parathyroid glands in
four patients, aiding surgical planning (231). The utility of
SPECT/CT for detection of cervical parathyroid adeno-
mas has been confirmed in multiple subsequent
SPECT/CT studies in primary, secondary, and tertiary hy-
perparathyroidism and subgroups of patients with con-
current nodular thyroid disease, multigland disease, and
in those with previously failed surgery (232–249). Regard-
less of the protocols used, parathyroid scintigraphy
SPECT/CT has been found to improve sensitivity and
specificity for detection of single adenomas, providing key
preoperative localization information. SPECT/CT aids
identification and surgical planning for ectopic parathy-
roid adenomas (211, 213, 216, 250) (Table 5).
2. Strategies and protocols for SPECT/CT
parathyroid scintigraphy
Most studies reporting parathyroid SPECT/CT have
used dual-phase
99m
Tc-sestamibi protocols with doses
ranging between 340 and 1110 MBq (231–237, 239
249).
99m
Tc-sestamibi/
123
I subtraction SPECT/CT with si-
multaneous dual-isotope acquisition is feasible, although
technically demanding (238). Most investigators use hy-
brid
-cameras with integrated SPECT/CT capability
(232–245), although some use software-fused CT-MIBI
from separately acquired SPECT and CT images (246
248). Typically CT parameters have been low-dose, “so-
called” nondiagnostic studies without iv contrast (eg, mAs
2.5 and kVP 140).
Some investigators advocate early time-point SPECT/CT
imaging (15–20 min postinjection) (233, 236–239, 244,
246 –248), whereas others recommend delayed time-point
acquisitions (60–120 min postinjection) (232, 234, 235,
237, 240–243), with no apparent difference in detection
rates between early and delayed SPECT/CT (237).
730 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
3. Image interpretation
SPECT/CT has sensitivities ranging from 70 to 100%
and specificities from 83 to 100%; these vary depending
on cohort studied, protocols used, surgical follow-up,
study design, and whether patient- or lesion-based anal-
ysis was used (232–246). SPECT/CT is clearly more sen-
sitive than planar imaging. In 110
patients with primary hyperparathy-
roidism, SPECT/CT had sensitivity
(72%) compared to SPECT (62%)
and planar (57%) imaging; all had
similar high specificity (98%) (237).
Comparing SPECT to SPECT/CT, a
few authors report similar sensitivity
(233, 238, 239), although most have
found that SPECT/CT increases both
sensitivity (235, 237, 241, 244, 246,
247, 249) and specificity (240, 244).
Overall, there appears to be a sequen-
tial step-up in sensitivity between pla-
nar (59–76%), SPECT (67–97%),
and SPECT/CT (77–100%) imaging
techniques (235, 239, 244).
One of the major advantages of
SPECT/CT has been for preoperative
identification and localization of ad-
enomas in unusual retrotracheal
sites and of ectopic parathyroid ad-
enomas (230, 232, 251–259). The
rates of ectopic locations vary be-
tween 10 and 20% in SPECT/CT co-
horts (231, 232, 234–246) (Figure
6). SPECT/CT is beneficial for surgi-
cal planning, with one study demon-
strating that at surgery, the loca-
Table 5. Utility of
99m
Tc-Sestamibi SPECT/CT for Preoperative Localization of Parathyroid Adenoma
Advantages of SPECT/CT (Refs) Comments and Findings
Lateralization (212, 230–249, 255–259) SPECT/CT is better able to indicate the side of the adenoma, compared to planar and
SPECT.
Neck quadrant localization (232, 235, 239,
242, 244)
SPECT/CT is better able to indicate the neck quadrant of the adenoma, compared to
planar and SPECT.
Ectopic localization (231–234, 236–244,
246–248, 256)
SPECT/CT provides incremental diagnostic value and improves localization of ectopic
glands (rate 10–20%).
Improved diagnostic performance (232, 234
236, 238–246, 248, 249)
99m
Tc-sestamibi SPECT/CT parathyroid scan
Planar: sensitivity, 57–76%; specificity, 77–100%
SPECT: sensitivity, 59–97%; specificity, 79–100%
SPECT/CT: sensitivity, 70–97%; specificity, 83–100%
99m
Tc-sestamibi/
123
I subtraction SPECT/CT parathyroid scan
SPECT: sensitivity, 71%; specificity, 48%
SPECT/CT: sensitivity, 70%; specificity, 96%
Improved interobserver agreement and reader
confidence (233, 237, 243)
Agreement
SPECT/CT
0.76–0.79
SPECT/CT
0.91, SPECT
0.39
Cost-effectiveness and surgical planning (232,
235, 239–241, 244, 246–249)
SPECT/CT use was cost-effective for planning minimally invasive parathyroid adenoma
resection, SPECT/CT assists surgical planning of eu to pic and ec to pic adenomas.
Mean operating time decreased from 56 to 38 min compared to SPECT in one
study and from 62 to 36 min in another study.
Figure 6.
Figure 6. Ectopic parathyroid adenoma in a 73-year-old woman with prior hemithyroidectomy,
presenting with hypercalcemia. A and B, Anterior early 20-minute and delayed 2-hour planar
images show focal radiotracer uptake and retention at the thoracic inlet (arrows). C–E, Axial
SPECT, CT, and SPECT/CT localizes this activity to a soft tissue abnormality in the anterior
mediastinum (arrows), consistent with ectopic parathyroid adenoma. This was surgically removed
via a lower neck incision approach.
doi: 10.1210/er.2013-1030 edrv.endojournals.org 731
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
tion(s) of resected adenomas was 19 mm from the
predicted location derived from the SPECT/CT scan (234).
SPECT/CT improves interobserver agreement and
reader confidence (243). One study found SPECT alone
had a poor
value of 0.39 compared to SPECT/CT
value
of 0.91 (233). Interobserver agreement in another study
was highest (
0.76 – 0.79) for dual-phase imaging when
SPECT/CT was included and lowest for single-phase im-
aging, regardless of protocol (
0.500.61) (237).
4. Special patient subgroups
SPECT/CT is useful in patients who have previously
undergone surgery (257). In 28 patients with a prior his-
tory of thyroid or parathyroid surgery, SPECT/CT pre-
dicted the site of the “missing” gland in 86% of patients,
compared to only 43% with SPECT alone (248), provid-
ing useful anatomic information in the presence of dis-
torted postsurgical neck anatomy owing to prior surgery
(236). In a cohort of patients selected for the presence of
multiglandular disease, the overall imaging sensitivity was
low, with SPECT positive in only four of 30 (13.3%) pa-
tients, CT in 11 of 30 (36.7%), but with slightly increased
detection rates using SPECT/CT (46.7%) (246). The ac-
curacy of SPECT/CT in patients with coexisting nodular
thyroid disease has been studied in 48 hyperparathyroid
patients with concurrent nodular thyroid glands; early
time-point SPECT/CT had a sensitivity of 77% and spec-
ificity of 97%, superior to SPECT sensitivity of 67% and
specificity of 87% (244). Others have also reported similar
findings (240, 242). In addition to multinodular thyroid
goiters, false-positive findings may also occur due to in-
flammatory disease, thyroid cancer, brown fat, osteitis
fibrosa cystica (brown tumors), and autotransplanted
glands (216, 260–263). Similar to FDG PET/CT, where
FDG uptake can occur in metabolically active brown ad-
ipose tissue,
99m
Tc-sestamibi uptake in mitochondria-rich
brown fat is identified on SPECT/CT in 6.3% cases with
implications for false-positive interpretative errors
(264–266).
5. Diagnostic workup of hyperparathyroidism and role of
parathyroid SPECT/CT
At the present time, most patients with hyperparathy-
roidism are asymptomatic at diagnosis (267). Selection for
surgery is based on calcium levels, renal function, bone
mineral densitometry, age 50 years, and symptomatol-
ogy. Nonparathyroid causes of hypercalcemia such as ma-
lignancy, familial hypocalciuric hypercalcemia, and med-
ication effects should be excluded (267). Neck ultrasound
and parathyroid scintigraphy have equivalent diagnostic
performance and are accepted as first-line imaging tests
(268–271). When there is unequivocal identification of a
parathyroid adenoma on either test, the surgeon may pro-
ceed confidently to a minimally invasive procedure (218).
Parathyroid scintigraphy has the advantage of being able
to detect extracervical ectopically located adenomas,
whereas neck ultrasound is best utilized when there is co-
existing nodular thyroid disease (121, 218). By contrast,
CT and MRI have not been recommended as primary im-
aging alternatives due mainly to low sensitivity for detec-
tion of parathyroid disease.
If both parathyroid scintigraphy and neck ultrasound
are negative, then four-dimensional (4D)-CT has been
proposed as an alternative test with good sensitivity and
specificity (218, 272). Its major disadvantage to date has
been the higher radiation dose to the thyroid gland, lim-
iting its use in the younger patient population. In the sit-
uation when all imaging tests are negative, some authors
have advocated proceeding to four-gland exploration, ac-
cepting a lower cure rate of around 89% (273, 274). Ob-
servation with serial imaging is another valid option. Pa-
tients with recurrent hyperparathyroidism in the setting of
a prior history of neck surgery are challenging to manage
due to more difficult reoperation and higher failure rates.
They warrant a comprehensive imaging evaluation, and
additional strategies proposed include selective venous
sampling (275), 4D-CT (218), and
11
C-methionine PET
(276). Table 6 summarizes diagnostic performance of var-
ious procedures and imaging tests for parathyroid ade-
noma localization.
Several studies have shown that the use of SPECT/CT
improves lateralization rates of parathyroid adenoma and
is more successful for quadrant localization compared to
planar and SPECT imaging. However, there is still some
difficulty in ascribing whether the location of a detected
adenoma is superior or inferior when correlated with the
surgical findings (232, 235, 240–242, 244, 277). The ad-
vantages of SPECT/CT for assisting the planning of the
minimally invasive surgical approach recommend its rou-
tine use, when available. Preoperative detection of an ec-
topic adenoma on SPECT/CT is unquestionably of value
before surgery (278). SPECT/CT provides accurate assess-
ment in patients with coexisting thyroid nodular disease
(240, 242, 279), particularly when combined with neck
ultrasound (242). However, for patients with multiglan-
dular disease or double adenomas, and for those under-
going reoperative surgical procedures, the sensitivity of
SPECT/CT appears lower (246, 248, 281) when com-
pared to selective venous sampling (281), and therefore it
is not expected to have a major impact for these more
challenging patient subgroups.
732 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
B.
99m
Tc-pertechnetate thyroid scans
SPECT/CT has been used with
99m
Tc-pertechnetate
thyroid scintigraphy to localize uptake within a thyroglos-
sal duct cyst (283) and to evaluate Marine-Lenhart syn-
drome, the rare entity of Graves’ disease combined with
hyperfunctioning thyroid nodules (284). Traditionally,
thyroid scintigraphy is correlated with thyroid ultra-
sound; however, SPECT/CT offers a more direct method
to assess the functional status of thyroid nodules (285,
286). Two studies have reported the use of
123
I SPECT/CT
to depict uptake in an obstructive lingual thyroid (124,
287). A lingual thyroid was successfully treated with ra-
dioiodine ablation in a patient at high surgical risk (287)
(Figure 7).
Table 6. Diagnostic Performance of Imaging Studies, Catheter-Based, and Surgical Procedures for Localization of
Parathyroid Adenoma
Test or Procedure
Overall
Sensitivity
Range, %
Pooled Sensitivity
(95% CI) Comments
Neck ultrasound Accepted role as first-line test
Primary HPT 70–100 76% (70–81) Ultrasound performed by surgeons or radiologists is equivalent.
Single adenoma N/A 79% (77–80) Readily available. No ionizing radiation.
Multiglandular disease N/A 35% (30–40) Concurrent evaluation for thyroid nodular disease.
Double adenoma N/A 16% (4–28) Operator dependent. Difficult in obese patients.
Thyroid abnormalities 47– 84 N/A Unable to detect ectopic glands.
Prior neck surgery 36–63 N/A
Sestamibi scintigraphy Accepted role as first-line test.
Planar and SPECT Able to detect ectopic glands.
Primary HPT 54–100 79% (64 –91) SPECT is more sensitive than planar.
Secondary HPT 35–90 58% (52–65) False-positive results due to thyroid nodules.
Single adenoma 54–96 88% (87– 89)
Multiglandular disease 25–58 45% (41–48)
Double adenoma N/A 30% (2to62)
Thyroid abnormalities Decreased N/A
SPECT/CT N/A Novel technology being validated.
Primary HPT 70–100 N/A SPECT/CT is more specific than SPECT.
Multiglandular disease 47 N/A Good localization of ectopic glands.
Thyroid abnormalities 77–90 N/A Improved lateralization and neck quadrant localization
compared to SPECT.Prior neck surgery 26–85
CT 40–86 45% Not considered first line.
Generally low sensitivity and high radiation exposure.
MRI 43–88 54% Not considered first line.
Generally low sensitivity and low specificity due to lymph nodes
and thyroid nodules.
Selective venous sampling “Gold standard” invasive test.
Primary HPT 71 N/A Possible role in reoperative cohorts.
Prior neck surgery 71–90 N/A
Four-gland exploration 90 –95% cure rate N/A Historical “gold standard.”
Intraoperative PTH monitoring 97 N/A Used for guiding minimally invasive surgery.
Useful for multiglandular or double adenomas.
High rates of cure (up to 97%) have been reported without use
of intraoperative PTH monitoring.
4D-CT 94 89% Novel technique being validated.
Possible role in reoperative cohorts.
High radiation dose to the thyroid.
Difficult interpretive criteria.
11
C-MET PET
Primary HPT
Prior neck surgery 83 81% (74–86) Novel technique being validated.
67 N/A Potential role in reoperative cohorts.
Poor availability.
FDG PET 86 N/A Novel technique being validated.
Surgical exploration with negative
imaging tests
89% cure rate N/A Lower cure rate when imaging tests are negative.
Balance lower cure rate against symptoms.
Resort to four-gland exploration vs observation with repeat
imaging.
Abbreviations: CI, confidence interval; HPT, hyperparathyroidism; MET, methionine; N/A, not available. Composite table from the following primary and secondary
sources: Refs. 121, 218, 225, 268–276.
doi: 10.1210/er.2013-1030 edrv.endojournals.org 733
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
C. Miscellaneous endocrine applications of SPECT/CT
SPECT/CT has been used in the research setting to de-
velop novel applications for the study of endocrine disor-
ders (288). For Graves’ eye disease,
99m
Tc-EDDA/
HYNIC-TOC SPECT/CT was used to localize radiotracer
uptake to the orbits. Orbital
99m
Tc-TOC uptake was re-
lated to the activity of Graves’ orbitopathy and may be a
feasible technique to assess treatment response to corti-
costeroids (289). Novel preoperative
99m
Tc-folate
SPECT/CT was used to investigate nonfunctioning pitu-
itary adenomas (290).
VIII. Current Clinical Role of SPECT/CT in the
Diagnostic Workup of Endocrine Disorders
A. Protocols for SPECT/CT use
Currently, SPECT/CT cameras, in a manner similar to
PET/CT, are becoming more widely disseminated in the
United States, predominately at tertiary medical institu-
tions. SPECT/CT is a technological tool applied to the
radionuclide study, rather than a type of imaging modality
in itself. Protocols for SPECT/CT include routine use vs
selective indications to localize and evaluate radioactivity
foci that are otherwise problematic to interpret on planar
or SPECT, based on the growing list of indications and
evidence for utility with endocrine scintigraphy (9). The
SPECT/CT component for parathy-
roid scintigraphy was recently ap-
proved for Centers for Medicare and
Medicaid Services reimbursement, al-
though other indications for SPECT/
CT do not as yet have separate bill
codings. In most institutions, the CT
portion of the study is acquired with
nondiagnostic (low-dose kVP and
mAs) parameters without iv con-
trast, with the intent to perform an-
atomical localization and attenua-
tion correction (291). Therefore,
SPECT/CT does not replace dedi-
cated diagnostic contrast-enhanced
CT, although SPECT/CT use in a
problem-solving capacity may obvi-
ate the need for further workup of
equivocal findings with diagnostic
CT imaging.
B. Economic evaluation and
cost-effectiveness
The current economic climate
mandates scrutiny directed at the ris-
ing costs of healthcare, and the cli-
nician should be mindful of the increasing costs related to
medical imaging while ordering appropriate investiga-
tions for their patients. Economic evaluation studies gen-
erally require descriptions of a given intervention in com-
parison to alternative tests or treatments, the perspective
of the study, the measured outcomes, the method of cost-
ing including adjustments, an outline of the time horizon,
and a discussion of uncertainties related to the analysis
(292, 293), helping to give insight into how best to allocate
scarce healthcare resources.
Cost-effectiveness analysis is the most appropriate type
of study for comparing an imaging modality to an alter-
native test, with the costs and beneficial effects on out-
comes examined and weighed against each other (294).
Economic evaluation studies in nuclear medicine in the
pre-SPECT/CT era have predominately been focused on
proving the effectiveness of PET for cancer staging and
myocardial perfusion studies for the diagnosis of coronary
artery disease. For endocrine disorders, cost-effectiveness
has been reported for radioiodine as first-line treatment of
Graves’ hyperthyroidism compared to surgery (294), ra-
dioiodine uptake measurements in the diagnostic workup
of thyrotoxicosis (295), evaluation of thyroid nodule ma-
lignant potential with sestamibi scans (296, 297), staging
of NETs using somatostatin scintigraphy (298, 299),
workup of adrenal incidentaloma with
131
I-NP59 imaging
Figure 7.
Figure 7. A 53-year-old woman with hypothyroidism had progressive globus sensation and
symptoms of airway obstruction when lying supine. Radioiodine 0.5 mCi
123
I planar scan at 4
and 24 hours showed absent thyroid gland in the neck with a focal uptake in the region of the
oral cavity (not shown). A–F, CT and
123
I SPECT/CT fusion images precisely localized radioiodine
uptake to a soft tissue mass at the base of the tongue (arrows), with higher attenuation than
surrounding soft tissue. The 24-hour
123
I uptake within this lingual thyroid was measured at
13.5%.
734 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
(300, 301), and use of
131
I-MIBG therapy for treatment of
carcinoid syndrome (302). Parathyroid scintigraphy for
preoperative localization of adenomas is cost-effective be-
cause the costs of protracted or failed surgery due to an
unidentified ectopic adenoma outweigh those related to
imaging (292, 303, 304). Another study showed that min-
imally invasive parathyroid surgery using preoperative
parathyroid scintigraphy led to a $3000 reduction in
charges compared to nondirected bilateral neck explora-
tion, attributed to reduced operative times, early hospital
discharge, and avoidance of re-exploration (305).
To date, the number of studies examining the cost-ef-
fectiveness of SPECT/CT applied to endocrine scintigra-
phy is limited to parathyroid scintigraphy (306). Table 7
summarizes the current published evidence on endocrine
SPECT/CT for diagnostic performance and economic
evaluation. Parathyroid SPECT/CT results in a reduction
in mean operating times from 56 to 38 minutes, compared
to SPECT alone. The same authors also reported in 55
patients (27 with SPECT and 28 with SPECT/CT) a re-
duction in mean operating times from 62 to 36 minutes,
resulting in estimated cost savings of 98 (US $136) per
patient (240). However, these results are not directly ap-
plicable to costs in the United States.
Economic evaluation of an emerging imaging technol-
ogy early in its clinical deployment may lack generaliz-
ability. Furthermore, outcome measurements and cost-ef-
fectiveness analyses of imaging tests are often difficult to
perform because technological advancement regularly
outpaces peer-reviewed medical literature. For example,
recent studies have reported cost-effectiveness of 4D-CT
or neck ultrasound when compared to parathyroid scin-
tigraphy (planar and SPECT); however, these imaging mo-
dalities have not yet been compared to parathyroid
SPECT/CT (307, 308). Future economic evaluation stud-
ies of SPECT/CT to determine its role in the diagnosis of
endocrine disorders are anticipated to be forthcoming.
C. Guidelines for indications and role
A number of medical organizations and societies have
advocated the use of SPECT/CT in their guidelines on the
management and treatment of endocrine disorders. For
example, the Society of Nuclear Medicine and Molecular
Imaging (SNMMI) 2006 procedure guideline for
SPECT/CT lists imaging of tumors, thyroid, and parathy-
roid disorders as appropriate indications for SPECT/CT
(291), with the strongest evidence of utility of SPECT/CT
found for parathyroid scintigraphy and thyroid cancer im-
aging, followed by octreotide and MIBG imaging (9). Both
the SNMMI and the European Association of Nuclear
Medicine and Molecular Imaging (EANMMI) guidelines
on parathyroid scintigraphy recommend a major role for
SPECT/CT for localization of ectopic parathyroid adeno-
mas (213, 214). Its routine use in the preoperative setting
remains investigational at present. The SNMMI 2012
guidelines on radioiodine therapy of thyroid disease dis-
cuss a potential role for preablation diagnostic
123
Ior
131
I
whole-body thyroid cancer scans with SPECT/CT in the
immediate postsurgical setting to detect unsuspected re-
gional and distant metastatic disease, potentially changing
staging and management (309). The American Thyroid
Association 2009 and EANMMI 2008 guidelines on thy-
roid cancer management discuss the use of SPECT/CT as
an emerging imaging technique, including a potential fu-
ture role for performing dosimetry (6, 310). The American
Association of Clinical Endocrinologists/American Col-
lege of Endocrinology thyroid cancer guidelines include
the use of diagnostic SPECT, although not SPECT/CT at
Table 7. Test Performance and Economic Evaluation Studies of SPECT/CT for Endocrine Scintigraphy
Parameters
131
I/
123
I Thyroid
Cancer Scan
111
In-DTPA
Octreotide
Scan
131
I/
123
I-MIBG
Scan
131
I-NP59
Adrenal
Cortical Scan
99m
Tc-Sestamibi
Parathyroid
Scan
Localization ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹ ⫹⫹⫹⫹
Characterization ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹ ⫹⫹⫹⫹
Clarification of equivocal uptake ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹ ⫹⫹ ⫹⫹
Sensitivity ⫹⫹ ⫹⫹ ⫹⫹⫹
Specificity ⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹⫹
Interobserver agreement ⫹⫹⫹ ⫹⫹⫹ ⫺ ⫹⫹⫹
Staging ⫹⫹⫹⫹ ⫹⫹⫹ ⫺ N/A N/A
Preoperative localization N/A ⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹⫹
Surgical times reduced N/A N/A N/A N/A ⫹⫹⫹
Dosimetry ⫹⫹⫹ ⫹⫹⫹ ⫹⫹ N/A N/A
Cost-effectiveness ⫺⫺
Ordering of additional imaging reduced ⫹⫹ ⫹⫹ ⫹⫹
Change in management ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹
Improved quality of life ⫺⫺
Increased life expectancy ⫺⫺
Abbreviation: N/A, not available.
doi: 10.1210/er.2013-1030 edrv.endojournals.org 735
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
present (311). The SNMMI and EANMMI guidelines on
octreotide imaging consider SPECT as the standard of
care, and when available, SPECT/CT is recommended for
attenuation correction and localization of octreotide-avid
lesions (312, 313). The SNMMI guidelines for MIBG im-
aging of paraganglioma and pheochromocytoma state
that the use of SPECT/CT is “highly recommended” due
to improved diagnostic accuracy (314), a sentiment shared
by the EANMMI guidelines (315). Dosimetry calculations
based on SPECT and SPECT/CT have been advocated for
131
I-MIBG therapy (314) and for somatostatin peptide
receptor therapy (316). The use of SPECT/CT for other
endocrine disorders remains investigational, with no cur-
rent role recommended in the workup of thyroid or ad-
renal nodules (6, 132, 136, 280, 282, 317).
D. Strategies for reducing radiation exposure
Radiation exposure due to medical sources continues to
increase, with much of the increase attributed to repetitive
radiological procedures both for diagnostic intent and fol-
low-up. The largest contribution to overall medical radi-
ation exposure is from contrast-enhanced CT, which may
require multiple image acquisitions during various phases
of contrast washout. Image quality for CT is strongly de-
pendent on x-ray intensity (ie, exposure); for this reason,
the compromise made is summarized with the As Low As
Reasonably Achievable (ALARA) principle to achieve the
highest diagnostic quality imaging while using the lowest
radiation exposure for patients. The issue of dose reduc-
tion from medical imaging is of great importance, and in
recent years software has been developed; in particular,
the latest-generation iterative reconstruction algorithms
designed for diagnostic CT are able to reduce image noise
while improving the overall image quality as compared to
older filtered back-projection image reconstruction meth-
ods. Increasing use of SPECT/CT, normally performed
with low-dose noncontrast enhanced CT protocols, is an-
ticipated to only modestly contribute to collective radia-
tion doses. Nonetheless, many strategies can be used to
achieve a reduction in patient dose from SPECT/CT, in-
cluding: 1) optimization of the CT acquisition protocols
(tube current, peak voltage, rotation time, helical pitch) in
order to obtain data for localization and SPECT attenu-
ation correction; 2) the systematic use of software able to
reduce the patient CT dose, in particular the automatic
exposure control, the anatomically adapted tube current
modulation, and adopting the latest-generation iterative
image reconstruction algorithms; 3) the use of the mini-
mum CT scan field-of-view coverage tailored for each im-
aging procedure; 4) correct selection of patients and
procedure; 5) consideration of alternative imaging proce-
dures, such as ultrasound or MRI, that do not have ion-
izing radiation effects, when appropriate; 6) a careful se-
lection in the pediatric population, using examination
protocols modified according to the child’s size; and 7)
avoidance of repetitive diagnostic CT for evaluations of
equivocal findings.
IX. Summary and Future Directions
Hybrid SPECT/CT is a powerful diagnostic tool that al-
lows precise localization of the distribution of radioactiv-
ity uptake from radiopharmaceuticals, reducing the num-
ber of equivocal findings encountered on traditional
planar scintigraphy, with the functional-anatomic coreg-
istration increasing accuracy of imaging interpretation.
Further advantages of the SPECT/CT approach include
incremental gains in interobserver agreement, reader con-
fidence, and improved staging of endocrine malignancies,
with SPECT/CT providing unique prognostic information
that alters subsequent patient management. Future re-
search directions include defining the role of SPECT/CT in
the investigation of endocrine disorders, improving quan-
titative methods for tumor-specific dosimetry, and explo-
ration of possible utility of novel hybrid SPECT/MRI, and
other unique combinations of imaging modalities.
Acknowledgments
Address all correspondence and requests for reprints to: Ka Kit Wong,
MBBS, University of Michigan Health System, Department of Radiol-
ogy, Division of Nuclear Medicine, B1G505 University Hospital SPC
5028, 1500 East Medical Center Drive, Ann Arbor, MI 48109–5028.
E-mail: kakit@med.umich.edu.
Disclosure Summary: The authors have nothing to declare.
References
1. Chung JK. Sodium iodide symporter: its role in nuclear
medicine. J Nucl Med. 2002;43(9):1188–1200.
2. Dai G, Levy O, Carrasco N. Cloning and characterization
of the thyroid iodide transporter. Nature. 1996;379(6564):
458460.
3. Smanik PA, Liu Q, Furminger TL, et al. Cloning of the
human sodium iodide symporter. Biochem Biophys Res
Commun. 1996;226(2):339–345.
4. Daniels GH. Radioactive iodine: a slice of history. Thyroid.
2013;23(3):253–258.
5. Means JH. Historical background of the use of radioactive
iodine in medicine. N Engl J Med. 1955;252(22):936 –940.
6. Cooper DS, Doherty GM, Haugen BR, et al. Revised Amer-
ican Thyroid Association management guidelines for pa-
tients with thyroid nodules and differentiated thyroid can-
cer. Thyroid. 2009;19(11):1167–1214.
736 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
7. Buck AK, Nekolla S, Ziegler S, et al. SPECT/CT. J Nucl
Med. 2008;49(8):1305–1319.
8. Histed SN, Lindenberg ML, Mena E, Turkbey B, Choyke
PL, Kurdziel KA. Review of functional/anatomical imaging
in oncology. Nucl Med Commun. 2012;33(4):349–361.
9. Mariani G, Bruselli L, Kuwert T, et al. A review on the
clinical uses of SPECT/CT. Eur J Nucl Med Mol Imaging.
2010;37(10):1959–1985.
10. Yap KS, Patel CN, Chowdhury FU, Scarsbrook AF. Less
commonly used and emerging clinical applications of
SPECT-CT in benign and malignant disease. Nucl Med
Commun. 2012;33(8):808818.
11. Anger HO. Scintillation camera. Rev Sci Instrum. 1958;
29:27–33.
12. Peterson TE, Furenlid LR. SPECT detectors: the Anger
camera and beyond. Phys Med Biol. 2011;56(17):R145–
R182.
13. Kuhl DE, Edwards RQ. Image separation radioisotope
scanning. Radiology. 1963;80:653–662.
14. Ambrose J, Hounsfield G. Computerized transverse axial
tomography. Br J Radiol. 1973;46:148–149.
15. Hutton B, Nuyts J, Zaidi H. Iterative reconstruction meth-
ods. In: Zaida H, ed. Quantitative Analysis in Nuclear
Medicine Imaging. New York, NY: Springer; 2006:107–
140.
16. Goitein M. Three dimensional density reconstruction from
a series of two-dimensional projections. Nucl Instrum
Methods. 1972;101(3):509–518.
17. Chang LT. A method for attenuation correction in radio-
nuclide computed tomography. IEEE Trans Nucl Sci.
1978;25:638643.
18. Bailey DL. Transmission scanning in emission tomogra-
phy. Eur J Nucl Med. 1998;25(7):774–787.
19. Lang TF, Hasegawa BH, Liew SC, et al. Description of a
prototype emission-transmission computed tomography
imaging system. J Nucl Med. 1992;33(10):1881–1887.
20. Even-Sapir E, Keidar Z, Sachs J, et al. The new technology
of combined transmission and emission tomography in
evaluation of endocrine neoplasms. J Nucl Med. 2001;
42(7):998–1004.
21. Schillaci O. Functional-anatomical image fusion in neu-
roendocrine tumors. Cancer Biother Radiopharm. 2004;
19(1):129–134.
22. Patton JA, Turkington TG. SPECT/CT physical principles
and attenuation correction. J Nucl Med Technol. 2008;
36(1):1–10.
23. Zaidi H, Hasegawa B. Determination of the attenuation
map in emission tomography. J Nucl Med. 2003;44(2):
291–315.
24. Goldman LW. Principles of CT: multislice CT. J Nucl Med
Technol. 2008;36(2):57–68; quiz 75–76..
25. Ritt P, Vija H, Hornegger J, Kuwert T. Absolute quanti-
fication in SPECT. Eur J Nucl Med Mol Imaging. 2011;
38(suppl 1):S69–S77.
26. Jaszczak RJ, Greer KL, Floyd CE Jr, Harris CC, Coleman
RE. Improved SPECT quantification using compensation
for scattered photons. J Nucl Med. 1984;25(8):893–900.
27. Koral KF, Wang XQ, Rogers WL, Clinthorne NH, Wang
XH. SPECT Compton-scattering correction by analysis of
energy spectra. J Nucl Med. 1988;29(2):195–202.
28. Meikle SR, Hutton BF, Bailey DL. A transmission-depen-
dent method for scatter correction in SPECT. J Nucl Med.
1994;35(2):360–367.
29. Welch A, Gullberg GT, Christian PE, Datz FL, Morgan
HT. A transmission-map-based scatter correction tech-
nique for SPECT in inhomogeneous media. Med Phys.
1995;22(10):1627–1635.
30. Erlandsson K, Buvat I, Pretorius PH, Thomas BA, Hutton
BF. A review of partial volume correction techniques for
emission tomography and their applications in neurology,
cardiology and oncology. Phys Med Biol. 2012;57(21):
R119–R159.
31. Du Y, Tsui BM, Frey EC. Partial volume effect compen-
sation for quantitative brain SPECT imaging. IEEE Trans
Med Imaging. 2005;24(8):969–976.
32. Soret M, Koulibaly PM, Darcourt J, Hapdey S, Buvat I.
Quantitative accuracy of dopaminergic neurotransmission
imaging with (123)I SPECT. J Nucl Med. 2003;44(7):
1184–1193.
33. Kovalski G, Israel O, Keidar Z, Frenkel A, Sachs J, Azhari
H. Correction of heart motion due to respiration in clinical
myocardial perfusion SPECT scans using respiratory gat-
ing. J Nucl Med. 2007;48(4):630636.
34. Eisner RL, Noever T, Nowak D, et al. Use of cross-corre-
lation function to detect patient motion during SPECT im-
aging. J Nucl Med. 1987;28(1):97–101.
35. Dey J, King MA. Theoretical and numerical study of
MLEM and OSEM reconstruction algorithms for motion
correction in emission tomography. IEEE Trans Nucl Sci.
2009;56(5):2739–2749.
36. Feng B, Gifford HC, Beach RD, Boening G, Gennert MA,
King MA. Use of three-dimensional Gaussian interpolation
in the projector/back projector pair of iterative reconstruc-
tion for compensation of known rigid-body motion in
SPECT. IEEE Trans Med Imaging. 2006;25(7):838844.
37. Schumacher H, Fischer B. A new and flexible reconstruc-
tion framework for motion correction in SPECT Imaging.
IEEE Trans Nucl Sci. 2007;54(3):480485.
38. Dewaraja YK, Frey EC, Sgouros G, et al. MIRD pamphlet
no. 23: quantitative SPECT for patient-specific 3-dimen-
sional dosimetry in internal radionuclide therapy. J Nucl
Med. 2012;53(8):1310–1325.
39. Larkin AM, Serulle Y, Wagner S, Noz ME, Friedman K.
Quantifying the increase in radiation exposure associated
with SPECT/CT compared to SPECT alone for routine nu-
clear medicine examinations. Int J Mol Imaging. 2011;
2011:897202.
40. Mettler FA Jr, Huda W, Yoshizumi TT, Mahesh M. Ef-
fective doses in radiology and diagnostic nuclear medicine:
a catalog. Radiology. 2008;248(1):254–263.
41. Park HM. 123I: almost a designer radioiodine for thyroid
scanning. J Nucl Med. 2002;43(1):77–78.
42. Al-Nahhas A, Win Z, Szyszko T, et al. Gallium-68 PET: a
new frontier in receptor cancer imaging. Anticancer Res.
2007;27(6B):4087–4094.
43. Ambrosini V, Castellucci P, Rubello D, et al. 68Ga-DOTA-
NOC: a new PET tracer for evaluating patients with bron-
chial carcinoid. Nucl Med Commun. 2009;30(4):281–
286.
44. Koopmans KP, Neels ON, Kema IP, et al. Molecular im-
aging in neuroendocrine tumors: molecular uptake mech-
doi: 10.1210/er.2013-1030 edrv.endojournals.org 737
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
anisms and clinical results. Crit Rev Oncol Hematol. 2009;
71(3):199–213.
45. Kwekkeboom DJ, Kam BL, van Essen M, et al. Somatosta-
tin-receptor-based imaging and therapy of gastroentero-
pancreatic neuroendocrine tumors. Endocr Relat Cancer.
2010;17(1):R53–R73.
46. Bombardieri E, Seregni E, Villano C, Chiti A, Bajetta E.
Position of nuclear medicine techniques in the diagnostic
work-up of neuroendocrine tumors. Q J Nucl Med Mol
Imaging. 2004;48(2):150–163.
47. Bushnell DL, Baum RP. Standard imaging techniques for
neuroendocrine tumors. Endocrinol Metab Clin North
Am. 2011;40(1):153–162, ix.
48. van Hulsteijn LT, Niemeijer ND, Dekkers OM, Corssmit
EP. (131)I-MIBG therapy for malignant paraganglioma
and phaeochromocytoma: systematic review and meta-
analysis. Clin Endocrinol (Oxf). 2014;80(4):487–501.
49. Hindié E, Zanotti-Fregonara P, Just PA, et al. Parathyroid
scintigraphy findings in chronic kidney disease patients
with recurrent hyperparathyroidism. Eur J Nucl Med Mol
Imaging. 2010;37(3):623–634.
50. Taillefer R, Boucher Y, Potvin C, Lambert R. Detection
and localization of parathyroid adenomas in patients with
hyperparathyroidism using a single radionuclide imaging
procedure with technetium-99m-sestamibi (double-phase
study). J Nucl Med. 1992;33(10):1801–1807.
51. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013.
CA Cancer J Clin. 2013;63(1):11–30.
52. Davies L, Welch HG. Increasing incidence of thyroid can-
cer in the United States, 1973–2002. JAMA. 2006;295(18):
2164–2167.
53. Chen AY, Jemal A, Ward EM. Increasing incidence of dif-
ferentiated thyroid cancer in the United States, 1988
2005. Cancer. 2009;115(16):3801–3807.
54. Hay ID, McConahey WM, Goellner JR. Managing pa-
tients with papillary thyroid carcinoma: insights gained
from the Mayo Clinic’s experience of treating 2,512 con-
secutive patients during 1940 through 2000. Trans Am
Clin Climatol Assoc. 2002;113:241–260.
55. Jonklaas J, Sarlis NJ, Litofsky D, et al. Outcomes of pa-
tients with differentiated thyroid carcinoma following ini-
tial therapy. Thyroid. 2006;16(12):1229–1242.
56. Mazzaferri EL, Jhiang SM. Long-term impact of initial sur-
gical and medical therapy on papillary and follicular thy-
roid cancer. Am J Med. 1994;97(5):418428.
57. British Thyroid Association, Royal College of Physicians.
Guidelines for the management of thyroid cancer. In: Per-
ros P, ed. Report of the Thyroid Cancer Guidelines Update
Group. 2nd ed. London: Royal College of Physicians;
2007.
58. Hay ID. Management of patients with low-risk papillary
thyroid carcinoma. Endocr Pract. 2007;13(5):521–533.
59. Mazzaferri EL. Management of low-risk differentiated
thyroid cancer. Endocr Pract. 2007;13(5):498–512.
60. Bourgeois P. A proposition for the use of radioiodine in
WDTC management. J Nucl Med. 2009;50(2):328–329.
61. Delbeke D, Schöder H, Martin WH, Wahl RL. Hybrid
imaging (SPECT/CT and PET/CT): improving therapeutic
decisions. Semin Nucl Med. 2009;39(5):308–340.
62. Filesi M, Signore A, Ventroni G, Melacrinis FF, Ronga G.
Role of initial iodine-131 whole-body scan and serum thy-
roglobulin in differentiated thyroid carcinoma metastases.
J Nucl Med. 1998;39(9):1542–1546.
63. Lind P, Kohlfürst S. Respective roles of thyroglobulin, ra-
dioiodine imaging, and positron emission tomography in
the assessment of thyroid cancer. Semin Nucl Med. 2006;
36(3):194–205.
64. Basu S, Abhyankar A. Role of SPECT/CT, versus tradi-
tional practices, in individualizing treatment of thyroid
carcinoma. Author reply. J Nucl Med. 2012;53:1819
1820.
65. Avram AM. Radioiodine scintigraphy with SPECT/CT: an
important diagnostic tool for thyroid cancer staging and
risk stratification. J Nucl Med. 2012;53(5):754–764.
66. Barwick TD, Dhawan RT, Lewington V. Role of
SPECT/CT in differentiated thyroid cancer. Nucl Med
Commun. 2012;33(8):787–798.
67. Toubert ME, Vija L, Vercellino L, et al. Additional diag-
nostic value of hybrid SPECT-CT systems imaging in pa-
tients with differentiated thyroid cancer. Am J Clin Oncol.
2014;37(3):305–313.
68. Xue YL, Qiu ZL, Song HJ, Luo QY. Value of
131
I
SPECT/CT for the evaluation of differentiated thyroid can-
cer: a systematic review of the literature. Eur J Nucl Med
Mol Imaging. 2013;40(5):768–778.
69. Wong KK, Dvorak RA, Marzola MC, Grassetto G, Gross
MD, Rubello D. Molecular imaging in the management of
thyroid cancer. Q J Nucl Med Mol Imaging. 2011;55(5):
541–559.
70. Aide N, Heutte N, Rame JP, et al. Clinical relevance of
single-photon emission computed tomography/computed
tomography of the neck and thorax in postablation (131)I
scintigraphy for thyroid cancer. J Clin Endocrinol Metab.
2009;94(6):2075–2084.
71. Aqueveque AC, González EP, Gutiérrez BD, et al. Fusion
of SPECT with computed tomography or magnetic reso-
nance for the interpretation of abnormal tracer uptake [in
Spanish]. Rev Med Chil. 2007;135(6):725–734.
72. Barwick T, Murray I, Megadmi H, et al. Single photon
emission computed tomography (SPECT)/computed to-
mography using iodine-123 in patients with differentiated
thyroid cancer: additional value over whole body planar
imaging and SPECT. Eur J Endocrinol. 2010;162(6):
1131–1139.
73. Blum M, Tiu S, Chu M, Goel S, Friedman K. I-131
SPECT/CT elucidates cryptic findings on planar whole-
body scans and can reduce needless therapy with I-131 in
post-thyroidectomy thyroid cancer patients. Thyroid.
2011;21(11):1235–1247.
74. Chen L, Luo Q, Shen Y, et al. Incremental value of 131I
SPECT/CT in the management of patients with differenti-
ated thyroid carcinoma. J Nucl Med. 2008;49(12):1952–
1957.
75. Ciappuccini R, Heutte N, Trzepla G, et al. Postablation
(131)I scintigraphy with neck and thorax SPECT-CT and
stimulated serum thyroglobulin level predict the outcome
of patients with differentiated thyroid cancer. Eur J Endo-
crinol. 2011;164(6):961–969.
76. Kim HY, Gelfand MJ, Sharp SE. SPECT/CT imaging in
children with papillary thyroid carcinoma. Pediatr Radiol.
2011;41(8):1008–1012.
77. Kohlfuerst S, Igerc I, Lobnig M, et al. Posttherapeutic
738 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
(131)I SPECT-CT offers high diagnostic accuracy when the
findings on conventional planar imaging are inconclusive
and allows a tailored patient treatment regimen. Eur J Nucl
Med Mol Imaging. 2009;36(6):886893.
78. Maruoka Y, Abe K, Baba S, et al. Incremental diagnostic
value of SPECT/CT with 131I scintigraphy after radioio-
dine therapy in patients with well-differentiated thyroid
carcinoma. Radiology. 2012;265(3):902–909.
79. Menges M, Uder M, Kuwert T, Schmidt D. 131I
SPECT/CT in the follow-up of patients with differentiated
thyroid carcinoma. Clin Nucl Med. 2012;37(6):555–560.
80. Mustafa M, Kuwert T, Weber K, et al. Regional lymph
node involvement in T1 papillary thyroid carcinoma: a
bicentric prospective SPECT/CT study. Eur J Nucl Med
Mol Imaging. 2010;37(8):1462–1466.
81. Oh JR, Byun BH, Hong SP, et al. Comparison of
131
I
whole-body imaging,
131
I SPECT/CT, and
18
F-FDG
PET/CT in the detection of metastatic thyroid cancer. Eur
J Nucl Med Mol Imaging. 2011;38(8):1459–1468.
82. Qiu ZL, Xu YH, Song HJ, Luo QY. Localization and iden-
tification of parapharyngeal metastases from differenti-
ated thyroid carcinoma by 131I-SPECT/CT. Head Neck.
2011;33(2):171–177.
83. Qiu ZL, Xue YL, Song HJ, Luo QY. Comparison of the
diagnostic and prognostic values of 99mTc-MDP-planar
bone scintigraphy, 131I-SPECT/CT and 18F-FDG-
PET/CT for the detection of bone metastases from differ-
entiated thyroid cancer. Nucl Med Commun. 2012;33(12):
1232–1242.
84. Ruf J, Lehmkuhl L, Bertram H, et al. Impact of SPECT and
integrated low-dose CT after radioiodine therapy on the
management of patients with thyroid carcinoma. Nucl
Med Commun. 2004;25(12):1177–1182.
85. Schmidt D, Linke R, Uder M, Kuwert T. Five months’
follow-up of patients with and without iodine-positive
lymph node metastases of thyroid carcinoma as disclosed
by (131)I-SPECT/CT at the first radioablation. Eur J Nucl
Med Mol Imaging. 2010;37(4):699–705.
86. Schmidt D, Szikszai A, Linke R, Bautz W, Kuwert T. Im-
pact of 131I SPECT/spiral CT on nodal staging of differ-
entiated thyroid carcinoma at the first radioablation.
J Nucl Med. 2009;50(1):18–23.
87. Song HJ, Xue YL, Qiu ZL, Luo QY. Uncommon metas-
tases from differentiated thyroid carcinoma. Hell J Nucl
Med. 2012;15(3):233–240.
88. Song HJ, Xue YL, Xu YH, Qiu ZL, Luo QY. Rare metas-
tases of differentiated thyroid carcinoma: pictorial review.
Endocr Relat Cancer. 2011;18(5):R165–R174.
89. Spanu A, Solinas ME, Chessa F, Sanna D, Nuvoli S,
Madeddu G. 131I SPECT/CT in the follow-up of differ-
entiated thyroid carcinoma: incremental value versus pla-
nar imaging. J Nucl Med. 2009;50(2):184–190.
90. Tharp K, Israel O, Hausmann J, et al. Impact of 131I-
SPECT/CT images obtained with an integrated system in
the follow-up of patients with thyroid carcinoma. Eur
J Nucl Med Mol Imaging. 2004;31(10):1435–1442.
91. Wakabayashi H, Nakajima K, Fukuoka M, et al. Double-
phase (131)I whole body scan and (131)I SPECT-CT im-
ages in patients with differentiated thyroid cancer: their
effectiveness for accurate identification. Ann Nucl Med.
2011;25(9):609615.
92. Wang H, Fu HL, Li JN, Zou RJ, Gu ZH, Wu JC. The role
of single-photon emission computed tomography/com-
puted tomography for precise localization of metastases in
patients with differentiated thyroid cancer. Clin Imaging.
2009;33(1):49–54.
93. Wong KK, Avram AM. Posttherapy I-131 thymic uptake
demonstrated with SPECT/CT in a young girl with papil-
lary thyroid carcinoma. Thyroid. 2008;18(8):919–920.
94. Wong KK, Sisson JC, Koral KF, Frey KA, Avram AM.
Staging of differentiated thyroid carcinoma using diagnos-
tic 131I SPECT/CT. AJR Am J Roentgenol. 2010;195(3):
730–736.
95. Wong KK, Zarzhevsky N, Cahill JM, Frey KA, Avram AM.
Incremental value of diagnostic 131I SPECT/CT fusion im-
aging in the evaluation of differentiated thyroid carcinoma.
AJR Am J Roentgenol. 2008;191(6):1785–1794.
96. Wong KK, Zarzhevsky N, Cahill JM, Frey KA, Avram AM.
Hybrid SPECT-CT and PET-CT imaging of differentiated
thyroid carcinoma. Br J Radiol. 2009;82(982):860876.
97. Yamamoto Y, Nishiyama Y, Monden T, Matsumura Y,
Satoh K, Ohkawa M. Clinical usefulness of fusion of 131I
SPECT and CT images in patients with differentiated thy-
roid carcinoma. J Nucl Med. 2003;44(12):1905–1910.
98. Weber K, Berger F, Mustafa M, Reiser MF, Bartenstein P,
Haug A. SPECT/CT for staging and treatment monitoring
in oncology. Applications in differentiated thyroid cancer
and liver tumors[in German]. Radiologe. 2012;52(7):646 –
652.
99. Grewal RK, Tuttle RM, Fox J, et al. The effect of post-
therapy 131I SPECT/CT on risk classification and man-
agement of patients with differentiated thyroid cancer.
J Nucl Med. 2010;51(9):1361–1367.
100. Avram AM, Fig LM, Frey KA, Gross MD, Wong KK.
Preablation 131-I Scans With SPECT/CT in Postoperative
Thyroid Cancer Patients: What Is the Impact on Staging?
J Clin Endocrinol Metab. 2013;98(3):1163–1171.
101. Mian C, Barollo S, Pennelli G, et al. Molecular character-
istics in papillary thyroid cancers (PTCs) with no 131I up-
take. Clin Endocrinol (Oxf). 2008;68(1):108–116.
102. Agriantonis DJ, Hall L, Wilson MA. Utility of SPECT/CT
as an adjunct to planar whole body I-131 imaging: liver
metastasis from papillary thyroid cancer. Clin Nucl Med.
2009;34(4):247–248.
103. Aide N, Lehembre E, Gervais R, Bardet S. Unusual intra-
tracheal metastasis of differentiated thyroid cancer accu-
rately depicted by SPECT/CT acquisition after radioiodine
ablation. Thyroid. 2007;17(12):1305–1306.
104. Dümcke CW, Madsen JL. Usefulness of SPECT/CT in the
diagnosis of intrathoracic goiter versus metastases from
cancer of the breast. Clin Nucl Med. 2007;32(2):156 –159.
105. Macdonald W, Armstrong J. Benign struma ovarii in a
patient with invasive papillary thyroid cancer: detection
with I-131 SPECT-CT. Clin Nucl Med. 2007;32(5):380
382.
106. Qiu ZL, Luo QY. Erector spinae metastases from differ-
entiated thyroid cancer identified by I-131 SPECT/CT.
Clin Nucl Med. 2009;34(3):137–140.
107. Rachinsky I, Driedger A. Iodine-131 uptake in a menstru-
ating uterus: value of SPECT/CT in distinguishing benign
and metastatic iodine-positive lesions. Thyroid. 2007;
17(9):901–902.
doi: 10.1210/er.2013-1030 edrv.endojournals.org 739
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
108. Thust S, Fernando R, Barwick T, Mohan H, Clarke SE.
SPECT/CT identification of post-radioactive iodine treat-
ment false-positive uptake in a simple renal cyst. Thyroid.
2009;19(1):75–76.
109. von Falck C, Beer G, Gratz KF, Galanski M. Renal me-
tastases from follicular thyroid cancer on SPECT/CT. Clin
Nucl Med. 2007;32(9):751–752.
110. Zhao LX, Li L, Li FL, Zhao Z. Rectus abdominis muscle
metastasis from papillary thyroid cancer identified by
I-131 SPECT/CT. Clin Nucl Med. 2010;35(5):360–361.
111. Carlisle MR, Lu C, McDougall IR. The interpretation of
131I scans in the evaluation of thyroid cancer, with an
emphasis on false positive findings. Nucl Med Commun.
2003;24(6):715–735.
112. Mitchell G, Pratt BE, Vini L, McCready VR, Harmer CL.
False positive 131I whole body scans in thyroid cancer. Br J
Radiol. 2000;73(870):627–635.
113. Shapiro B, Rufini V, Jarwan A, et al. Artifacts, anatomical
and physiological variants, and unrelated diseases that
might cause false-positive whole-body 131-I scans in pa-
tients with thyroid cancer. Semin Nucl Med. 2000;30(2):
115–132.
114. Bizhanova A, Kopp P. Minireview: the sodium-iodide sym-
porter NIS and pendrin in iodide homeostasis of the thy-
roid. Endocrinology. 2009;150(3):1084–1090.
115. Filetti S, Bidart JM, Arturi F, Caillou B, Russo D, Schlum-
berger M. Sodium/iodide symporter: a key transport sys-
tem in thyroid cancer cell metabolism. Eur J Endocrinol.
1999;141(5):443–457.
116. Shen DH, Kloos RT, Mazzaferri EL, Jhian SM. Sodium
iodide symporter in health and disease. Thyroid. 2001;
11(5):415–425.
117. Bruno R, Giannasio P, Ronga G, et al. Sodium iodide sym-
porter expression and radioiodine distribution in extrathy-
roidal tissues. J Endocrinol Invest. 2004;27(11):1010
1014.
118. Carvalho DP, Ferreira AC. The importance of sodium/io-
dide symporter (NIS) for thyroid cancer management. Arq
Bras Endocrinol Metabol. 2007;51(5):672–682.
119. Dohán O, De la Vieja A, Paroder V, et al. The sodium/
iodide symporter (NIS): characterization, regulation, and
medical significance. Endocr Rev. 2003;24(1):48–77.
120. Glazer DI, Brown RK, Wong KK, Savas H, Gross MD,
Avram AM. SPECT/CT evaluation of unusual physiologic
radioiodine biodistributions: pearls and pitfalls in image
interpretation. Radiographics. 2013;33(2):397–418.
121. Oh JR, Ahn BC. False-positive uptake on radioiodine
whole-body scintigraphy: physiologic and pathologic vari-
ants unrelated to thyroid cancer. Am J Nucl Med Mol Im-
aging. 2012;2(3):362–385.
122. Jammah AA, Driedger A, Rachinsky I. Incidental finding of
ovarian teratoma on post-therapy scan for papillary thy-
roid cancer and impact of SPECT/CT imaging. Arq Bras
Endocrinol Metabol. 2011;55(7):490493.
123. Yuoness S, Rachinsky I, Driedger AA, Belhocine TZ. Dif-
ferentiated thyroid cancer with epiphora: detection of na-
solacrimal duct obstruction on I-131 SPECT/CT. Clin
Nucl Med. 2011;36(12):1149–1152.
124. Vercellino L, Alaoui NI, Faugeron I, et al. Lingual thyroid
imaging with
123
I SPECT/CT. Eur J Nucl Med Mol Imag-
ing. 2011;38(6):1173.
125. Mebarki M, Menemani A, Medjahedi A, et al. Radioiodine
accumulation in a giant ovarian cystadenofibroma de-
tected incidentally by 131-I whole body scans. Case Rep
Radiol. 2012;2012:295617.
126. Sakahara H, Yamashita S, Suzuki K, Imai M, Kosugi T.
Visualization of nasolacrimal drainage system after radio-
iodine therapy in patients with thyroid cancer. Ann Nucl
Med. 2007;21(9):525–527.
127. Grünwald F, Kälicke T, Feine U, et al. Fluorine-18 fluo-
rodeoxyglucose positron emission tomography in thyroid
cancer: results of a multicentre study. Eur J Nucl Med.
1999;26(12):1547–1552.
128. Grünwald F, Schomburg A, Bender H, et al. Fluorine-18
fluorodeoxyglucose positron emission tomography in the
follow-up of differentiated thyroid cancer. Eur J Nucl Med.
1996;23(3):312–319.
129. Helal BO, Merlet P, Toubert ME, et al. Clinical impact of
(18)F-FDG PET in thyroid carcinoma patients with ele-
vated thyroglobulin levels and negative (131)I scanning
results after therapy. J Nucl Med. 2001;42(10):1464
1469.
130. Hung GU, Lee KW, Liao PY, Yang LH, Yang KT. The
influence of I-131 therapy on FDG uptake in differentiated
thyroid cancer. Ann Nucl Med. 2008;22(6):481–485.
131. Iwata M, Kasagi K, Misaki T, et al. Comparison of whole-
body 18F-FDG PET, 99mTc-MIBI SPET, and post-thera-
peutic 131I-Na scintigraphy in the detection of metastatic
thyroid cancer. Eur J Nucl Med Mol Imaging. 2004;31(4):
491–498.
132. Gharib H, Papini E, Paschke R, et al. American Association
of Clinical Endocrinologists, Associazione Medici Endo-
crinologi, and European Thyroid Association medical
guidelines for clinical practice for the diagnosis and man-
agement of thyroid nodules: executive summary of recom-
mendations. J Endocrinol Invest. 2010;33(5 suppl):51–56.
133. Tuttle RM, Leboeuf R, Robbins RJ, et al. Empiric radio-
active iodine dosing regimens frequently exceed maximum
tolerated activity levels in elderly patients with thyroid can-
cer. J Nucl Med. 2006;47(10):1587–1591.
134. Kulkarni K, Van Nostrand D, Atkins F, Aiken M, Burman
K, Wartofsky L. The relative frequency in which empiric
dosages of radioiodine would potentially overtreat or un-
dertreat patients who have metastatic well-differentiated
thyroid cancer. Thyroid. 2006;16(10):1019–1023.
135. Van Nostrand D, Aiken M, Atkins F, et al. The utility of
radioiodine scans prior to iodine 131 ablation in patients
with well-differentiated thyroid cancer. Thyroid. 2009;
19(8):849855.
136. Bahn RS, Burch HB, Cooper DS, et al. Hyperthyroidism
and other causes of thyrotoxicosis: management guidelines
of the American Thyroid Association and American Asso-
ciation of Clinical Endocrinologists. Endocr Pract. 2011;
17(3):456–520.
137. Maxon HR, Thomas SR, Hertzberg VS, et al. Relation
between effective radiation dose and outcome of radioio-
dine therapy for thyroid cancer. N Engl J Med. 1983;
309(16):937–941.
138. Koral KF, Adler RS, Carey JE, Beierwaltes WH. Iodine-
131 treatment of thyroid cancer: absorbed dose calculated
from post-therapy scans. J Nucl Med. 1986;27(7):1207–
1211.
740 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
139. Sisson JC, Dewaraja YK, Wizauer EJ, Giordano TJ, Avram
AM. Thyroid carcinoma metastasis to skull with infringe-
ment of brain: treatment with radioiodine. Thyroid. 2009;
19(3):297–303.
140. Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S.
The pathologic classification of neuroendocrine tumors: a
review of nomenclature, grading, and staging systems.
Pancreas. 2010;39(6):707–712.
141. Vinik AI, Woltering EA, Warner RR, et al. NANETS con-
sensus guidelines for the diagnosis of neuroendocrine tu-
mor. Pancreas. 2010;39(6):713–734.
142. Mottaghy FM, Reske SN. Functional imaging of neuroen-
docrine tumours with PET. Pituitary. 2006;9(3):237–242.
143. Ilias I, Pacak K. A clinical overview of pheochromocyto-
mas/paragangliomas and carcinoid tumors. Nucl Med
Biol. 2008;35(suppl 1):S27–S34.
144. Pacak K, Eisenhofer G, Goldstein DS. Functional imaging
of endocrine tumors: role of positron emission tomogra-
phy. Endocr Rev. 2004;25(4):568–580.
145. Ingui CJ, Shah NP, Oates ME. Endocrine neoplasm scin-
tigraphy: added value of fusing SPECT/CT images com-
pared with traditional side-by-side analysis. Clin Nucl
Med. 2006;31(11):665–672.
146. Roach PJ, Schembri GP, Ho Shon IA, Bailey EA, Bailey DL.
SPECT/CT imaging using a spiral CT scanner for anatom-
ical localization: impact on diagnostic accuracy and re-
porter confidence in clinical practice. Nucl Med Commun.
2006;27(12):977–987.
147. Amthauer H, Denecke T, Rohlfing T, et al. Value of image
fusion using single photon emission computed tomogra-
phy with integrated low dose computed tomography in
comparison with a retrospective voxel-based method in
neuroendocrine tumours. Eur Radiol. 2005;15(7):1456
1462.
148. Apostolova I, Riethdorf S, Buchert R, et al. SPECT/CT
stabilizes the interpretation of somatostatin receptor scin-
tigraphy findings: a retrospective analysis of inter-rater
agreement. Ann Nucl Med. 2010;24(6):477–483.
149. Castaldi P, Rufini V, Treglia G, et al. Impact of 111In-
DTPA-octreotide SPECT/CT fusion images in the manage-
ment of neuroendocrine tumours. Radiol Med. 2008;
113(7):1056–1067.
150. Hillel PG, van Beek EJ, Taylor C, et al. The clinical impact
of a combined
camera/CT imaging system on somatosta-
tin receptor imaging of neuroendocrine tumours. Clin Ra-
diol. 2006;61(7):579–587.
151. Krausz Y, Keidar Z, Kogan I, et al. SPECT/CT hybrid im-
aging with 111In-pentetreotide in assessment of neuroen-
docrine tumours. Clin Endocrinol (Oxf). 2003;59(5):565–
573.
152. Perri M, Erba P, Volterrani D, et al. Octreo-SPECT/CT
imaging for accurate detection and localization of sus-
pected neuroendocrine tumors. Q J Nucl Med Mol Imag-
ing. 2008;52(4):323–333.
153. Ruf J, Steffen I, Mehl S, et al. Influence of attenuation
correction by integrated low-dose CT on somatostatin re-
ceptor SPECT. Nucl Med Commun. 2007;28(10):782–
788.
154. Wong KK, Cahill JM, Frey KA, Avram AM. Incremental
value of 111-in pentetreotide SPECT/CT fusion imaging of
neuroendocrine tumors. Acad Radiol. 2010;17(3):291–
297.
155. Wong KK, Wynn EA, Myles J, Ackermann RJ, Frey KA,
Avram AM. Comparison of single time-point [111-In] pen-
tetreotide SPECT/CT with dual time-point imaging of neu-
roendocrine tumors. Clin Nucl Med. 2011;36(1):25–31.
156. Gabriel M, Hausler F, Bale R, et al. Image fusion analysis
of (99m)Tc-HYNIC-Tyr(3)-octreotide SPECT and diag-
nostic CT using an immobilisation device with external
markers in patients with endocrine tumours. Eur J Nucl
Med Mol Imaging. 2005;32(12):1440–1451.
157. Pfannenberg AC, Eschmann SM, Horger M, et al. Benefit
of anatomical-functional image fusion in the diagnostic
work-up of neuroendocrine neoplasms. Eur J Nucl Med
Mol Imaging. 2003;30(6):835–843.
158. Sepúlveda-Méndez J, de Murphy CA, Pedraza-López M,
Murphy-Stack E, Rojas-Bautista JC, González-Treviño O.
Specificity and sensitivity of
99
mTc-EDDA/HYNIC-Tyr
3
-
octreotide (
99
mTc-TOC) for imaging neuroendocrine tu-
mors. Nucl Med Commun. 2012;33(1):69–79.
159. Sandström M, Garske-Román U, Granberg D, et al. Indi-
vidualized dosimetry of kidney and bone marrow in pa-
tients undergoing 177Lu-DOTA-octreotate treatment.
J Nucl Med. 2013;54(1):33–41.
160. Grimes J, Celler A, Birkenfeld B, et al. Patient-specific ra-
diation dosimetry of 99mTc-HYNIC-Tyr3-octreotide in
neuroendocrine tumors. J Nucl Med. 2011;52(9):1474
1481.
161. Rufini V, Castaldi P, Treglia G, et al. Nuclear medicine
procedures in the diagnosis and therapy of medullary thy-
roid carcinoma. Biomed Pharmacother. 2008;62(3):139
146.
162. Bozkurt MF, Ugur O, Banti E, Grassetto G, Rubello D.
Functional nuclear medicine imaging of medullary thyroid
cancer. Nucl Med Commun. 2008;29(11):934–942.
163. Czepczynski R, Parisella MG, Kosowicz J, et al. Soma-
tostatin receptor scintigraphy using 99mTc-EDDA/
HYNIC-TOC in patients with medullary thyroid carci-
noma. Eur J Nucl Med Mol Imaging. 2007;34(10):1635–
1645.
164. Fiore D, Rubello D, Casara D, Pelizzo MR, Franchi A,
Muzzio PC. Role of CT/111In-octreotide SPECT digital
fusion imaging in the localization of loco-regional recur-
rence of medullary thyroid carcinoma [in Italian]. Minerva
Chir. 2004;59(3):295–299.
165. De Bonilla-Damiá A1, Calvo-Morón C, De La Riva-Pérez
PA, Iglesias-Jerez R, Molina-Mora M, Castro-Montaño J.
Detection by SPECT-CT scan with
99m
Tc-(V) DMSA of
bone metastases in patient with medullary thyroid cancer
[in Spanish]. Rev Esp Med Nucl. 2011;30(6):365–367.
166. Chowdhury FU, Scarsbrook AF. The role of hybrid
SPECT-CT in oncology: current and emerging clinical ap-
plications. Clin Radiol. 2008;63(3):241–251.
167. Patel CN, Chowdhury FU, Scarsbrook AF. Clinical utility
of hybrid SPECT-CT in endocrine neoplasia. AJR Am J
Roentgenol. 2008;190(3):815–824.
168. Sarkar SD, Beierwaltes H, Ice RD, et al. A new and superior
adrenal scanning agent, NP-59. J Nucl Med. 1975;16(11):
1038–1042.
169. Kloos RT, Korobkin M, Thompson NW, Francis IR, Sha-
doi: 10.1210/er.2013-1030 edrv.endojournals.org 741
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
piro B, Gross MD. Incidentally discovered adrenal masses.
Cancer Treat Res. 1997;89:263–292.
170. Yu KC, Alexander HR, Ziessman HA, et al. Role of pre-
operative iodocholesterol scintiscanning in patients under-
going adrenalectomy for Cushing’s syndrome. Surgery.
1995;118(6):981–986; discussion 986–987.
171. White ML, Gauger PG, Doherty GM, et al. The role of
radiologic studies in the evaluation and management of
primary hyperaldosteronism. Surgery. 2008;144(6):926
933.
172. Volpe C, Enberg U, Sjögren A, et al. The role of adrenal
scintigraphy in the preoperative management of primary
aldosteronism. Scand J Surg. 2008;97(3):248–253.
173. Izaki H, Fukumori T, Takahashi M, et al. Indications for
laparoscopic adrenalectomy for non-functional adrenal
tumor with hypertension: usefulness of adrenocortical
scintigraphy. Int J Urol. 2006;13(6):677–681.
174. Bardet S, Rohmer V, Murat A, et al. 131I-6
-iodometh-
ylnorcholesterol scintigraphy: an assessment of its role in
the investigation of adrenocortical incidentalomas. Clin
Endocrinol (Oxf). 1996;44(5):587–596.
175. Kazerooni EA, Sisson JC, Shapiro B, et al. Diagnostic ac-
curacy and pitfalls of [iodine-131]6-
-iodomethyl-19-nor-
cholesterol (NP-59) imaging. J Nucl Med. 1990;31(4):
526–534.
176. Hwang I, Balingit AG, Georgitis WJ, Sisson JC, Shapiro B.
Adrenocortical SPECT using iodine-131 NP-59. J Nucl
Med. 1998;39(8):1460–1463.
177. La Cava G, Imperiale A, Olianti C, et al. SPECT semiquan-
titative analysis of adrenocortical (131)I-6
iodomethyl-
norcholesterol uptake to discriminate subclinical and pre-
clinical functioning adrenal incidentaloma. J Nucl Med.
2003;44(7):1057–1064.
178. Imperiale A, Olianti C, Mannelli M, La Cava G, Pupi A.
Tomographic evaluation of [131I] 6
-iodomethyl-norcho-
lesterol standardised uptake trend in clinically silent mono-
lateral and bilateral adrenocortical incidentalomas. Q
J Nucl Med Mol Imaging. 2005;49(3):287–296.
179. Chen YC, Wei CK, Chen PF, Tzeng JE, Chuang TL, Wang
YF. Seeking the invisible: I-131 NP-59 SPECT/CT for pri-
mary hyperaldosteronism. Kidney Int. 2009;75(6):663.
180. Chen YC, Su YC, Wei CK, et al. Diagnostic value of I-131
NP-59 SPECT/CT scintigraphy in patients with subclinical
or atypical features of primary aldosteronism. J Biomed
Biotechnol. 2011;2011:209787.
181. Chen YC, Su YC, Chiu JS, Tseng CE. Diagnosis of primary
aldosteronism in chronic kidney disease by I-131 NP-59
SPECT/CT imaging. Clin Nucl Med. 2012;37(2):e30 – e32.
182. Yen RF, Wu VC, Liu KL, et al. 131I-6
-iodomethyl-19-
norcholesterol SPECT/CT for primary aldosteronism pa-
tients with inconclusive adrenal venous sampling and CT
results. J Nucl Med. 2009;50(10):1631–1637.
183. Chen H, Sippel RS, O’Dorisio MS, Vinik AI, Lloyd RV,
Pacak K. The North American Neuroendocrine Tumor So-
ciety consensus guideline for the diagnosis and manage-
ment of neuroendocrine tumors: pheochromocytoma,
paraganglioma, and medullary thyroid cancer. Pancreas.
2010;39(6):775–783.
184. Havekes B, King K, Lai EW, Romijn JA, Corssmit EP,
Pacak K. New imaging approaches to phaeochromocyto-
mas and paragangliomas. Clin Endocrinol (Oxf). 2010;
72(2):137–145.
185. Jacobson AF, Deng H, Lombard J, Lessig HJ, Black RR.
123I-meta-iodobenzylguanidine scintigraphy for the de-
tection of neuroblastoma and pheochromocytoma: results
of a meta-analysis. J Clin Endocrinol Metab. 2010;95(6):
2596–2606.
186. Duet M, Sauvaget E, Pételle B, et al. Clinical impact of
somatostatin receptor scintigraphy in the management of
paragangliomas of the head and neck. J Nucl Med. 2003;
44(11):1767–1774.
187. Muros MA, Llamas-Elvira JM, Rodríguez A, et al. 111In-
pentetreotide scintigraphy is superior to 123I-MIBG scin-
tigraphy in the diagnosis and location of chemodectoma.
Nucl Med Commun. 1998;19(8):735–742.
188. Telischi FF, Bustillo A, Whiteman ML, et al. Octreotide
scintigraphy for the detection of paragangliomas. Otolar-
yngol Head Neck Surg. 2000;122(3):358–362.
189. Whiteman ML, Serafini AN, Telischi FF, Civantos FJ, Fal-
cone S. 111In octreotide scintigraphy in the evaluation of
head and neck lesions. AJNR Am J Neuroradiol. 1997;
18(6):1073–1080.
190. Mihai R, Gleeson F, Roskell D, Parker A, Sadler G. Routine
preoperative (123)I-MIBG scintigraphy for patients with
phaeochromocytoma is not necessary. Langenbecks Arch
Surg. 2008;393(5):725–727.
191. Taïeb D, Sebag F, Hubbard JG, Mundler O, Henry JF,
Conte-Devolx B. Does iodine-131 meta-iodobenzylguani-
dine (MIBG) scintigraphy have an impact on the manage-
ment of sporadic and familial phaeochromocytoma? Clin
Endocrinol Oxf. 2004;61(1):102–108.
192. Meyer-Rochow GY, Schembri GP, Benn DE, et al. The
utility of metaiodobenzylguanidine single photon emission
computed tomography/computed tomography (MIBG
SPECT/CT) for the diagnosis of pheochromocytoma. Ann
Surg Oncol. 2010;17(2):392–400.
193. Núñez R, Erwin WD, Wendt RE 3rd, et al. Acquisition
parameters for oncologic imaging with a new SPECT/mul-
tislice CT scanner. Mol Imaging Biol. 2010;12(2):110
138.
194. Acharya J, Chang PT, Gerard P. Abnormal MIBG uptake
in a neuroblastoma patient with right upper lobe atelecta-
sis. Pediatr Radiol. 2012;42(10):1259–1262.
195. Aide N, Reznik Y, Icard P, Franson T, Bardet S. Paraneo-
plastic ACTH secretion: bronchial carcinoid overlooked
by planar indium-111 pentetreotide scintigraphy and ac-
curately localized by SPECT/CT acquisition. Clin Nucl
Med. 2007;32(5):398400.
196. Canbaz F, Basoglu T, Ersoy D, Dabak N. Interesting im-
age. Rapid verification of unusual tracer uptake patterns
by means of SPECT/CT: a case of unexpected 111indium
pentetreotide accumulation in a splenic variant. Clin Nucl
Med. 2010;35(11):904–906.
197. Caobelli F, Bertagna F, Panarotto MB, Paghera B, Giub-
bini R. An unusual muscular metastasis in a patient af-
fected by ileal carcinoid imaged with a
111
In-pentetreotide
SPECT/CT scan and confirmed by biopsy. Clin Nucl Med.
2011;36(8):696697.
198. Patel M, Santos P, Jong I, Nandurkar D, McKay J. Malig-
nant pheochromocytoma metastasis to the breast shown
742 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
on I-123 MIBG scan. Clin Nucl Med. 2010;35(10):816
817.
199. Singer J, Koch CA, Kassahun W, et al. A patient with a large
recurrent pheochromocytoma demonstrating the pitfalls
of diagnosis. Nat Rev Endocrinol. 2011;7(12):749–755.
200. Strobel K, Burger C, Schneider P, Weber M, Hany TF.
MIBG-SPECT/CT-angiography with 3-D reconstruction
of an extra-adrenal phaeochromocytoma with dissection
of an aortic aneurysm. Eur J Nucl Med Mol Imaging. 2007;
34(1):150.
201. Udall D, Cho SY. Congenital agenesis of right parotid
gland confounds MIBG scan interpretation in craniocer-
vical neuroblastoma. Clin Nucl Med. 2011;36(11):e162–
e164.
202. Rozovsky K, Koplewitz BZ, Krausz Y, et al. Added value
of SPECT/CT for correlation of MIBG scintigraphy and
diagnostic CT in neuroblastoma and pheochromocytoma.
AJR Am J Roentgenol. 2008;190(4):1085–1090.
203. Fukuoka M, Taki J, Mochizuki T, Kinuya S. Comparison
of diagnostic value of I-123 MIBG and high-dose I-131
MIBG scintigraphy including incremental value of
SPECT/CT over planar image in patients with malignant
pheochromocytoma/paraganglioma and neuroblastoma.
Clin Nucl Med. 2011;36(1):1–7.
204. Derlin T, Busch JD, Wisotzki C, et al. Intraindividual com-
parison of 123I-mIBG SPECT/MRI, 123I-mIBG SPECT/
CT, and MRI for the detection of adrenal pheochromocy-
toma in patients with elevated urine or plasma
catecholamines. Clin Nucl Med. 2013;38(1):e1–e6.
205. Rufini V, Treglia G, Castaldi P, et al. Comparison of 123I-
MIBG SPECT-CT and 18F-DOPA PET-CT in the evalua-
tion of patients with known or suspected recurrent para-
ganglioma. Nucl Med Commun. 2011;32(7):575–582.
206. Franzius C, Hermann K, Weckesser M, et al. Whole-body
PET/CT with 11C-meta-hydroxyephedrine in tumors of
the sympathetic nervous system: feasibility study and com-
parison with 123I-MIBG SPECT/CT. J Nucl Med. 2006;
47(10):1635–1642.
207. Howman-Giles R, Shaw PJ, Uren RF, Chung DK. Neuro-
blastoma and other neuroendocrine tumors. Semin Nucl
Med. 2007;37(4):286–302.
208. Tang HR, Da Silva AJ, Matthay KK, et al. Neuroblastoma
imaging using a combined CT scanner-scintillation camera
and 131I-MIBG. J Nucl Med. 2001;42(2):237–247.
209. Andersen JB, Mortensen J, Bech BH, Højgaard L, Borg-
wardt L. First experiences from Copenhagen with paedi-
atric single photon emission computed tomography/com-
puted tomography. Nucl Med Commun. 2011;32(5):
356–362.
210. Biermann M, Schwarzlmüller T, Fasmer KE, Reitan BC,
Johnsen B, Rosendahl K. Is there a role for PET-CT and
SPECT-CT in pediatric oncology? Acta Radiol. 2013;
54(9):1037–1045.
211. Taubman ML, Goldfarb M, Lew JI. Role of SPECT and
SPECT/CT in the surgical treatment of primary hyperpara-
thyroidism. Int J Mol Imaging. 2011;2011:141593.
212. Eslamy HK, Ziessman HA. Parathyroid scintigraphy in
patients with primary hyperparathyroidism: 99mTc sesta-
mibi SPECT and SPECT/CT. Radiographics. 2008;28(5):
1461–1476.
213. Hindié E, Ugur O, Fuster D, et al. 2009 EANM parathy-
roid guidelines. Eur J Nucl Med Mol Imaging. 2009;36(7):
1201–1216.
214. Greenspan BS, Dillehay G, Intenzo C, et al. SNM practice
guideline for parathyroid scintigraphy 4.0. J Nucl Med
Technol. 2012;40(2):111–118.
215. Palestro CJ, Tomas MB, Tronco GG. Radionuclide imag-
ing of the parathyroid glands. Semin Nucl Med. 2005;
35(4):266–276.
216. Chien D, Jacene H. Imaging of parathyroid glands. Oto-
laryngol Clin North Am. 2010;43(2):399415, x.
217. Berber E, Parikh RT, Ballem N, Garner CN, Milas M,
Siperstein AE. Factors contributing to negative parathy-
roid localization: an analysis of 1000 patients. Surgery.
2008;144(1):74–79.
218. Kunstman JW, Kirsch JD, Mahajan A, Udelsman R. Clin-
ical review: parathyroid localization and implications for
clinical management. J Clin Endocrinol Metab. 2013;
98(3):902–912.
219. Okuda I, Nakajima Y, Miura D, Maruno H, Kohno T,
Hirata K. Diagnostic localization of ectopic parathyroid
lesions: developmental consideration. Jpn J Radiol. 2010;
28(10):707–713.
220. Ali L, Loutfi I, Biswas G, Hadi N, Girgis T. Improved
delineation of parathyroid lesions in patients with chronic
renal failure using magnified pinhole imaging. J Nucl Med
Technol. 2011;39(1):35–39.
221. Thomas DL, Bartel T, Menda Y, Howe J, Graham MM,
Juweid ME. Single photon emission computed tomogra-
phy (SPECT) should be routinely performed for the detec-
tion of parathyroid abnormalities utilizing technetium-
99m sestamibi parathyroid scintigraphy. Clin Nucl Med.
2009;34(10):651–655.
222. Smith JR, Oates ME. Radionuclide imaging of the para-
thyroid glands: patterns, pearls, and pitfalls. Radiograph-
ics. 2004;24(4):1101–1115.
223. Martínez-Rodríguez I, Banzo I, Quirce R, et al. Early pla-
nar and early SPECT Tc-99m sestamibi imaging: can it
replace the dual-phase technique for the localization of
parathyroid adenomas by omitting the delayed phase? Clin
Nucl Med. 2011;36(9):749–753.
224. Swanson TW, Chan SK, Jones SJ, et al. Determinants of
Tc-99m sestamibi SPECT scan sensitivity in primary hy-
perparathyroidism. Am J Surg. 2010;199(5):614620.
225. Ruda JM, Hollenbeak CS, Stack BC Jr. A systematic review
of the diagnosis and treatment of primary hyperparathy-
roidism from 1995 to 2003. Otolaryngol Head Neck Surg.
2005;132(3):359–372.
226. Witteveen JE, Kievit J, Stokkel MP, Morreau H, Romijn
JA, Hamdy NA. Limitations of Tc99m-MIBI-SPECT im-
aging scans in persistent primary hyperparathyroidism.
World J Surg. 2011;35(1):128–139.
227. Cook MI, Qureshi YA, Todd CE, Cummins RS. An un-
usual ectopic location of a parathyroid carcinoma arising
within the thyroid gland. J Clin Endocrinol Metab. 2012;
97(6):1829–1833.
228. Loftus KA, Anderson S, Mulloy AL, Terris DJ. Value of
sestamibi scans in tertiary hyperparathyroidism. Laryngo-
scope. 2007;117(12):2135–2138.
229. Elaraj DM, Sippel RS, Lindsay S, et al. Are additional lo-
calization studies and referral indicated for patients with
doi: 10.1210/er.2013-1030 edrv.endojournals.org 743
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
primary hyperparathyroidism who have negative sesta-
mibi scan results? Arch Surg. 2010;145(6):578–581.
230. Rubello D, Casara D, Fiore D, Muzzio P, Zonzin G, Sha-
piro B. An ectopic mediastinal parathyroid adenoma ac-
curately located by a single-day imaging protocol of Tc-
99m pertechnetate-MIBI subtraction scintigraphy and
MIBI-SPECT-computed tomographic image fusion. Clin
Nucl Med. 2002;27(3):186–190.
231. Kaczirek K, Prager G, Kienast O, et al. Combined trans-
mission and (99m)Tc-sestamibi emission tomography for
localization of mediastinal parathyroid glands. Nuklear-
medizin. 2003;42(5):220–223.
232. Ciappuccini R, Morera J, Pascal P, et al. Dual-phase
99mTc sestamibi scintigraphy with neck and thorax
SPECT/CT in primary hyperparathyroidism: a single-in-
stitution experience. Clin Nucl Med. 2012;37(3):223–228.
233. Gayed IW, Kim EE, Broussard WF, et al. The value of
99mTc-sestamibi SPECT/CT over conventional SPECT in
the evaluation of parathyroid adenomas or hyperplasia.
J Nucl Med. 2005;46(2):248–252.
234. Harris L, Yoo J, Driedger A, et al. Accuracy of technetium-
99m SPECT-CT hybrid images in predicting the precise
intraoperative anatomical location of parathyroid adeno-
mas. Head Neck. 2008;30(4):509–517.
235. Kim YI, Jung YH, Hwang KT, Lee HY. Efficacy of
99
mTc-
sestamibi SPECT/CT for minimally invasive parathyroid-
ectomy: comparative study with
99
mTc-sestamibi scintig-
raphy, SPECT, US and CT. Ann Nucl Med. 2012;26(10):
804810.
236. Krausz Y, Bettman L, Guralnik L, et al. Technetium-99m-
MIBI SPECT/CT in primary hyperparathyroidism. World
J Surg. 2006;30(1):7683.
237. Lavely WC, Goetze S, Friedman KP, et al. Comparison of
SPECT/CT, SPECT, and planar imaging with single- and
dual-phase (99m)Tc-sestamibi parathyroid scintigraphy.
J Nucl Med. 2007;48(7):1084–1089.
238. Neumann DR, Obuchowski NA, Difilippo FP. Preopera-
tive 123I/99mTc-sestamibi subtraction SPECT and
SPECT/CT in primary hyperparathyroidism. J Nucl Med.
2008;49(12):2012–2017.
239. Oksüz MO, Dittmann H, Wicke C, et al. Accuracy of para-
thyroid imaging: a comparison of planar scintigraphy,
SPECT, SPECT-CT, and C-11 methionine PET for the de-
tection of parathyroid adenomas and glandular hyperpla-
sia. Diagn Interv Radiol. 2011;17(4):297–307.
240. Pata G, Casella C, Besuzio S, Mittempergher F, Salerni B.
Clinical appraisal of 99m technetium-sestamibi
SPECT/CT compared to conventional SPECT in patients
with primary hyperparathyroidism and concomitant nod-
ular goiter. Thyroid. 2010;20(10):1121–1127.
241. Pata G, Casella C, Magri GC, et al. Financial and clinical
implications of low-energy CT combined with 99m Tech-
netium-sestamibi SPECT for primary hyperparathyroid-
ism. Ann Surg Oncol. 2011;18(9):2555–2563.
242. Patel CN, Salahudeen HM, Lansdown M, Scarsbrook AF.
Clinical utility of ultrasound and 99mTc sestamibi
SPECT/CT for preoperative localization of parathyroid
adenoma in patients with primary hyperparathyroidism.
Clin Radiol. 2010;65(4):278–287.
243. Serra A, Bolasco P, Satta L, Nicolosi A, Uccheddu A, Piga
M. Role of SPECT/CT in the preoperative assessment of
hyperparathyroid patients. Radiol Med. 2006;111(7):
999–1008.
244. Shafiei B, Hoseinzadeh S, Fotouhi F, et al. Preoperative
99mTc-sestamibi scintigraphy in patients with primary hy-
perparathyroidism and concomitant nodular goiter: com-
parison of SPECT-CT, SPECT, and planar imaging. Nucl
Med Commun. 2012;33(10):1070–1076.
245. Sharma J, Mazzaglia P, Milas M, et al. Radionuclide im-
aging for hyperparathyroidism (HPT): which is the best
technetium-99m sestamibi modality? Surgery. 2006;
140(6):856863; discussion 863–865.
246. Wimmer G, Profanter C, Kovacs P, et al. CT-MIBI-SPECT
image fusion predicts multiglandular disease in hyperpara-
thyroidism. Langenbecks Arch Surg. 2010;395(1):73–80.
247. Prommegger R, Wimmer G, Profanter C, et al. Virtual neck
exploration: a new method for localizing abnormal para-
thyroid glands. Ann Surg. 2009;250(5):761–765.
248. Wimmer G, Bale R, Kovacs P, et al. Virtual neck explora-
tion in patients with hyperparathyroidism and former cer-
vical operations. Langenbecks Arch Surg. 2008;393(5):
687–692.
249. Profanter C, Wetscher GJ, Gabriel M, et al. CT-MIBI im-
age fusion: a new preoperative localization technique for
primary, recurrent, and persistent hyperparathyroidism.
Surgery. 2004;135(2):157–162.
250. Levine DS, Wiseman SM. Fusion imaging for parathyroid
localization in primary hyperparathyroidism. Expert Rev
Anticancer Ther. 2010;10(3):353–363.
251. Baird CW, Parker M, Shah N, McKay C. Minimally in-
vasive resection of mediastinal parathyroid adenoma using
SPECT/CT and intact PTH monitoring. Eur J Pediatr Surg.
2011;21(6):399400.
252. Gouveia S, Rodrigues D, Barros L, et al. Persistent primary
hyperparathyroidism: an uncommon location for an ecto-
pic gland–case report and review. Arq Bras Endocrinol
Metabol. 2012;56(6):393–403.
253. Li L, Chen L, Yang Y, et al. Giant anterior mediastinal
parathyroid adenoma. Clin Nucl Med. 2012;37(9):889
891.
254. Radan L, Gorenberg M. The value of SPECT/CT 99mTc-
sestamibi scintigraphy in the diagnosis of ectopic parathy-
roid adenoma. Isr Med Assoc J. 2009;11(7):448.
255. Akram K, Parker JA, Donohoe K, Kolodny G. Role of
single photon emission computed tomography/computed
tomography in localization of ectopic parathyroid adeno-
ma: a pictorial case series and review of the current liter-
ature. Clin Nucl Med. 2009;34(8):500–502.
256. Levine DS, Belzberg AS, Wiseman SM. Hybrid SPECT/CT
imaging for primary hyperparathyroidism: case reports
and pictorial review. Clin Nucl Med. 2009;34(11):779
784.
257. Papathanassiou D, Flament JB, Pochart JM, et al.
SPECT/CT in localization of parathyroid adenoma or hy-
perplasia in patients with previous neck surgery. Clin Nucl
Med. 2008;33(6):394–397.
258. Vaz A, Griffiths M. Parathyroid imaging and localization
using SPECT/CT: initial results. J Nucl Med Technol.
2011;39(3):195–200.
259. Chakraborty D, Mittal BR, Harisankar CN, Bhattacharya
A, Bhadada S. Spectrum of single photon emission com-
puted tomography/computed tomography findings in pa-
744 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
tients with parathyroid adenomas. Indian J Nucl Med.
2011;26(1):52–55.
260. Kirk J, Au-Yong I, Ganatra R. Multimodality imaging of a
retrosternal parathyroid adenoma with multiple brown tu-
mors. Clin Nucl Med. 2009;34(9):555–559.
261. Meng Z, Zhu M, He Q, et al. Clinical implications of
brown tumor uptake in whole-body 99mTc-sestamibi
scans for primary hyperparathyroidism. Nucl Med Com-
mun. 2011;32(8):708–715.
262. Mori H, Okada Y, Arao T, Shimaziri S, Tanaka Y. A case
of multiple brown tumors with primary hyperparathyroid-
ism. J Bone Miner Metab. 2013;31(1):123–127.
263. Treglia G, Dambra DP, Bruno I, Mulè A, Giordano A.
Costal brown tumor detected by dual-phase parathyroid
imaging and SPECT-CT in primary hyperparathyroidism.
Clin Nucl Med. 2008;33(3):193–195.
264. Belhocine T, Shastry A, Driedger A, Urbain JL. Detection
of 99mTc-sestamibi uptake in brown adipose tissue with
SPECT-CT. Eur J Nucl Med Mol Imaging. 2007;34(1):
149.
265. Goetze S, Lavely WC, Ziessman HA, Wahl RL. Visualiza-
tion of brown adipose tissue with 99mTc-methoxyisobu-
tylisonitrile on SPECT/CT. J Nucl Med. 2008;49(5):752–
756.
266. Wong KK, Brown RK, Avram AM. Potential false positive
Tc-99m sestamibi parathyroid study due to uptake in
brown adipose tissue. Clin Nucl Med. 2008;33(5):346
348.
267. Silverberg SJ, Bilezikian JP. The diagnosis and manage-
ment of asymptomatic primary hyperparathyroidism. Nat
Clin Pract Endocrinol Metab. 2006;2(9):494–503.
268. Caldarella C, Treglia G, Pontecorvi A, Giordano A. Diag-
nostic performance of planar scintigraphy using 99mTc-
MIBI in patients with secondary hyperparathyroidism: a
meta-analysis. Ann Nucl Med. 2012;26(10):794803.
269. Cheung K, Wang TS, Farrokhyar F, Roman SA, Sosa JA.
A meta-analysis of preoperative localization techniques for
patients with primary hyperparathyroidism. Ann Surg On-
col. 2012;19(2):577–583.
270. Levy JM, Kandil E, Yau LC, Cuda JD, Sheth SN, Tufano
RP. Can ultrasound be used as the primary screening mo-
dality for the localization of parathyroid disease prior to
surgery for primary hyperparathyroidism? A review of 440
cases. ORL J Otorhinolaryngol Relat Spec. 2011;73(2):
116–120.
271. Untch BR, Adam MA, Scheri RP, et al. Surgeon-performed
ultrasound is superior to 99Tc-sestamibi scanning to lo-
calize parathyroid adenomas in patients with primary hy-
perparathyroidism: results in 516 patients over 10 years.
J Am Coll Surg. 2011;212(4):522–529; discussion 529
531.
272. Kutler DI, Moquete R, Kazam E, Kuhel WI. Parathyroid
localization with modified 4D-computed tomography and
ultrasonography for patients with primary hyperparathy-
roidism. Laryngoscope. 2011;121(6):1219–1224.
273. Dy BM, Richards ML, Vazquez BJ, Thompson GB, Farley
DR, Grant CS. Primary hyperparathyroidism and negative
Tc99 sestamibi imaging: to operate or not? Ann Surg On-
col. 2012;19(7):2272–2278.
274. Hoda NE, Phillips P, Ahmed N. Recommendations after
non-localizing sestamibi and ultrasound scans in primary
hyperparathyroid disease: order more scans or explore sur-
gically? J Miss State Med Assoc. 2013;54(2):3641.
275. Seehofer D, Steinmüller T, Rayes N, et al. Parathyroid hor-
mone venous sampling before reoperative surgery in renal
hyperparathyroidism: comparison with noninvasive local-
ization procedures and review of the literature. Arch Surg.
2004;139(12):1331–1338.
276. Caldarella C, Treglia G, Isgrò MA, Giordano A. Diagnos-
tic performance of positron emission tomography using
11C-methionine in patients with suspected parathyroid
adenoma: a meta-analysis. Endocrine. 2013;43(1):78 –83.
277. Zhen L, Li H, Liu X, Ge BH, Yan J, Yang J. The application
of SPECT/CT for preoperative planning in patients with
secondary hyperparathyroidism. Nucl Med Commun.
2013;34(5):439444.
278. Tardin L, Prats E, Andrés A, et al. Ectopic parathyroid
adenoma: scintigraphic detection and radioguided sur-
gery[in Spanish]. Rev Esp Med Nucl. 2011;30(1):19–23.
279. Hassler S, Ben-Sellem D, Hubele F, Constantinesco A,
Goetz C. Dual-isotope 99mTc-MIBI/123I parathyroid
scintigraphy in primary hyperparathyroidism: comparison
of subtraction SPECT/CT and pinhole planar scan. Clin
Nucl Med. 2014;39(1):32–36.
280. Stokkel MP, Handkiewicz Junak D, Lassmann M, Dietlein
M, Luster M. EANM procedure guidelines for therapy of
benign thyroid disease. Eur J Nucl Med Mol Imaging.
2010;37(11):2218–2228.
281. Schalin-Jäntti C, Ryhänen E, Heiskanen I, et al. Planar
scintigraphy with 123I/99mTc-sestamibi, 99mTc-sesta-
mibi SPECT/CT, 11C-methionine PET/CT, or selective ve-
nous sampling before reoperation of primary hyperpara-
thyroidism? J Nucl Med. 2013;54(5):739–747.
282. Zeiger MA, Thompson GB, Duh QY, et al. American As-
sociation of Clinical Endocrinologists and American As-
sociation of Endocrine Surgeons Medical Guidelines for
the Management of Adrenal Incidentalomas: executive
summary of recommendations. Endocr Pract. 2009;15(5):
450453.
283. Harisankar CN, Preethi GR, George M. Hybrid
SPECT/CT evaluation of dual ectopia of thyroid in the
absence of orthotopic thyroid gland. Clin Nucl Med. 2012;
37(6):602–603.
284. Harisankar CN, Preethi GR, Chungath BB. Hybrid
SPECT/CT evaluation of Marine-Lenhart syndrome. Clin
Nucl Med. 2013;38(2):e89e90.
285. Listewnik MH, Birkenfeld B, Piwowarska-Bilska H, et al.
The application of SPECT/CT scintigraphy with MIBI-
Tc99(m) in the diagnosis of thyroid nodules - a preliminary
report. Endokrynol Pol. 2010;61(5):422–426.
286. Tuncel M, Akdemir E. 99mTc-MDP uptake in thyroid
nodule: contribution of SPECT-CT and ultrasonography.
Rev Esp Med Nucl Imagen Mol. 2012;31(1):49–50.
287. Patel Z, Johnson L. Iodine 131 ablation of an obstructive
lingual thyroid. J Radiol Case Rep. 2009;3(2):3–6.
288. Richard-Fiardo P, Franken PR, Lamit A, et al. Normali-
sation to blood activity is required for the accurate quan-
tification of Na/I symporter ectopic expression by
SPECT/CT in individual subjects. PLoS One. 2012;7(3):
e34086.
289. Zhao R, Wang J, Deng J, Yang W. Efficacy of (99m)Tc-
EDDA/HYNIC-TOC SPECT/CT scintigraphy in Graves’
doi: 10.1210/er.2013-1030 edrv.endojournals.org 745
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
ophthalmopathy. Am J Nucl Med Mol Imaging. 2012;
2(2):242–247.
290. Galt JR, Halkar RK, Evans CO, et al. In vivo assay of folate
receptors in nonfunctional pituitary adenomas with
99mTc-folate SPECT/CT. J Nucl Med. 2010;51(11):
1716–1723.
291. Delbeke D, Coleman RE, Guiberteau MJ, et al. Procedure
guideline for SPECT/CT imaging 1.0. J Nucl Med. 2006;
47(7):1227–1234.
292. Dietlein M, Knapp WH, Lauterbach KW, Schicha H. Eco-
nomic evaluation studies in nuclear medicine: the need for
standardization. Eur J Nucl Med. 1999;26(6):663–680.
293. Siegel JE, Weinstein MC, Russell LB, Gold MR. Recom-
mendations for reporting cost-effectiveness analyses. Panel
on Cost-Effectiveness in Health and Medicine. JAMA.
1996;276(16):1339–1341.
294. Brockhuis B, Lass P, Popowski P, Scheffler J. An introduc-
tion to economic analysis in medicine–the basics of meth-
odology and chosen terms. Examples of results of evalua-
tion in nuclear medicine. Nucl Med Rev Cent East Eur.
2002;5(1):55–59.
295. Franklyn JA. What is the role of radioiodine uptake mea-
surement and thyroid scintigraphy in the diagnosis and
management of hyperthyroidism? Clin Endocrinol (Oxf).
2010;72(1):11–12.
296. Verburg FA, Behrendt FF, Mottaghy FM, Heinzel A. Cost-
effectiveness of (99m)Tc-MIBI in the evaluation of thyroid
nodules for malignancy: a new lease of life for an old ra-
diopharmaceutical? Eur J Nucl Med Mol Imaging. 2014;
41(1):102–104.
297. Wale A, Miles KA, Young B, et al. Combined (99m)Tc-
methoxyisobutylisonitrile scintigraphy and fine-needle as-
piration cytology offers an accurate and potentially cost-
effective investigative strategy for the assessment of
solitary or dominant thyroid nodules. Eur J Nucl Med Mol
Imaging. 2014;41(1):105–115.
298. Kwekkeboom DJ, Lamberts SW, Habbema JD, Krenning
EP. Cost-effectiveness analysis of somatostatin receptor
scintigraphy. J Nucl Med. 1996;37(6):886892.
299. Dimitroulopoulos D, Xynopoulos D, Tsamakidis K, et al.
Scintigraphic detection of carcinoid tumors with a cost
effectiveness analysis. World J Gastroenterol. 2004;
10(24):3628–3633.
300. Dwamena BA, Kloos RT, Fendrick AM, et al. Diagnostic
evaluation of the adrenal incidentaloma: decision and cost-
effectiveness analyses. J Nucl Med. 1998;39(4):707–712.
301. Lumachi F, Basso SM, Borsato S, et al. Role and cost-
effectiveness of adrenal imaging and image-guided FNA
cytology in the management of incidentally discovered ad-
renal tumours. Anticancer Res. 2005;25(6C):4559 –4562.
302. Pathirana AA, Vinjamuri S, Byrne C, Ghaneh P, Vora J,
Poston GJ. (131)I-MIBG radionuclide therapy is safe and
cost-effective in the control of symptoms of the carcinoid
syndrome. Eur J Surg Oncol. 2001;27(4):404408.
303. Ruda J, Stack BC Jr, Hollenbeak CS. The cost-effectiveness
of sestamibi scanning compared to bilateral neck explora-
tion for the treatment of primary hyperparathyroidism.
Otolaryngol Clin North Am. 2004;37(4):855–870, x-xi.
304. Wei JP, Burke GJ. Analysis of savings in operative time for
primary hyperparathyroidism using localization with tech-
netium 99m sestamibi scan. Am J Surg. 1995;170(5):488 –
491.
305. Fahy BN, Bold RJ, Beckett L, Schneider PD. Modern para-
thyroid surgery: a cost-benefit analysis of localizing strat-
egies. Arch Surg. 2002;137(8):917–922; discussion 922–
923.
306. Judson BL, Shaha AR. Nuclear imaging and minimally
invasive surgery in the management of hyperparathyroid-
ism. J Nucl Med. 2008;49(11):1813–1818.
307. Lubitz CC, Stephen AE, Hodin RA, Pandharipande P. Pre-
operative localization strategies for primary hyperparathy-
roidism: an economic analysis. Ann Surg Oncol. 2012;
19(13):4202–4209.
308. Madorin CA, Owen R, Coakley B, et al. Comparison of
radiation exposure and cost between dynamic computed
tomography and sestamibi scintigraphy for preoperative
localization of parathyroid lesions. JAMA Surg. 2013;
148(6):500–503.
309. Silberstein EB, Alavi A, Balon HR, et al. The SNMMI
practice guideline for therapy of thyroid disease with 131I
3.0. J Nucl Med. 2012;53(10):1633–1651.
310. Luster M, Clarke SE, Dietlein M, et al. Guidelines for ra-
dioiodine therapy of differentiated thyroid cancer. Eur
J Nucl Med Mol Imaging. 2008;35(10):1941–1959.
311. Cobin RH, Gharib H, Bergman DA, et al. AACE/AAES
medical/surgical guidelines for clinical practice: manage-
ment of thyroid carcinoma. American Association of Clin-
ical Endocrinologists. American College of Endocrinol-
ogy. Endocr Pract. 2001;7(3):202–220.
312. Bombardieri E, Ambrosini V, Aktolun C, et al. 111In-pen-
tetreotide scintigraphy: procedure guidelines for tumour
imaging. Eur J Nucl Med Mol Imaging. 2010;37(7):1441–
1448.
313. Balon HR, Brown TL, Goldsmith SJ, et al. The SNM prac-
tice guideline for somatostatin receptor scintigraphy 2.0.
J Nucl Med Technol. 2011;39(4):317–324.
314. Bombardieri E, Giammarile F, Aktolun C, et al. 131I/123I-
metaiodobenzylguanidine (mIBG) scintigraphy: proce-
dure guidelines for tumour imaging. Eur J Nucl Med Mol
Imaging. 2010;37(12):2436–2446.
315. Taïeb D, Timmers HJ, Hindié E, et al. EANM 2012 guide-
lines for radionuclide imaging of phaeochromocytoma and
paraganglioma. Eur J Nucl Med Mol Imaging. 2012;
39(12):1977–1995.
316. Bodei L, Mueller-Brand J, Baum RP, et al. The joint IAEA,
EANM, and SNMMI practical guidance on peptide recep-
tor radionuclide therapy (PRRNT) in neuroendocrine tu-
mours. Eur J Nucl Med Mol Imaging. 2013;40(5):800
816.
317. Gharib H, Papini E, Paschke R. Thyroid nodules: a review
of current guidelines, practices, and prospects. Eur J En-
docrinol. 2008;159(5):493–505.
318. Wong KK, Komissarova M, Avram AM, Fig LM, Gross
MD. Adrenal cortical imaging with I-131 NP-59 SPECT-
CT. Clin Nucl Med. 2010;35(11):865–869.
746 Wong et al Endocrine SPECT/CT Endocrine Reviews, October 2014, 35(5):717–746
The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 27 August 2015. at 02:50 For personal use only. No other uses without permission. . All rights reserved.
... Somatostatin receptors are over expressed in MTC and thus allows the use of radiolabeled somatostatin analogues for scintigraphic imaging before and after treatment for proper staging and follow-up of these patients [4,5]. Recently, SPECT-CT γ-cameras have become widely available which allows the fusion of anatomical CT and functional SPECT modalities [6,7]. SPECT-CT is used to optimize somatostatin-receptor scintigraphic protocols for MTC imaging. ...
... Fusion SPECT-CT images improve image quality, show correct topography and morphological characteristics of metastatic lesions and provide exact differential diagnosis of the physiological from abnormal uncertain "hot" spots, thus increasing diagnostic accuracy of somatostatin-receptor SPECT studies [6][7][8]. Radionuclide targeting of somatostatin receptors for internal radiation therapy provides another therapeutic approach in advanced symptomatic non-operable MTC [8,10]. ...
... Single-photon emission computed tomography (SPECT) has been widely used to diagnose various kinds of human diseases, such as myocardial diseases, endocrine disorders, and central nervous system diseases, since its invention in the 1990s [1][2][3][4]. Most of the available SPECT systems are based on the well-known Anger camera with NaI (Tl) as a scintillation material, which determines the position of an event by the centroid of the scintillation light [5]. ...
Article
Full-text available
Abstract Background The digital cadmium–zinc–telluride (CZT)-based SPECT system has many advantages, including better spatial and energy resolution. However, the impacts of different acquisition and reconstruction parameters on CZT SPECT quantification might still need to be validated. This study aimed to evaluate the impacts of acquisition parameters (the main energy window and acquisition time per frame) and reconstruction parameters (the number of iterations, subsets in iterative reconstruction, post-filter, and image correction methods) on the technetium quantification of CZT SPECT/CT. Methods A phantom (PET NEMA/IEC image quality, USA) was filled with four target-to-background (T/B) ratios (32:1, 16:1, 8:1, and 4:1) of technetium. Mean uptake values (the calculated mean concentrations for spheres) were measured to evaluate the recovery coefficient (RC) changes under different acquisition and reconstruction parameters. The corresponding standard deviations of mean uptake values were also measured to evaluate the quantification error. Image quality was evaluated using the National Electrical Manufacturers Association (NEMA) NU 2–2012 standard. Results For all T/B ratios, significant correlations were found between iterations and RCs (r = 0.62–0.96 for 1–35 iterations, r = 0.94–0.99 for 35–90 iterations) as well as between the full width at half maximum (FWHM) of the Gaussian filter and RCs (r = − 0.86 to − 1.00, all P values
... Along with this, patients must withdraw from interfering medications that may provoke (together with dexamethasone) a hypertension flair. Use of SPECT/CT with NP-59 scintigraphy allows precise localization of the functional activity to anatomic structures improving diagnostic interpretation (20). ...
Article
Incidentalomas are reported in 3%-4% of patients who undergo abdominal anatomic imaging, making adrenal mass evaluation a common occurrence. An adrenal mass can be caused by a variety of pathologies, such as benign cortical and medullary tumors, malignant tumors (primary or secondary), cysts, hyperplasia, hemorrhage, or more rarely infection/inflammation processes. Functioning tumors usually have increased hormonal production but they are less common. Regardless of their functional status, some tumors have the potential to behave aggressively. Anatomic and functional imaging together with biologic evaluation play a vital role in adrenal pathology subtyping. Most patients are initially evaluated by CT or MRI, which allows for tumor characterization (to a certain extent) and can rule out malignant behavior based on the absence of tumor growth during longitudinal follow-up. In the remaining patients for whom CT or MRI fail to characterize the pathogenesis of adrenal tumors, the use of specialized molecular imaging techniques should be performed after hormonal screening. This review emphasizes well-established and emerging nuclear medicine imaging modalities and describes their use across various clinical scenarios.
... The single photon emission computed tomography (SPECT) has been widely used to diagnose various kinds of human diseases such as myocardial diseases, endocrine disorders, central nervous system diseases, and so on since its invention in the 1990s [1][2][3][4]. Most of the available SPECT systems are based on the well-known Anger camera with NaI (Tl) as a scintillation material, which determines the position of an event by the centroid of the scintillation light [5]. ...
Preprint
Full-text available
Background Various acquisition and reconstruction parameters may affect the accuracy of the absolute SPECT quantification. However, many of the impacts of these parameters have not well been studied. This study aimed to evaluate the impacts of acquisition parameters (main energy window and acquisition time per frame) and reconstruction parameters (the number of iterations and subsets in iterative reconstruction, post-filter, and image correction methods) on the technetium quantification of the cadmium-zinc-telluride based SPECT/CT system. Methods A phantom (PET NEMA/IEC image quality, USA) was filled with a 16:1 sphere-to-background activity concentration ratio of technetium and all spheres had 132400.81Bq/ml of radioactivity. Mean uptake values (calculated mean concentrations for spheres) were measured to evaluate the recovery coefficient (RC) changes under different acquisition and reconstruction parameters. Corresponding standard deviations of mean uptake values were also measured to evaluate the quantification error. Image quality was evaluated using the National Electrical Manufacturers Association (NEMA) NU 2-2012 standard. Results For six spheres of the phantom, significant correlations were found between iterations and RCs (r=0.60~0.98 for 1~35 iterations, r=0.96~0.99 for 35~90 iterations, all P-values <0.05) as well as between the full width at half maximum (FWHM) of the Gauss filter and RCs (r=-0.90~-1.00, all P-values <0.05). 1~35 iterations had higher regression coefficients compared with those of 35~90 iterations (0.67~1.16 vs. 0.02~0.17). The AC (attenuation correction)+SC (scatter correction) +RR (resolution recovery correction) combination had more close to 100% RCs (42.42%~98.04%) with better image quality (31.52%~83.57%) than those of other correction combinations (all P-values <0.05). No significant statistical difference was found between the 15% energy window and the 20% energy window (P-value=0.061), nor between the 5 seconds/frame and 120 seconds/frame of acquisition time (P-value=0.943) in terms of RCs. Conclusions The CZT-SPECT/CT showed a good quantification accuracy of technetium. The favorable acquisition parameters may be 15% energy window and 40 seconds/frame of acquisition time. The favorable reconstruction parameters could be 35 iterations, 20 subsets, the AC+SC+RR correction combination, and FWHM 0.7mm of Gauss filter.
... Due to the weak radiation dose, Tc-99m O4 is preferred. Tc-99m O4 is a radionuclide which is reliable, easily accessible, widely used in clinical nuclear medicine applications [16][17][18][19][20][21][22][23][24][25] . ...
Article
Objective: Oxidative/nitrosative stress may be triggered by various sources, and ionizing radiation may also initiate oxidative/nitrosative stress. This is the first study; we aimed to investigate the induction of oxidative and nitrosative stress due to ionizing radiation in patients undergoing Tc99m pertechnetate thyroid scintigraphy. Method: Totally 26 patients (16 female,10 male) undergoing Tc-99m pertechnetate thyroid scintigraphy were included in this study. The patients were aged between 20 and 50 years (58.0±16.3 years). The blood samples were taken from patients 20 minutes after intravenous injection of Tc99m pertechnetate in the dose used clinically (5 miliCurie) before the patients were taken to the thyroid imaging. Control group was selected from 30 healthy subjects (15 female,15 male). The control group was aged between 17 and 72 years (57.0±14.0 years). The blood samples were taken both patients and control group for measuring antioxidant enzymes (catalase and superoxide dismutase), malondialdehyde, nitric oxide, and nitrotyrosine as oxidative/nitrosative stress biomarkers. Results: In this study, we found that activities of antioxidant enzymes increased in patients compared to control (p<0.05). Further, malondialdehyde levels as an indicator of oxidative stress were higher in patients than control group (p<0.05).The levels of nitric oxide and nitrotyrosine as nitrosative stress biomarkers also increased in patients compared to control groups (p<0.05). Conclusions: We thought that Tc-99m pertechnetate might cause an increase in reactive oxygen and nitrogen species and may cause oxidative/nitrosative damage at the cellular level. Our results indicated that the dose of Tc-99m pertechnetate given in these patients undergoing thyroid scintigraphy could be tolerable.
Chapter
Scintigraphy is a medical examination performed in gamma cameras after the detection of gamma ray emissions from radioactive isotopes. It can acquire static or dynamic planar images, including whole-body sweep scans or tridimensional images after the reconstruction of multiple-angle planar acquisitions (single-photon emission computed tomography—SPECT). After radiation emission, a few steps are necessary before imaging reconstruction including collimation to improve spatial resolution, signal amplification, and a computational positioning system. Hybrid equipment is available to simultaneously acquire SPECT images and anatomic images through a conventional computed tomography (SPECT-CT).KeywordsScintigraphyPlanarSPECTSPECT-CT
Article
Adrenal masses are a frequent finding in clinical practice. Many of them are incidentally discovered with a prevalence of 4% in patients undergoing abdominal anatomic imaging and require a differential diagnosis. Biochemical tests, evaluating hormonal production of both adrenal cortex and medulla (in particular, mineralocorticoids, glucocorticoids and catecholamines), have a primary importance in distinguishing functional or non-functional lesions. Conventional imaging techniques, in particular computerized tomography (CT) and magnetic resonance imaging (MRI), are required to differentiate between benign and malignant lesions according to their appearance (size stability, contrast enhanced CT and/or chemical shift on MRI). In selected patients, functional imaging is a non-invasive tool able to explore the metabolic pathways involved thus providing additional diagnostic information. Several single photon emission tomography (SPET) and positron emission tomography (PET) radiopharmaceuticals have been developed and are available, each of them suitable for studying specific pathological conditions. In functional masses causing hypersecreting diseases (mainly adrenal hypercortisolism, primary hyperaldosteronism and pheochromocytoma), functional imaging can lateralize the involvement and guide the therapeutic strategy in both unilateral and bilateral lesions. In non-functioning adrenal masses with inconclusive imaging findings at CT/MR, [18F]-FDG evaluation of tumor metabolism can be helpful to characterize them by distinguishing between benign nodules and primary malignant adrenal disease (mainly adrenocortical carcinoma), thus modulating the surgical approach. In oncologic patients, [18F]-FDG uptake can differentiate between benign nodule and adrenal metastasis from extra-adrenal primary malignancies.
Article
Identification of cervical metastatic lymph nodes (LN) on I-131 post-ablation whole-body planar scans (WBS) for cancer staging is crucial for patients with papillary thyroid cancer (PTC). The existing deep network is plagued by instability in finding the under-represented LN classes on highly complex WBS, where end-to-end tuned models that can account for this uncertainty region for multi-class locations are needed. Hence, as a key contribution of this study, we designed a novel leveraging segmentation network with input guidance (LSIG) end-to-end training model without pre and post-processing features that can learn ideal parameter settings depending on the quantity of multiple-object instances. To improve the co-occurrence of classes and control the false positive regions, we proposed a re-weighting negative control (RNC) mechanism that combines two key components, namely the re-weighting (Rw) term and the negative control function (NcF). This unified approximation of weighted training would leverage the network to control and learn the desired weights of true positives towards the LN region. As an end-to-end network training, we utilize a Unet-like convolution neural network (CNN) model. The performance of the LSIG is compared with the CNN-based networks, based on the ground truth (GT) mask developed using post-ablation single-photon emission computed tomography (SPECT/CT). Furthermore, the effectiveness of the two components used in the LSIG framework is evaluated on WBS datasets. Our proposed LSIG with a fully guided (Fg) LS-FgCNN model yielded a superior performance with high AUC value of 94.9%, which is 14.6% higher than the previous network for PTC.
Article
Full-text available
The accurate detection of radioactive iodine-avid lymph node (LN) metastasis on 131I post-ablation whole-body planar scans (RxWBSs) is important in tracking the progression of the metastatic lymph nodes (mLNs) of patients with papillary thyroid cancer (PTC). However, severe noise artifacts and the indiscernible location of the mLN from adjacent tissues with similar gray-scale values make clinical decisions extremely challenging. This study aims (i) to develop a multilayer fully connected deep network (MFDN) for the automatic recognition of mLNs from thyroid remnant tissue by utilizing the dataset of RxWBSs and (ii) to evaluate its diagnostic performance using post-ablation single-photon emission computed tomography. Image patches focused on the mLN and remnant tissues along with their variations of probability of pixel positions were fed as inputs to the network. With this efficient automatic approach, we achieved a high F1-score and outperformed the physician score (P < 0.001) in detecting mLNs. Competitive segmentation networks on RxWBS displayed moderate performance for the mLN but remained robust for the remnant tissue. Our results demonstrated that the generalization performance with the multiple layers by replicating signal transmission overcome the constraint of local minimum optimization, it can be suitable to localize the unstable location of mLN region on RxWBS and therefore MFDN can be useful in clinical decision-making to track mLN progression for PTC.
Article
Full-text available
The aim of this document is to provide general information about mIBG scintigraphy in cancer patients. The guidelines describe the mIBG scintigraphy protocol currently used in clinical routine, but do not include all existing procedures for neuroendocrine tumours. The guidelines should therefore not be taken as exclusive of other nuclear medicine modalities that can be used to obtain comparable results. It is important to remember that the resources and facilities available for patient care may vary from one country to another and from one medical institution to another. The present guidelines have been prepared for nuclear medicine physicians and intend to offer assistance in optimizing the diagnostic information that can currently be obtained from mIBG scintigraphy. The corresponding guidelines of the Society of Nuclear Medicine (SNM) and the Dosimetry, Therapy and Paediatric Committee of the EANM have been taken into consideration, and partially integrated into this text. The same has been done with the most relevant literature on this topic, and the final result has been discussed within a group of distinguished experts. Bombardieri E, Giammarile F, Aktolun C, Baum RP, Bischof Delaloye A, Maffioli L, Moncayo R, Mortelmans L, Pepe G, Reske SN, Castellani MR, Chiti A; European Association for Nuclear Medicine. Eur J Nucl Med Mol Imaging. 2010 Dec;37(12):2436-46. doi: 10.1007/s00259-010-1545-7. PMID: 20644928
Article
This document provides general information about somatostatin receptor scintigraphy with (111)In-pentetreotide. This guideline should not be regarded as the only approach to visualise tumours expressing somatostatin receptors or as exclusive of other nuclear medicine procedures useful to obtain comparable results. The aim of this guideline is to assist nuclear medicine physicians in recommending, performing, reporting and interpreting the results of (111)In-pentetreotide scintigraphy. Bombardieri E, Ambrosini V, Aktolun C, Baum RP, Bishof-Delaloye A, Del Vecchio S, Maffioli L, Mortelmans L, Oyen W, Pepe G, Chiti A; Oncology Committee of the EANM. Eur J Nucl Med Mol Imaging. 2010 Jul;37(7):1441-8. doi: 10.1007/s00259-010-1473-6. PMID: 20461371
Article
Aim: Although parathyroid scintigraphy using 99mTc-sestamibi is considered the best preoperative localization method for hyperfunctioning parathyroid tissue it lacks the anatomical details required for successful, minimal invasive surgery of ectopic parathyroid lesions. This study presents the role of combined SPECT/X-ray-CT imaging in a single device for localization of mediastinal parathyroid glands. Methods: 99mTc-sestamibi SPECT/ X-ray-CT was performed by gamma camera-mounted anatomical X-ray tomography (GMAXT; GE Medical systems, Millenium VG with Hawkeye) in four patients with ectopic parathyroid glands (two patients with primary, two with persistent secondary hyperparathyroidism). The device contains an X-ray tube and a set of detectors that rotate around the patient combined with a gamma camera. For comparison with GMAXT additionally high resolution computed tomography images of the neck and mediastinum were performed. Results: Correct preoperative localization was achieved. The parathyroid glands were located in the anterior mediastinum. High resolution computed tomography could not provide further details. Three patients were operated by a minimal invasive open and one patient by a transsternal approach because of concomitant aortic valve replacement. Conclusion: 99mTc-sestamibi/X-ray-CT fusion imaging in a single device can accurately localise ectopic or supernumerary mediastinal parathyroid tumours in primary and secondary hyperparathyroidism. Morbidity, radiation exposure, time, and costs are reduced by avoiding multiple diagnostic examinations and minimal invasive parathyroid surgery becomes possible.
Conference Paper
In recent years nuclear medicine has contributed to the impressive development of the knowledge of neuroendocrine tumors in terms of biology (receptor scintigraphy), pharmacology (development of new tracers), and therapy (radiometabolic therapy). At present, it is impossible to plan the management of a patient affected by a neuroendocrine tumor without performing nuclear medicine examinations. The contribution of nuclear medicine had affected and improved the management of these patients by offering various important options that are part of the modern diagnosis and treatment protocols. The clinical experience and the literature confirm that, among the wide variety of tracers and nuclear medicine modalities available today, metaiodobenzylguanidine (MIBG) and DTPA-D-Phe-octreotide (pentetreotide) are the radiopharmaceuticals of current clinical use. Several new somatostatin analogues are under investigation. Positron emission tomography (PET) supplies a range of labelled compounds to be used for the visualization of tumor biochemistry. In addition to the first routinely used PET tracer in oncology, F-18-labelled deoxyglucose (FDG), a number of radiopharmaceuticals based on different precursors such as fluorodopamine and 5-hydroxytryptophan (5-HTP) are going to gain a clinical role. Of course, the diagnosis of neuroendocrine tumors has to be based on integrated information derived from different examinations including nuclear medicine studies. The clinical presentation of neuroendocrine tumors is highly variable: sometimes they manifest typical or atypical symptoms but they may also be detected by chance during an X-ray or ultrasound examination carried out for other reasons. At disease presentation nuclear medicine modalities are sometimes able to direct physicians towards the clinical diagnosis thanks to the specificity of their imaging mechanisms. They also play a role in disease staging and restaging, patient follow-up and treatment monitoring. In addition, the biological characterisation of neuroendocrine tissues (receptor status, glucose metabolism, differentiation, etc.) allows the interpretation of radiopharmaceutical uptake as a prognostic parameter and sometimes as a predictor of the response to treatment.
Article
The parathyroid glands are located posterior to the upper and lower poles of the thyroid and are derived from the third and fourth pharyngeal pouches. Usually there are only 2 superior glands, whereas only 40% of patients have their inferior glands located near the inferior thyroid poles. Ectopic locations include the carotid sheath, anterior mediastinum, retropharynx, or intrathyroidal locations. Single photon emission computed tomography/computed tomography (SPECT/CT) offers the advantage of combining function and anatomy for exact localization of ectopic parathyroid adenomas. In this pictorial review, we present 4 cases of hyperparathyroidism caused by ectopic parathyroid adenomas and review the literature on the additional value of their localization with SPECT/CT. Combined SPECT/CT scanners permit more reliable localization of ectopic adenomas. The additional information can aid in exact preoperative localization. In one study of 16 patients, SPECT/CT identified 39% more lesions compared with SPECT imaging alone. In other comparisons of planar, SPECT and SPECT/CT imaging modalities, SPECT/CT permitted the highest reader confidence in localization, especially for mediastinal adenomas. Larger studies are needed to establish the role and cost-effectiveness of SPECT/CT.
Article
Well differentiated thyroid cancer is a rare disease in the UK. It is the only cancer which, having metastasized, remains curable by radioisotope: therapy with I-131. The main indication for administering repeat doses of I-131 is the appearance of abnormal uptake in a whole body scan following diagnostic or therapeutic I-131 administration. False positive scans, showing the presence of I-131 uptake in the absence of residual thyroid tissue or metastases can occur, although they are uncommon. Unless recognized as a false positive,I-131 uptake may result in diagnostic error and lead to administration of an unnecessary therapy dose. We describe a series of nine patients in whom the scans showed false positive uptake of I-131, including cases where the cause of the uptake is still uncertain. We demonstrate the common sites of false positive uptake, discuss the underlying mechanisms and suggest a systematic approach to the interpretation of whole body scans in order to prevent unnecessary treatment with I-131.