Download minerva medica copyright

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
REVIEWS
Q J NUCL MED MOL IMAGING 2007;51:99-110
Nuclear medicine applications in molecular imaging:
2007 update
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
R
M OP
C
F. G. BLANKENBERG 1, H. W. STRAUSS 2
This review examines several classes of radiolabeled
agents, including analogs localizing in somatostatin,
benzodiazepine and dopamine receptors; analogs of
progesterone and estrogen; and agents localizing in
lesions with hypoxia. It concludes the status of agents
advocated for detecting angiogenesis and inflammation. The current clinical status of these agents, and
their potential roles in diagnosis and treatment are discussed.
1Division
of Pediatric Radiology
F.G.B.-Department of Radiology
Lucile Salter Packard Children’s Hospital, Stanford, CA, USA
2Section of Nuclear Medicine
H.W.S.-Department of Radiology
Memorial Sloan Kettering Cancer Center, NY, USA
KEY WORDS: Tomography, emission-computed, single-photon
- Radiopharmaceuticals - Molecular imaging - Receptors.
Fusion imaging with
magnetic resonance imaging
and computed tomography
adionuclide detectors are exceptionally sensitive
to small amounts of radioactivity. Collimated single photon imaging systems typically provide sensitivities of 2-4 counts/s/mCi of technetium-99m in the
field of view, while positron emission tomography
(PET) full ring imaging systems typically have sensitivities of ~2-5 thousand counts/s/mCi of fluorine-18
in the field of view. These sensitivities can be used to
detect nanomolar concentrations of tracer.1, 2 The high
sensitivity permits mapping of metabolism, perfusion,
receptor expression, and dynamic imaging of compartments, pools and spaces. Although these systems
Supported in part by NIH Grant # EB000898.
Address reprint requests to: F. G. Blankenberg, M.D., 725 Welch Road
Palo Alto, CA 94304 USA. E-mail: [email protected]
Vol. 51 - No. 2
are sensitive, they lack the anatomic resolution of
ultrasound, computed tomography (CT), and magnetic resonance imaging (MRI). Combining single
photon emission computed tomography (SPECT) and
PET with CT and MRI to make hybrid PET/CT,3, 4
SPECT/CT,5-7 and PET/MRI 8, 9 scanners provides
images containing both functional and anatomic information in co-registered fusion images (Figure 1),10 to
define a specific physiologic parameter in a clearly
defined anatomic location. Hybrid units have several
distinct advantages: the systems provide non-image
based spatial co-registration of the multimodal image
data, assuming the patient remains motionless between
studies; the CT data allows attenuation correction of
PET or SPECT data; the combined PET-CT facilitates
biopsy of lesions visible on radionuclide scans, that
cannot be appreciated on CT (as often observed in
malignant peritoneal disease from gynecological cancers).11 A number of recent investigations have successfully used fusion imaging performed directly with
dual modality units for preoperative localization of
tumor, detection of recurrent disease, tumor staging,
and the prediction of tumor response.12-16 In all stud-
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
99
BLANKENBERG
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
The somatostatins
Somatostatin is a peptide hormone that inhibits cell
proliferation. Proliferating cells in some tumors express
somatostatin receptors. Five different somatostatin
receptors, SSTR-1 through SSTR-5,27 have been identified. SSTR-3 activation leads to target cell apoptosis
while the other four inhibit growth by cell-cycle
arrest.28 The marked somatostatin receptor (mostly
SSTR-2) density, found in breast carcinoma, lymphoma
and neuroendocrine tumors, permits clinical SPECT
imaging of primary and metastatic tumor with currently available radiolabeled somatostatin analogs
namely, [111In]D-Phe-DTPA-octreotide (Octreoscan,
which primarily recognizes SSTR-2) 29, 30 and [99mTc]
depreotide (Neotect, which primarily recognizes SSTR3).31 Reports describe the use of these agents for the
detection of malignant lung nodules,32, 33 bronchial
carcinoids,34 paragangliomas of the head and neck,35
primary and metastatic insulinoma,36 ectopic Cushing’s
syndromes,37 metastatic iodine negative thyroid carcinoma,38 predicting the endocrine response of
metastatic breast carcinoma,39 as well as pathologic
lymphocytic processes, such as lymphoma,40 Graves’
opthalmopathy,41, 42 granulomatous disease,43 cardiac
allograft rejection,44 and the formation of unstable
(vulnerable) atherosclerotic plaques, in which T-lymphocytes accumulate and overexpress the SSTR-2
receptor.45 When [111In]D-Phe-DTPA-octreotide was
imaged with hybrid SPECT/CT scanners, preoperative localization of abdominal neuroendocrine tumors
improved significantly compared to SPECT alone.
This improvement caused a change of surgical or
radiotherapeutic management in 28% of patients studied.46
Somatostatin analogues can also be used as a carrier for targeted radionuclide therapy to treat neuroendocrine tumors expressing SSTR-2 and STTR-5
receptors.47 For therapy, a β-emitting nuclide, such
as yttrium-90 (90Y), is used in place of the γ emitter
111In.48 Using the same peptide platform for diagnostic lesion localization, dose-planning and therapy
allows more precise calculation of tumor dose, as
well as the dose to bone marrow and kidneys. The
high concentration of somatostatin peptides in the
renal cortex is a major factor limiting the administered dose for tumor therapy. There are several strategies to reduce the renal uptake of somatostatin
analogs, including preloading with infusions of amino
acids (such as lysine), succinylated gelatin,49, 50 and
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Figure 1.—FDG PET/CT scan of a 51 year old man with stage IV
Hodgkins disease. The coronal PET (A), transaxial through the lesion
in the right axilla (B), CT at the same level as the transaxial PET (C),
and the transaxial fusion image (D) depicting the anatomic correlation of the enlarged node seen on CT with the metabolic focus seen
on the transaxial PET.
ies, the fusion of cross-sectional image data with
radionuclide images resulted in improved detection of
tumor that lead to changes in patient staging, prognosis, or therapeutic management. Misregistration of
data, caused by respiratory or cardiac motion, remains
significant challenges for these hybrid imaging techniques.
Oncology has been the major focus of fusion imaging with fluorodeoxyglucose (FDG) PET/CT for the
past 5 years. Applications of combined imaging
include: radioiodine studies in patients with metastatic thyroid cancer 17 (to distinguish osseous lesions
from uptake in nodes and soft tissue), somatostatin
and meta-iodo-benzylguanidine imaging 18, 19 (to precisely localize uptake in bowel, nodes, adrenal glands
and liver), bone scans in patients suspected of metastatic disease,20 programmed cell death with radiolabeled-annexin V,21 tissue hypoxia with misonidazole
and 2-nitroimadazole analogs.22 In addition to oncologic applications, hybrid imaging provides detailed
information about brain pathophysiology with labeled
neurotransmitter analogs,23 provides opportunities to
study the mechanisms of inflammation,24 the progression of angiogenesis,25 and the distribution of βamyloid in Alzheimer’s disease.26 We discuss these
and other radiotracers below.
100
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
June 2007
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
recently the use of angiotensin converting enzyme
inhibitors. Work is ongoing to see which of these
approaches will be best suited to the clinic.
The SSTR-2 receptor gene has also been successfully
used as a molecular reporter of the incorporation and
expression of DNA constructs in vivo.51, 52 As somatostatin analogues have proven safe and effective in
humans, it is possible that cells genetically engineered
to express the SSTR-2 receptor in vitro or via viral or
other in vivo vector could be in the near future noninvasively detected and monitored with SPECT/CT.
BLANKENBERG
Dopamine transporter and D2 dopamine receptor
imaging
The presynaptic dopamine transporter (DAT) and
the postsynaptic D2 receptor are two of the most
extensively studied neurotransmitter-receptor systems
in the CNS.62, 63 Several disease states, including
depression, the antipsychotic drug induced negative
syndrome of schizophrenia, Parkinson’s disease, and
extrapyramidal Parkinson-plus neurodegenerative
syndromes are characterized by focal or regional
decreases in DAT and D2 receptor binding.64-67 Opiates
and Parkinson’s disease both effect the DAT system
with decreased uptake on SPECT images recorded
with [123I]β-CIT, a cocaine analog with a binding constant of 1.6 nM for the DAT and [123I]FP-CIT, a tracer
that has been successful at documenting the accelerated presynaptic dopaminergic degeneration found
in Parkinson’s patients.68 Diseases characterized by
abnormal increases in D2 receptor binding potential
include attention deficit-hyperactivity disorder, mania
and schizophrenia. Iodine-123-iodo-benzamide or
[123I]iodolisuride are also useful for SPECT. Lisuride, an
ergolene derivative used in the treatment of
Parkinson’s disease, k=0.27 nM, is similar to 76 Br-bromolisuride used for PET. These studies all show excellent correlations of D2 binding potential with neuropsychiatric function and may have an immediate
benefit in the diagnosis and treatment of the depression that occurs in over 1/3 of patients undergoing
anti-psychotic treatment for schizophrenia.69, 70
One fascinating area of clinical research is the specific identification of abnormalities of the dopaminergic
system in patients with substance abuse.71, 72 Patients
abusing cocaine, methamphetamine, methylenedioxymethaphetamine, alcohol, opiates, tobacco, marijuana, and inhalants all appeared to be associated
with abnormalities of the brain dopamine system, the
primary force behind the reward center in humans.
Dopamine producing cell bodies are located in the
midbrain within the substantia nigra and the ventral
tegmental area with projections to the striatal area,
that is known as the reward center, the nucleus accumbens. All abused substances, despite different mechanisms of action, increase synaptic levels of dopamine.
Chronic substance abusers have stimulant-induced
highs associated with increases in brain dopamine,
but abnormally low numbers of dopamine D2 receptors at rest as measured by the PET radioligand
[11C]raclopride.73-75 Carbon-11-raclopride is a radio-
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Gamma-aminobutyric acid A-benzodiazepine receptor agonists
Gamma-aminobutyric acid (GABA) is the most
abundant inhibitory transmitter in the central nervous
system (CNS).53 This neurotransmitter is distributed
within GABAergic neurons throughout the brain.
When GABA inhibitory activity exceeds that of excitatory inputs (mainly glutaminergic) sedation, amnesia and ataxia appear. Benzodiazepines work by
potentiating the effects of GABA on the chloride ion
channel of GABAA-benzodiazepine receptor complex.
Benzodiazepine derivatives [11C]flumazenil for PET
and [123I]iomazenil for SPECT, that primarily image
the peripheral (as opposed to central) benzodiazepine
receptors, are commercially available and have been
applied to the neuroimaging of a variety of neurologic disorders including, anxiety and temporal lobe
epilepsy, characterized in part by decreases in GABAA
receptors within the brain.54-56 In addition, reductions
in temporal-mesial uptake of [123I]iomazenil can be
found in regions that are structurally intact by MRI in
patients with medically refractory temporal lobe
epilepsy.57 It is now recommended that presurgical
studies with [123I]iomazenil (SPECT) or [11C]flumazenil
(PET) be performed as part of routine imaging along
with MRI.
Iomazenil also binds selectively to activated
microglial cells (brain macrophages). These cells have
no significant binding of tracer in their quiescent state.
Microglial cells make up 1 out of 10 cells in the normal brain. Activated microglial cells are abundant in
the entorhinal, temporoparietal, and cingulate cortex
of patients with Alzheimer’s presenile dementia and
multiple sclerosis, suggesting that these diseases can
be readily imaged, quantified, and serially followed
with [123I]iomazenil SPECT or [11C]flumazenil PET imaging.58-61
Vol. 51 - No. 2
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
101
BLANKENBERG
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
pharmaceutical that binds to the postsynaptic D2/3
receptors and, therefore, is an indirect indicator of
endogenous concentrations of dopamine. This effect
coupled to decreases in dopamine release in response
to chemical stimuli leads to the compulsive drug seeking behavior in chronic substance abusers and is an
active area of neuropsychiatric investigation.76
The nitroimidazoles are chemically reduced when
they enter tissues. In the presence of adequate tissue
oxygen levels, the molecule is re-oxidized, remains soluble, and diffuses out of the tissue.85, 86 If the oxygen
tension in the cell is low, further reduction occurs,
the molecule becomes insoluble and is trapped in
the tissue. Misonidazole analogs have been labeled
with 18F for PET imaging and 2-nitroimadazole analogs
have been labeled with 123I and 99mTc for SPECT imaging of hypoxia. These agents have demonstrated
increased uptake in hypoxic and low flow ischemic
myocardium and brain as well as in tumors.87-92
Technetium-99m labeled HL91 and BRU 59-21 as well
as 18F-labeled fluoromisonidazole ([18F]FMISO) have
been recently been used to study temporal changes in
tumor hypoxia in patients undergoing radiation and
chemotherapy for primary and recurrent squamous
head and neck carcinoma.93-95 Hypoxia may become
more important as PET/CT imaging is gaining importance in radiation treatment planning. FDG PET/CT
identifies areas of metabolically active tissue within the
tumor volume. This information is used to define the
areas to ‘boost’ with intensity modulated radiation
therapy (IMRT). Mapping the distribution of hypoxia
in tumors offers additional information to integrate
into the IMRT radiation therapy plan.96
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Estrogen and progesterone
Selection of therapy in breast cancer is influenced
by the expression of estrogen receptors by the tumor
cells. In the absence of imaging, receptor status is
determined by multiple biopsies of the primary tumor
and regional lymph nodes.77 Both 18F estrogen
analogs 78 and iodinated compounds have been synthesized for imaging.79 An iodinated estrogen analog,
123-labeled cis-11β-methoxy-17α-iodovinyl estradiol
(Z-[123I]MIVE) has proven successful as a predictor of
tamoxifen therapy in a recent study of breast cancer
patients in which all patients with faint baseline uptake
or mixed or no estrogen receptor (ER) blockade after
tamoxifen showed progressive disease, while patients
with clear baseline uptake and complete ER blockade
after tamoxifen had a significantly longer progression-free interval.80 Progesterone receptor SPECT imaging with a progesterone analog, Z-[123I]IPG2, may also
be possible in the near future.81
Hypoxia
Most tissues require a sufficient level of oxygen to
maintain function. To facilitate the maintenance of
adequate oxygen supplies, there are oxygen sensors82,
83 that initially respond to low oxygen levels by complex signaling for vascular dilatation (typically by
nitric oxide mediated responses). When this is insufficient, hypoxic tissues produce a number of growth
factors that stimulate the growth of new capillaries,
including vascular endothelial growth factor (VEGF).84
Neoplasms are often hypoxic due to their relatively
unstructured cellular proliferation, and lack of well
developed arteries. Lack of oxygen in these lesions
leads to the production of substantial amounts of
angiogenic factors, which ultimately results in the
neovascularity associated with neoplasia. One
approach to treating tumors interferes with hypoxia
generated angiogenic signaling.
Decreased oxygen tension in tissues can be imaged
with radiolabeled agents, such as the nitroimidazoles.
102
Imaging of the αvβ3 integrin and vascular endothelial
growth factor receptors
The integrins, a family of heterodimeric endothelial
cell membrane proteins, serve as adhesion receptors
for extracellular matrix proteins that contain exposed
arginine, glycine, and, aspartate (single letter coding
RGD) amino acid sequences.97 These include laminin,
fibronectin, collagens, and vitronectin that help form
blood vessels. The most abundant integrin expressed
on the surface of proliferating endothelial cells is the
αvβ3 receptor.98 In the adult human the αvβ3 integrin
has a limited tissue distribution. It is not expressed on
quiescent epithelial cells and appears at minimal levels on smooth muscle cells.99 In contrast, both activated
endothelial cells in tumor capillaries,100 and some
tumor cells101 express high levels of αvβ3.
The cyclic pentapeptide cyclo (-Arg-Gly-Asp-DPhe-Val-) has been identified as a potent (Kd <10
nmol/L) inhibitor of αvβ3 integrin binding to extracellular matrix proteins.102 Modifications of this peptide at position 4 or 5 have allowed radiolabeling with
iodine for SPECT and 18F for PET imaging.103, 104
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
June 2007
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
Contrast between tumor and normal tissues (especially liver) has been since improved by addition of
sugar to the amino acids of the peptide.105-107
Radiolabeled RGD-peptides have been recently used
to image αvβ3 expression in tumor prior to administration of αvβ3 antagonists, such as EMD-121974 to
allow selection of patients entering clinical trials.
These peptides in the near future will be used to
assess the effectiveness of αvβ3 integrin blockade by
specific doses of other αvβ3 antagonists. This approach
will permit optimization of dose for a specific patient
and tumor type.
Sites of neoangiogenesis may be imaged with a
number of tracers. Maschauer et al.108 demonstrated an
82% increase in FDG uptake in vascular endothelial
cells stimulated with VEGF, a potent pro-angiogenic
growth factor. This relationship has been borne out by
several clinical studies in which FDG uptake positively correlated with VEGF expression.109 Specific
growth factor receptors at sites of neoangiogenesis
may also be imaged using several radiolabeled forms
of VEGF.110-112 Although these radiolabeled VEGF
analogs localize, the random localization of the radiolabel chelates (such as HYNIC) on the VEGF molecule, may have a significant effect on the biodistribution of the labeled molecule. A new recombinant
VEGF combining two 3-112 aa fragments of VEGF121
into a single-chain (sc) protein expressed with N-terminal 15-aa cysteine-containing Cys-tag (scVEGF) was
developed to allow reproducible site-specific labeling
for SPECT, PET, and fluorescent imaging via facile
malemide based chemistry.113 sc-VEGF also has the
advantage of extreme heat stability and can withstand
90 °C up to 10 min that maybe helpful for the chelation of metal isotopes such as 177Lu, 90Y, and 64Cu;
and has no pro-angiogenic potential as this domain
has been removed from the recombinant protein.
Another isoform of VEGF, VEGF165 labeled with 123I for
SPECT, has also proven successful for the imaging of
tumor in 9 patients with pancreatic carcinoma.114
BLANKENBERG
very high concentrations in the placenta and lower
concentrations in endothelial cells, kidney, myocardium, skeletal muscle, skin, red cells, platelets and
monocytes. Although the precise physiologic function of annexin is uncertain, the protein has several
well studied functions, including: inhibition of coagulation (annexin was originally discovered because of
its ability to trap calcium [i.e. “annex” calcium] and prevent clotting); inhibition of phospholipase A2, an
enzyme responsible for the release of arachidonic
acid from the cell membranea component of the
inflammatory process; and inhibition of protein kinase
C, a system responsible for intracellular signaling.
The binding of annexin V to sites of phosphatidylserine (PS) expression in vivo has been found
to be extremely complex and difficult to model. While
annexin V is a relatively large protein (about half the
molecular weight of albumin) it was shown early on
that the protein can be internalized at sites of ischemic
injury both in the heart and brain and cross the intact
blood brain barrier.128 The mechanism of annexin V
uptake into PS expressing cells appears to be via a
newly described energy dependent form of pinocytosis.129
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Imaging of apoptosis
While many tracers are being developed as imaging agents for apoptosis 115-126 radiolabeled annexin V
so far is the only radiopharmaceutical that has been
studied extensively in animals and in humans.127
Annexin V (MW ⊕36 000) is an endogenous human
protein that is widely distributed intracellularly, with
Vol. 51 - No. 2
Positron emission tomography
and single photon emission
computed tomography imaging
with radiolabeled annexin V
Annexin has been radiolabeled with 125I, 124I, 18F,
and 68Ga. An array of human imaging studies has
been performed with recombinant human annexin
V. Two clinical trials were performed with the first
form of radiolabeled annexin V, [99mTc]N2S2-rh annexin, the same formulation used in an unrelated clot
detection trial.130 The first study was designed to detect
graft rejection in heart transplant recipients by Narula
et al.131 that studied 18 cardiac allograft recipients. In
the second trial, Belhocine et al.132 studied the
increased uptake of tracer with in days after the start
of chemotherapy in 15 cancer patients in late stages
small cell and non-small cell lung cancers.
To reduce the complexity of preparing the radiolabeled material, alternative labeling approaches were
sought and hydrazino nicotinamide (HYNIC) was
selected as the coupling molecule.133 The whole labeling procedure has been reduced to a standardized
two-vial kit that can be ready for patient use within 30
99mTc
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
103
BLANKENBERG
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
min of receiving 20-30 mCi of sterile [99mTc]pertechnetate from a local radiopharmacy or generator.
Unfortunately, although [99mTc]HYNIC annexin V is
not concentrated in the liver or excreted in the bowel, it concentrates in the cortex of the kidney, limiting
visualization of any structures in this region.134 In spite
of this shortcoming, clinical trials tested the clinical utility of HYNIC-annexin V to determine the efficacy of
chemotherapy in patients with tumors,135-139 detect
apoptosis in areas of acute myocardial infarction,140, 141
define activity of rheumatoid arthritis (personal communication R. Hustinx and C. Beckers, Liege,
Belgium), ischemic preconditioning,142-144 identify vulnerable atherosclerotic plaque,145 acute stroke,146, 147
and Alzheimer’s dementia.148
There are alternative methods to radiolabel annexin V, including the use of self-chelating annexin V
mutants that have lower concentrations in the kidneys of rodents compared with HYNIC-annexin V.149
An alternative to random modification of annexin V
with bifunctional agents, such as HYNIC, is a selfchelating annexin V mutant, known as V-128.150, 151
Annexin V-128 is a fusion protein with an endogenous
Tc chelation site (Ala-Gly-Gly-Cys-Gly-His) added to
the N-terminus of annexin V, that can be rapidly
labeled with 99mTc using glucoheptonate as the
exchange reagent. This form of radiolabeled annexin V has major advantages over the HYNIC chelator
with regard to renal retention of 99mTc, with attendant
decreased abdominal background and renal radiation dose and because it is site specifically labeled
(as opposed to randomly modified) has at least twice
the in vivo uptake of other annexin V based tracers.
It will be helpful to quantify radiolabeled annexin
concentration before and after therapy in lesions.
Since PET has major advantages for quantitative imaging, several approaches to label annexin V with 18F
have been developed. Two laboratories have used
N-succinimidyl 4-fluorobenzoate 152, 153 to synthesize
[18F]annexin V. The fluorine-labeled agent has lower
uptake in the liver, spleen, and kidney compared to
HYNIC-annexin V.
tance.154-158 The These agents work by different mechanisms: radiolabeled phagocytes depend on chemotaxis to attract the cells to the lesion site; gallium
depends on the transfer of the metal transferrin complex into cells at the site of inflammation; while FDG
is concentrated in metabolically active cells, most likely macrophages, undergoing respiratory burst activation. When blood is incubated with FDG, it is possible to label white cells in vitro, likely due to the cells
activation in course of blood withdrawal into a syringe.
Trials in humans are ongoing.159, 160 Other investigational agents for imaging inflammation include: antigranulocytic antibodies and antibody fragments, nonspecific immunoglobulin G, liposomes, chemotactic
peptides, interleukins and chemokines. For each of
these agents reliable and efficient one-step with 99mTcor 123I-based labeling methods have been developed
and tested in humans.161 However, these agents have
the potential for unforeseen immunologic (usually a
mild drop in white blood cells [WBC] count due to
transient margination) or allergic responses. One of the
most promising of the anti-granulocyte antibody based
tracers, [99mTc]fanolesomab, an agent that binds to the
CD15 antigen expressed on the surface of neutrophils,
eosinophils, and lymphocytes was withdrawn from the
market in 2005 because of two cardiopulmonary
deaths occurring within 30 min after administration of
tracer.162
In general, it appears that liposomes, antibodies
against specific immune markers, nonspecific IgG,
and radiolabeled immune cells lack rapid background
clearance while the agents with relative fast pharmacokinetics (chemotactic peptides, interleukins) lack
high uptake in the target. The side effects of some of
the chemotactic peptides and interleukins (e.g. IL-1)
also need to be addressed by the use of lower doses
(<10 µg/kg of protein), such as has been done with
131I-labeled IL-8 a marker of acute inflammation163, 164
or the use of another less toxic member within a specific family of immune related proteins or peptides,
such as the substitution of IL-1 (either the α or β isoforms) with IL-1 receptor antagonist that has the same
affinity for IL-1 receptors but is lacking in biologic
activity.165 Despite the successful labeling of the IL-1
receptor antagonist with 123I, studies in humans have
so far been disappointing as the tracer appears to
localize non-specifically to regions of reactive edema in a similar fashion as polyclonal IgG.166
There are, however, notable exceptions to the rules
above including several agents that target the CXC-
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Imaging of inflammatory cells
A number of radiotracers have been proposed to
image inflammation, yet to date only white blood cell
scanning, 67Ga, and [18F]FDG have gained accep-
104
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
June 2007
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
receptor family (recognizing a chemokine characterized by 4 cysteines that form essential disulfide bonds),
particularly IL-2, platelet factor (PF4), IL-8, all of which
display remarkably efficient target uptake in combination with rapid background clearance. Both IL-2 167
and PF4 168 have a high affinity for monocytes and
may, therefore, be useful for imaging chronic inflammation, a process that is difficult to detect with the
granulocyte specific tracers, such as radiolabeled
leukocytes or neutrophil specific markers such as
leukotriene B4 (imaged with the antagonist,
DPC11870), NAP-2 and its variants (neutrophil activating petide-2), NCA-90 and NCA-95 (LeukoScan)
antibodies 169, 170 used for acute inflammation imaging.
The first of these agents is IL-2, a cytokine that is a
member of the interleukin family. IL-2 binds to the Th1
(T-helper 1) receptor expressed on surface of T-cells,
that are an integral part of the chronic lymphocytic
infiltrates seen in Crohn’s disease, celiac disease, type1 diabetes and autoimmune thyroiditis.171-173 IL-2 can
be readily radiolabeled with either 123I or 99mTc; however, 99mTc labeling has proven to be a complex procedure limiting its routine use in the clinic.174
IL-8 binds to neutrophils with nanomolar affinity and
has been successfully tested in human trials.175-177
Similar success has been found patients with acute
osteomyelitis using 99mTc-labeled anti-granulocyte Fab
antibody scintigraphy (LeukoScan).178 Despite the success of Leukoscan for imaging osteomyelitis and soft
tissue infection, such as acute appendicitis, caution
must be exercised as with this and other immune
tracers as it has not proven to be of use in other types
of inflammation such as with inflammatory bowel disease.179
Another major chemokine that can bind to monocytes and macrophages (Mφs) is monocyte chemoattractant peptide-1 (MCP-1), an endogenous human
peptide with isoforms weighing 9-12 kDa, that selectively binds to the CCR-2 receptor (a C-C based receptor) with a nanomolar affinity.180 MCP-1 has so far
been successful in the imaging of experimental atherosclerosis however no human trials have yet to be
conducted.181, 182
The latest generation of inflammatory markers selectively target the extracellular components of an infectious process and include a naturally occurring antimicrobial peptides, such as 99mTc-UBI 29-41
([99mTc]ubiquicidin 29-41), that target bacteria directly.183, 184 Another example is chitin, which is expressed
in the fungal cell wall, but is absent in mammalian and
BLANKENBERG
bacterial cells. Chitin can be detected with SPECT
using 123I-labeled chitinase: a marker that may prove
a great benefit in the work up and treatment of polymicrobial or opportunistic infections in immunocompromised hosts that lack sufficient numbers of leukocytes for standard WBC labeling techniques.185 The last
class of direct microbial labeling agents are radiolabeled antibiotic drugs, including [99mTc]ciprofloxacin
(and related fluoroquinolones) and ceftizoxmine, that
are now in clinical trials.186
The last major approach to the imaging of infection
is the targeting of the effects of inflammation on the
host’s cells and tissues. Acute inflammation is resolved
through the PS-specific recognition and clearance of
apoptotic granulocytes, cells that have outlived their
useful function.187, 188 Monocytes and Mφs also accelerate the apoptosis of bystander (unwanted) granulocytes at sites of inflammation.189 The degree of
macrophage infiltration and its associated granulocytic apoptotic response can, therefore be imaged
with radiolabeled annexin V.190 Annexin V also has the
advantage that it should be able to image all types of
inflammation including that seen in unstable atherosclerotic plaques and while non-specific with respect
to the exact type of infection it is far more robust and
versatile as compared with more specific immune
markers.191-193
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Vol. 51 - No. 2
References
1. Martin WH, Delbeke D, Patton JA, Sandler MP. Detection of malignancies with SPECT versus PET, with 2-[fluorine-18]fluoro-2-deoxyD-glucose. Radiology 1996;198:225-31.
2. Behr TM, Gotthardt M, Barth A, Behe M. Imaging tumors with
peptide-based radioligands. Q J Nucl Med 2001;45:189-200.
3. Jarritt PH, Carson KJ, Hounsell AR, Visvikis D. The role of PET/CT
scanning in radiotherapy planning. Br J Radiol 2006;79 Spec No
1:S27-35.
4. Tsukamoto E, Ochi S. PET/CT today: system and its impact on cancer diagnosis. Ann Nucl Med 2006;20:255-67.
5. Schillaci O, Filippi L, Danieli R, Simonetti G. Single-photon emission computed tomography/computed tomography in abdominal diseases. Semin Nucl Med 2007;37:48-61.
6. Schillaci O, Filippi L, Manni C, Santoni R. Single-photon emission
computed tomography/computed tomography in brain tumors.
Semin Nucl Med 2007;37:34-47.
7. Sodee DB, Sodee AE, Bakale G. Synergistic value of single-photon emission computed tomography/computed tomography fusion
to radioimmunoscintigraphic imaging of prostate cancer. Semin
Nucl Med 2007;37:17-28.
8. Pichler BJ, Judenhofer MS, Catana C, Walton JH, Kneilling M,
Nutt RE et al. Performance test of an LSO-APD detector in a 7-T
MRI scanner for simultaneous PET/MRI. J Nucl Med 2006;47:
639-47.
9. Seemann MD. Whole-body PET/MRI: the future in oncological
imaging. Technol Cancer Res Treat 2005;4:577-82.
10. Yamamoto Y, Nishiyama Y, Monden T, Matsumura Y, Satoh K,
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
105
BLANKENBERG
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
Ohkawa M. Clinical usefulness of fusion of 131I SPECT and CT
images in patients with differentiated thyroid carcinoma. J Nucl Med
2003;44:1905-10.
Schillaci O, Simonetti G Fusion imaging in nuclear medicine:
applications of dual-modality systems in oncology. Cancer Biother
Radiopharm 2004;19:1-10.
Antoch G, Kanja J, Bauer S, Kuehl H, Renzing-Koehler K, Schuette
J et al. Comparison of PET, CT, and dual-modality PET/CT imaging for monitoring of imatinib (STI571) therapy in patients with gastrointestinal stromal tumors. J Nucl Med 2004;45:357-65.
Schillaci O, Spanu A, Madeddu G. [99mTc]sestamibi and
[99mTc]tetrofosmin in oncology: SPET and fusion imaging in lung
cancer, malignant lymphomas and brain tumors. Q J Nucl Med Mol
Imaging 2005;49:133-44.
Grosu AL, Weber WA, Franz M, Stark S, Piert M, Thamm R et al.
Reirradiation of recurrent high-grade gliomas using amino acid
PET (SPECT)/CT/MRI image fusion to determine gross tumor volume for stereotactic fractionated radiotherapy. Int J Radiat Oncol
Biol Phys 2005;63:511-9.
von Schulthess GK. Integrated modality imaging with PET-CT and
SPECT-CT: CT issues. Eur Radiol 2005;15 Suppl 4:D121-6.
Schillaci O. Single-photon emission computed tomography/computed tomography in lung cancer and malignant lymphoma. Semin
Nucl Med 2006;36:275-85.
Lind P, Kohlfurst S. Respective roles of thyroglobulin, radioiodine
imaging, and positron emission tomography in the assessment of
thyroid cancer. Semin Nucl Med 2006;36:194-205.
Gabriel M, Hausler F, Bale R, Moncayo R, Decristoforo C, Kovacs
P et al. Image fusion analysis of (99m)Tc-HYNIC-Tyr(3)-octreotide
SPECT and diagnostic CT using an immobilisation device with
external markers in patients with endocrine tumours. Eur J Nucl
Med Mol Imaging 2005;32:1440-51.
Ingui CJ, Shah NP, Oates ME. Endocrine neoplasm scintigraphy:
added value of fusing SPECT/CT images compared with traditional side-by-side analysis. Clin Nucl Med 2006;31:665-72.
Groves AM, Bird N, Tabor I, Cheow HK, Balan KK. 16-Detector
multislice CT-skeletal scintigraphy image co-registration. Nucl
Med Commun 2004;25:1151-5.
Kartachova MS, Valdes Olmos RA, Haas RL, Hoebers FJ, van den
Brekel MW, van Zandwijk N et al. Mapping of treatment-induced
apoptosis in normal structures: 99mTc-Hynic-rh-annexin V SPECT
and CT image fusion. Eur J Nucl Med Mol Imaging 2006;33:893-9.
Padhani A. PET imaging of tumour hypoxia. Cancer Imaging
2006;6:S117-21.
Rufini V, Calcagni ML, Baum RP. Imaging of neuroendocrine
tumors. Semin Nucl Med 2006;36:228-47.
Gemmel F, Dumarey N, Palestro CJ. Radionuclide imaging of
spinal infections. Eur J Nucl Med Mol Imaging 2006;33:1226-37.
Daldrup-Link HE, Simon GH, Brasch RC. Imaging of tumor angiogenesis: current approaches and future prospects. Curr Pharm
Des 2006;12:2661-72.
Small GW, Kepe V, Ercoli LM, Siddarth P, Bookheimer SY, Miller
KJ et al. PET of brain amyloid and tau in mild cognitive impairment.
N Engl J Med 2006;355:2652-63.
Reichlin S. Somatostatin. N Engl J Med 1983;309:1495-501.
Kvols LK, Woltering EA. Role of somatostatin analogs in the clinical management of non-neuroendocrine solid tumors. Anticancer
Drugs 2006;17:601-8.
Hillel PG, van Beek EJ, Taylor C, Lorenz E, Bax ND, Prakash V et
al. The clinical impact of a combined gamma camera/CT imaging
system on somatostatin receptor imaging of neuroendocrine
tumours. Clin Radiol 2006;61:579-87.
Modlin IM, Latich I, Zikusoka M, Kidd M, Eick G, Chan AK.
Gastrointestinal carcinoids: the evolution of diagnostic strategies.
J Clin Gastroenterol 2006;40:572-82.
Weiner RE, Thakur ML. Radiolabeled peptides in oncology: role in
diagnosis and treatment. BioDrugs 2005;19:145-63.
Plachcinska A, Mikolajczak R, Maecke H, Kozak J, Michalski A,
Rzeszutek K et al. Efficacy of 99mTc-EDDA/HYNIC-TOC scintig-
106
33.
34.
raphy in differential diagnosis of solitary pulmonary nodules.
Cancer Biother Radiopharm 2004;19:613-20.
Ferran N, Ricart Y, Lopez M, Martinez-Ballarin I, Roca M, Gamez
C et al. Characterization of radiologically indeterminate lung
lesions: 99mTc-depreotide SPECT versus 18F-FDG PET. Nucl Med
Commun 2006;27:507-14.
Granberg D, Sundin A, Janson ET, Oberg K, Skogseid B, Westlin
JE. Octreoscan in patients with bronchial carcinoid tumours. Clin
Endocrinol 2003;59:793-9.
Duet M, Sauvaget E, Petelle B, Rizzo N, Guichard JP, Wassef M et
al. Clinical impact of somatostatin receptor scintigraphy in the
management of paragangliomas of the head and neck. J Nucl Med
2003;44:1767-74.
Bertherat J, Tenenbaum F, Perlemoine K, Videau C, Alberini JL,
Richard B et al. Somatostatin receptors 2 and 5 are the major
somatostatin receptors in insulinomas: an in vivo and in vitro
study. J Clin Endocrinol Metab 2003;88:5353-60.
Tsagarakis S, Christoforaki M, Giannopoulou H, Rondogianni F,
Housianakou I, Malagari C et al. A reappraisal of the utility of
somatostatin receptor scintigraphy in patients with ectopic adrenocorticotropin Cushing’s syndrome. J Clin Endocrinol Metab
2003;88:4754-8.
Rodrigues M, Li S, Gabriel M, Heute D, Greifeneder M, Virgolini
I. 99mTc-depreotide scintigraphy versus 18F-FDG-PET in the diagnosis of radioiodine-negative thyroid cancer. J Clin Endocrinol
Metab 2006;91:3997-4000.
Van Den Bossche B, Van Belle S, De Winter F, Signore A, Van de
Wiele C. Early prediction of endocrine therapy effect in advanced
breast cancer patients using 99mTc-depreotide scintigraphy. J Nucl
Med 2006;47:6-13.
Dalm VA, Hofland LJ, Mooy CM, Waaijers MA, van Koetsveld PM,
Langerak AW et al. Somatostatin receptors in malignant lymphomas: targets for radiotherapy? J Nucl Med 2004;45:8-16.
Nocaudie M, Bailliez A, Itti E, Bauters C, Wemeau JL, Marchandise
X. Somatostatin receptor scintigraphy to predict the clinical evolution and therapeutic response of thyroid-associated ophthalmopathy. Eur J Nucl Med 1999;26511-7.
Cascini GL, Cuccurullo V, Rambaldi PF, Mansi L. Immunological
imaging using marked octreotide. Minerva Endocrinol 2001;2612933.
Galuska L, Leovey A, Szucs-Farkas Z, Szabados L, Garai I, Berta A
et al. Imaging of disease activity in Graves’ orbitopathy with different methods: comparison of (99m)Tc-DTPA and (99m)Tcdepreotide single photon emission tomography, magnetic resonance imaging and clinical activity scores. Nucl Med Commun
2005;26:407-14.
Balon HR, Goldsmith SJ, Siegal BA, Silberstein EB, Krenning EP,
Lang O et al. Procedure guideline for somatostatin receptor scintigraphy with 111In-pentetreotide. J Nucl Med 2001;42:1134-8.
Davies JR, Rudd JH, Weissberg PL, Narula J. Radionuclide imaging
for the detection of inflammation in vulnerable plaques. J Am Coll
Cardiol 2006;47(8 Suppl):C57-68.
Kowalski J, Henze M, Schuhmacher J, Macke HR, Hofmann M,
Haberkorn U. Evaluation of positron emission tomography imaging using [68Ga]-DOTA-D Phe(1)-Tyr(3)-Octreotide in comparison to [111In]-DTPAOC SPECT. First results in patients with neuroendocrine tumors. Mol Imaging Biol 2003;5:42-8.
Lewington VJ. Targeted radionuclide therapy for neuroendocrine
tumours. Endocr Relat Cancer 2003;10:497-501.
Forrer F, Waldherr C, Maecke HR, Mueller-Brand J. Targeted
radionuclide therapy with 90Y-DOTATOC in patients with neuroendocrine tumors. Anticancer Res 2006;26:703-7.
Weiner RE, Thakur ML. Radiolabeled peptides in oncology: role in
diagnosis and treatment. BioDrugs 2005;19:145-63.
Vegt E, Wetzels JF, Russel FG, Masereeuw R, Boerman OC, van Eerd
JE et al. Renal uptake of radiolabeled octreotide in human subjects
is efficiently inhibited by succinylated gelatin. J Nucl Med
2006;47:432-6.
Zinn KR, Buchsbaum DJ, Chaudhuri TR, Mountz JM, Grizzle WE,
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
June 2007
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
Rogers BE. Noninvasive monitoring of gene transfer using a
reporter receptor imaged with a high-affinity peptide radiolabeled
with 99mTc or 188Re. J Nucl Med 2000;41:887-95.
McCart JA, Mehta N, Scollard D, Reilly RM, Carrasquillo JA, Tang
N et al. Oncolytic vaccinia virus expressing the human somatostatin
receptor SSTR2: molecular imaging after systemic delivery using
111In-pentetreotide. Mol Ther 2004;10:553-61.
Nutt DJ, Malizia AL. New insights into the role of the GABA(A)-benzodiazepine receptor in psychiatric disorder. Br J Psychiatry
2001;179.390-6.
Matheja P, Ludemann P, Kuwert T, Weckesser M, Kellinghaus C,
Weitemeyer L et al. Disturbed benzodiazepine receptor function
at the onset of temporal lobe epilepsy: lomanzenil-binding in denovo TLE. J Neurol 2001;248:585-91.
Morimoto K, Tamagami H, Matsuda K. Central-type benzodiazepine receptors and epileptogenesis: basic mechanisms and
clinical validity. Epilepsia. 2005;46 Suppl 5:184-8.
Millet P, Graf C, Moulin M, Ibanez V. SPECT quantification of benzodiazepine receptor concentration using a dual-ligand approach.
J Nucl Med. 2006;47:783-92.
Saur D, Buchert R, Knab R, Weiller C, Rother J. Iomazenil-singlephoton emission computed tomography reveals selective neuronal loss in magnetic resonance-defined mismatch areas. Stroke
2006;37:2713-9.
Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer
FE et al. In-vivo measurement of activated microglia in dementia.
Lancet 2001;358:461-7. Erratum in: Lancet 2001;358:766.
Debruyne JC, Versijpt J, Van Laere KJ, De Vos F, Keppens J,
Strijckmans K et al. PET visualization of microglia in multiple
sclerosis patients using [11C]PK11195. Eur J Neurol 2003;10:
257-64.
Versijpt JJ, Dumont F, Van Laere KJ, Decoo D, Santens P, Audenaert
K et al. Assessment of neuroinflammation and microglial activation
in Alzheimer’s disease with radiolabelled PK11195 and single photon emission computed tomography. A pilot study. Eur Neurol
2003;50:39-47.
Mattner F, Katsifis A, Staykova M, Ballantyne P, Willenborg DO.
Evaluation of a radiolabelled peripheral benzodiazepine receptor
ligand in the central nervous system inflammation of experimental autoimmune encephalomyelitis: a possible probe for imaging
multiple sclerosis. Eur J Nucl Med Mol Imaging 2005;32:557-63.
Bergstrom KA, Jolkkonen J, Kuikka JT, Akerman KK, Viinamaki H,
Airaksinen O et al. Fentanyl decreases beta-CIT binding to the
dopamine transporter. Synapse 1998;29:413-5.
Voruganti L, Slomka P, Zabel P, Costa G, So A, Mattar A et al.
Subjective effects of AMPT-induced dopamine depletion in schizophrenia: correlation between dysphoric responses and striatal
D(2) binding ratios on SPECT imaging. Neuropsychopharmacology
2001;25:642-50.
Wong DF. In vivo imaging of D2 dopamine receptors in schizophrenia; the ups and downs of neuroimaging research. Arch Gen
Pyschiatry 2002;59:31-4.
Prunier C, Tranquart F, Cottier JP, Giraudeau B, Chalon S, Guilloteau
D et al. Quantitative analysis of striatal dopamine D2 receptors with
123I-iodolisuride SPECT in degenerative extrapyramidal diseases.
Nucl Med Commun 2001;22:1207-14.
Ilgin N, Senol S, Gucuyener K, Gokcora N, Sener S. Is increased
D2 receptor availability associated with response to stimulant
medication in ADHD. Dev Med Child Neurol 2001;43:755-60.
Winogrodzka A, Bergmans P, Booij J, van Royen EA, Janssen AGM,
Wolters EC. [123I]FP-CIT SPECT is a useful method to monitor the
rate of dopaminergic degeneration in early-stage Parkinson’s disease. J Neural Transm 2001;108:1001-19.
Seppi K, Scherfler C, Donnemiller E, Virgolini I, Schocke MF,
Goebel G et al. Topography of dopamine transporter availability
in progressive supranuclear palsy: a voxelwise [123I]beta-CIT
SPECT analysis. Arch Neurol 2006;63:1154-60.
Perez V, Catafau AM, Corripio I, Martin JC, Alvarez E. Preliminary
evidence of striatal D2 receptor density as a possible biological
BLANKENBERG
70.
71.
marker of prognosis in naive schizophrenic patients. Prog
Neuropsychopharmacol Biol Psychiatry 2003;27:767-70.
Glenthoj BY, Mackeprang T, Svarer C, Rasmussen H, Pinborg LH,
Friberg L et al. Frontal dopamine D(2/3) receptor binding in drugnaive first-episode schizophrenic patients correlates with positive
psychotic symptoms and gender. Biol Psychiatry 2006;60:621-9.
Volkow HD, Fowler JS, Wang GJ. Positron emission tomography
and single-photon emission computed tomography in substance
abuse research. Semin Nucl Med 2003;33:114-28.
Elkashef A, Vocci F. Biological markers of cocaine addiction: implications for medications development. Addict Biol 2003;8:123-39.
Doudet DJ, Jivan S, Holden JE. In vivo measurement of receptor
density and affinity: comparison of the routine sequential method
with a nonsequential method in studies of dopamine D2 receptors
with [11C]raclopride. J Cereb Blood Flow Metab 2003;23:280-4.
Martinez D, Gil R, Slifstein M, Hwang DR, Huang Y, Perez A et al.
Alcohol dependence is associated with blunted dopamine transmission in the ventral striatum. Biol Psychiatry 2005;58:779-86.
Volkow ND, Wang GJ, Ma Y, Fowler JS, Wong C, Ding YS et al.
Activation of orbital and medial prefrontal cortex by methylphenidate in cocaine-addicted subjects but not in controls: relevance to
addiction. J Neurosci 2005;25:3932-9.
Glenthoj BY, Mackeprang T, Svarer C, Rasmussen H, Pinborg LH,
Friberg L et al. Frontal dopamine D(2/3) receptor binding in drugnaive first-episode schizophrenic patients correlates with positive
psychotic symptoms and gender. Biol Psychiatry 2006;60:621-9.
Rose C, Thorpe SM, Andersen KW, Pedersen BV, Mouridsen HT,
Blichert-Toft M et al. Beneficial effect of adjuvant tamoxifen therapy in primary breast cancer patients with high oestrogen receptor values. Lancet 1985;1:16-9.
Linden HM, Stekhova SA, Link JM, Gralow JR, Livingston RB, Ellis
GK et al. Quantitative fluoroestradiol positron emission tomography imaging predicts response to endocrine treatment in breast cancer. J Clin Oncol 2006;24:2793-9.
Bennink RJ, Rijks LJ, van Tienhoven G, Noorduyn LA, Janssen
AG, Sloof GW. Estrogen receptor status in primary breast cancer:
iodine 123-labeled cis-11beta-methoxy-17alpha-iodovinyl estradiol scintigraphy. Radiology 2001;220:774-9.
Bennink RJ, van Tienhoven G, Rijks LJ, Noorduyn AL, Janssen
AG, Sloof GW. In vivo prediction of response to antiestrogen treatment in estrogen receptor-positive breast cancer. J Nucl Med
2004;45:1-7.
Zhou D, Carlson KE, Katzenellenbogen JA, Welch MJ. Bromine- and
iodine-substituted 16alpha,17alpha-dioxolane progestins for breast
tumor imaging and radiotherapy: synthesis and receptor binding
affinity. J Med Chem 2006;49:4737-44.
Poburko D, Lee CH, van Breemen C. Vascular smooth muscle
mitochondria at the cross roads of Ca(2+) regulation. Cell Calcium
2004;35:509-21.
Tang XD, Garcia ML, Heinemann SH, Hoshi T. Reactive oxygen
species impair Slo1 BK channel function by altering cysteinemediated calcium sensing. Nat Struct Mol Biol 2004;11:171-8.
Brogi E, Wu T, Namiki A, Isner JM. Indirect angiogenic cytokines
upregulate VEGF and bFGF gene expression in vascular smooth
muscle cells, whereas hypoxia upregulates VEGF expression only.
Circulation 1994;90:649-52.
Nunn A. Linder K. Strauss HW. Nitroimidazoles and imaging hypoxia. Eur J Nucl Med 1995;22:265-80.
Markus R, Donnan GA, Kazui S, Read S, Hirano T, Scott AM et al.
Statistical parametric mapping of hypoxic tissue identified by
[(18)F]fluoromisonidazole and positron emission tomography following acute ischemic stroke. Neuroimage 2002;16:425-33.
Parliament MB, Chapman JD, Urtasun RC, McEwan AJ, Golberg L,
Mercer JR et al. Non-invasive assessment of human tumour hypoxia with 123I-iodoazomycin arabinoside: preliminary report of a
clinical study. Br J Cancer 1992;65:90-5.
Cook GJ, Houston S, Barrington SF, Fogelman I. Technetium-99mlabeled HL91 to identify tumor hypoxia: correlation with fluorine18-FDG. J Nucl Med 1998;39:99-103.
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Vol. 51 - No. 2
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
107
BLANKENBERG
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
89. Melo T, Duncan J, Ballinger JR, Rauth AM. BRU59-21, a secondgeneration 99mTc-labeled 2-nitroimidazole for imaging hypoxia
in tumors. J Nucl Med 2000;41:169-76.
90. Hoffend J, Linke G, Mohammed A, Tiefenbacher CP, Eisenhut M,
Haberkorn U. 99mTcO(BAT-NI), a novel nitroimidazole tracer:
in vivo uptake studies in ischaemic myocardium. Eur J Nucl Med
Mol Imaging 2003;30:494-501.
91. Song HC, Bom HS, Cho KH, Kim BC, Seo JJ, Kim CG et al.
Prognostication of recovery in patients with acute ischemic stroke
through the use of brain SPECT with Technetium-99m-labeled
metronidazole. Stroke 2003;34:982-6.
92. Padhani A. PET imaging of tumour hypoxia. Cancer Imaging
2006;6:S117-21.
93. Van De Wiele C, Versijpt J, Dierckx RA, Moerman M, Lemmerling
M, D’Asseler Y et al. 99Tc(m) labelled HL91 versus computed
tomography and biopsy for the visualization of tumour recurrence
of squamous head and neck carcinoma. Nucl Med Commun
2001;22:269-75.
94. Hoebers FJ, Janssen HL, Olmos AV, Sprong D, Nunn AD, Balm AJ
et al. Phase 1 study to identify tumour hypoxia in patients with
head and neck cancer using technetium-99m BRU 59-21. Eur J
Nucl Med Mol Imaging 2002;29:1206-11.
95. Li L, Yu J, Xing L, Ma K, Zhu H, Guo H et al. Serial hypoxia
imaging with 99mTc-HL91 SPECT to predict radiotherapy response
in nonsmall cell lung cancer. Am J Clin Oncol 2006;29:628-33.
96. Schinagl DA, Kaanders JH, Oyen WJ. From anatomical to biological target volumes: the role of PET in radiation treatment
planning. Cancer Imaging 2006;6:S107-16.
97. Ruoslahti E, Engvall E. Integrins and vascular extracellular matrix
assembly. J Clin Invest 1997;100:S53-S56.
98. Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 1994;264:569-71.
99. Witmer AN, Dai J, Weich HA, Vrensen GF, Schlingemann RO.
Expression of vascular endothelial growth factor receptors 1, 2,
and 3 in quiescent endothelia. J Histochem Cytochem 2002;50:76777.
100. Eliceiri BP, Cheresh DA. The role of alphav integrins during
angiogenesis: insights into potential mechanisms of action and
clinical development. J Clin Invest 1999;103:1227-30.
101. Cheresh DA. Structure, function and biological properties of integrin alpha v beta 3 on human melanoma cells. Cancer Metastasis
Rev 1991;10:3-10.
102. Pfaff M, Tangemann K, Muller B, Gurrath M, Muller G, Kessler H
et al. Selective recognition of cyclic RGD peptides of NMR defined
conformation by alpha IIb beta 3, alpha V beta 3, and alpha 5 beta
1 integrins. J Biol Chem 1994;269:20233-8.
103. Haubner R, Wester HJ, Burkhart F, Senekowitsch-Schmidtke R,
Weber W, Goodman SL et al. Glycosylated RGD-containing peptides: tracer for tumor targeting and angiogenesis imaging with
improved biokinetics. J Nucl Med 2001;42:326-36.
104. Liu S. Radiolabeled multimeric cyclic RGD peptides as integrin
alphavbeta3 targeted radiotracers for tumor imaging. Mol Pharm
2006;3:472-87.
105. Haubner R, Wester HJ, Reuning U, Senekowitsch-Schmidtke R,
Diefenbach B, Kessler H et al. Radiolabeled alpha(v)beta3 integrin antagonists: a new class of tracers for tumor targeting. J Nucl
Med 1999;40:1061-71.
106. Decristoforo C, Faintuch-Linkowski B, Rey A, von Guggenberg
E, Rupprich M, Hernandez-Gonzales I et al. [(99m)Tc]HYNICRGD for imaging integrin alpha(v)beta(3) expression. Nucl Med
Biol 2006;33:945-52.
107. Indrevoll B, Kindberg GM, Karlsen H, Mendizabal M, Morrison M,
Healey A et al. CMR 2005: 3.03: New RGD peptide-based molecular imaging agents for detection of angiogenesis. Contrast
Media Mol Imaging 2006;1:58.
108. Maschauer S, Prante O, Hoffmann M, Deichen JT, Kuwert T.
Characterization of 18F-FDG uptake in human endothelial cells
in vitro. J Nucl Med 2004;45:455-60.
109. Cher LM, Murone C, Lawrentschuk N, Ramdave S, Papenfuss A,
110.
Hannah A et al. Correlation of hypoxic cell fraction and angiogenesis with glucose metabolic rate in gliomas using 18F-fluoromisonidazole, 18F-FDG PET, and immunohistochemical studies. J Nucl Med 2006;47:410-8.
Blankenberg FG, Mandl S, Cao YA, O’Connell-Rodwell C, Contag
C, Mari C et al. Tumor imaging using a standardized radiolabeled adapter protein docked to vascular endothelial growth
factor. J Nucl Med 2004;45:1373-80.
Chan C, Sandhu J, Guha A, Scollard DA, Wang J, Chen P et al. A
human transferrin-vascular endothelial growth factor (hnTf-VEGF)
fusion protein containing an integrated binding site for (111)In
for imaging tumor angiogenesis. J Nucl Med 2005;46:1745-52.
Cai W, Chen K, Mohamedali KA, Cao Q, Gambhir SS, Rosenblum
MG et al. PET of vascular endothelial growth factor receptor
expression. J Nucl Med 2006;47:2048-56.
Backer MV, Levashova Z, Patel V, Jehning BT, Claffey K,
Blankenberg FG et al. Molecular imaging of VEGF receptors in
angiogenic vasculature with single-chain VEGF based probes.
Nat Med. In press 2007.
Li S, Peck-Radosavljevic M, Kienast O, Preitfellner J, Havlik E,
Schima W et al. Iodine-123-vascular endothelial growth factor-165
(123I-VEGF165). Biodistribution, safety and radiation dosimetry
in patients with pancreatic carcinoma. Q J Nucl Med Mol Imaging
2004;48:198-206.
Sosnovik DE, Schellenberger EA, Nahrendorf M, Novikov MS,
Matsui T, Dai G et al. Magnetic resonance imaging of cardiomyocyte apoptosis with a novel magneto-optical nanoparticle. Magn
Reson Med 2005;54:718-24.
Yang SK, Attipoe S, Klausner AP, Tian R, Pan D, Rich TA et al. In
vivo detection of apoptotic cells in the testis using fluorescence
labeled annexin V in a mouse model of testicular torsion. J Urol
2006;176:830-5.
van Tilborg GAF, Mulder WJM, Chin PTK, Storm G,
Reutelingsperger CP, Nicolay K et al. Annexin A5-Conjugated
Quantum Dots with a Paramagnetic Lipidic Coating for the
Multimodal Detection of Apoptotic Cells. Bioconjugate Chem.
2006;17;865-868.
Cauchon N, Langlois R, Rousseau JA, Tessier G, Cadorette J,
Lecomte R, Hunting DJ, Pavan RA, Zeisler SK, van Lier JE. PET
imaging of apoptosis with (64)Cu-labeled streptavidin following
pretargeting of phosphatidylserine with biotinylated annexin-V.
Eur J Nucl Med Mol Imaging 2007;34:247-58.
Luo QY, Zhang ZY, Wang F, Lu HK, Guo YZ, Zhu RS. Preparation,
in vitro and in vivo evaluation of 99mTc-Annexin B1: a novel
radioligand for apoptosis imaging. Biochem Biophys Res Commun
2005;335:1102-6.
Laumonier C, Segers J, Laurent S, Michel A, Coppee F, Belayew
A et al. A new peptidic vector for molecular imaging of apoptosis, identified by phage display technology. J Biomol Screen
2006;11: 537-45.
Bose S, Tuunainen I, Parry M, Medina OP, Mancini G, Kinnunena
PKJ. Binding of cationic liposomes to apoptotic cells. Anal
Biochem 2004;331:385-94.
Quinti L, Weissleder R, Tung CH. A fluorescent nanosensor for
apoptotic cells. Nano Lett 2006;6:488-90.
Hanshaw RG, Smith BD. New reagents for phosphatidylserine
recognition and detection of apoptosis. Bioorg Med Chem
2005;13:5035-42.
Zhao M, Zhu X, Ji S, Zhou J, Ozker KS, Fang W et al. 99mTclabeled C2A domain of synaptotagmin I as a target-specific molecular probe for noninvasive imaging of acute myocardial infarction. J Nucl Med 2006;47:1367-74.
Bullok K, Piwnica-Worms D. Synthesis and characterization of a
small, membrane-permeant, caspase-activatable far-red fluorescent
peptide for imaging apoptosis. J Med Chem 2005;48:5404-7.
Zhou D, Chu W, Rothfuss J, Zeng C, Xu J, Jones L et al. Synthesis,
radiolabeling, and in vivo evaluation of an 18F-labeled isatin analog for imaging caspase-3 activation in apoptosis. Bioorg Med
Chem Lett 2006;16:5041-6.
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
108
111.
112.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
125.
126.
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
June 2007
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
127. Boersma HH, Kietselaer BL, Stolk LM, Bennaghmouch A, Hofstra
L, Narula J et al. Past, present, and future of annexin A5: from protein discovery to clinical applications. J Nucl Med 2005;46:203550.
128. D’Arceuil H, Rhine W, de Crespigny A, Yenari M, Tait JF, Strauss
WH et al. 99mTc annexin V imaging of neonatal hypoxic brain
injury. Stroke 2000;31:2692-700.
129. Kenis H, van Genderen H, Bennaghmouch A, Rinia HA, Frederik
P, Narula J et al. Cell surface-expressed phosphatidylserine and
annexin A5 open a novel portal of cell entry. J Biol Chem
2004;279:52623-9.
130. Stratton JR, Dewhurst TA, Kasina S, Reno JM, Cerqueira MD,
Baskin DG et al. Selective uptake of radiolabeled annexin V on
acute porcine left atrial thrombi. Circulation 1995;92:3113-21.
131. Narula J, Acio ER, Narula N, Samuels LE, Fyfe B, Wood D et al.
Annexin-V imaging for noninvasive detection of cardiac allograft
rejection. Nat Med 2001;7:1347-52.
132. Belhocine T, Steinmetz N, Hustinx R, Bartsch P, Jerusalem G,
Seidel L et al. Increased uptake of the apoptosis-imaging agent
(99m)Tc recombinant human Annexin V in human tumors after
one course of chemotherapy as a predictor of tumor response and
patient prognosis. Clin Cancer Res 2002;8:2766-74.
133. Kemerink GJ, Liu X, Kieffer D, Ceyssens S, Mortelmans L,
Verbruggen AM et al. Safety, biodistribution, and dosimetry of
99mTc-HYNIC-annexin V, a novel human recombinant annexin v
for human application. J Nucl Med 2003;44:947-52.
134. Boersma HH, Liem IH, Kemerink GJ, Thimister PW, Hofstra L,
Stolk LM et al. Comparison between human pharmacokinetics and
imaging properties of two conjugation methods for 99mTc-annexin A5. Br J Radiol 2003;76:553-60.
135. Blankenberg FG. Molecular imaging: the latest generation of
contrast agents and tissue characterization techniques. J Cell
Biochem 2003;90:443-53.
136. Haas RL, de Jong D, Valdes Olmos RA, Hoefnagel CA, van den
Heuvel I, Zerp SF et al. In vivo imaging of radiation-induced
apoptosis in follicular lymphoma patients. Int J Radiat Oncol
Biol Phys 2004;59:782-7.
137. Vermeersch H, Ham H, Rottey S, Lahorte C, Corsetti F, Dierckx R
et al. Intraobserver, interobserver, and day-to-day reproducibility of quantitative 99mTc-HYNIC annexin-V imaging in head and
neck carcinoma. Cancer Biother Radiopharm 2004;19:205-10.
138. Kartachova M, Haas RL, Olmos RA, Hoebers FJ, van Zandwijk N,
Verheij M. In vivo imaging of apoptosis by 99mTc-Annexin V
scintigraphy: visual analysis in relation to treatment response.
Radiother Oncol 2004;72:333-9.
139. Rottey S, Slegers G, Van Belle S, Goethals I, Van de Wiele C.
Sequential 99mTc-hydrazinonicotinamide-annexin V imaging for predicting response to chemotherapy. J Nucl Med 2006;47:1813-8.
140. Thimister PW, Hofstra L, Liem IH, Boersma HH, Kemerink G,
Reutelingsperger CP et al. In vivo detection of cell death in the area
at risk in acute myocardial infarction. J Nucl Med 2003;44:391-6.
141. Narula J, Strauss HW. Invited commentary: P.S.* I love you: implications of phosphatidyl serine (PS) reversal in acute ischemic
syndromes. J Nucl Med 2003;44:397-9.
142. Rongen GA, Oyen WJ, Ramakers BP, Riksen NP, Boerman OC,
Steinmetz N et al. Annexin A5 scintigraphy of forearm as a novel in vivo model of skeletal muscle preconditioning in humans.
Circulation 2005;111:173-8.
143. Riksen NP, Oyen WJ, Ramakers BP, Van den Broek PH, Engbersen
R, Boerman OC et al. Oral therapy with dipyridamole limits
ischemia-reperfusion injury in humans. Clin Pharmacol Ther
2005;78:52-9.
144. Riksen NP, Zhou Z, Oyen WJ, Jaspers R, Ramakers BP, Brouwer
RM et al. Caffeine prevents protection in two human models of
ischemic preconditioning. J Am Coll Cardiol 2006;48:700-7.
145. Kiestelaer BLJH, Reutelingsperger CPM, Heidendal GAK, Daemen
MJAP, Mess WH, Hofstra L. Noninvasive detection of plaque
instability with use of radiolabeled annexin A5 in patients with
carotid-artery atherosclerosis. N Engl J Med 2004;350:1472-3.
BLANKENBERG
146. Blankenberg FG, Kalinyak J, Liu L, Koike M, Cheng D, Goris ML
et al. 99mTc-HYNIC-annexin V SPECT imaging of acute stroke and
its response to neuroprotective therapy with anti-Fas ligand antibody. Eur J Nucl Med Mol Imaging 2006;33:566-74.
147. Lorberboym M, Blankenberg FG, Sadeh M, Lampl Y. In vivo
imaging of apoptosis in patients with acute stroke: correlation with
blood-brain barrier permeability. Brain Res 2006;1103:13-9.
148. Lampl Y, Lorberboym M, Blankenberg FG, Sadeh M, Gilad R.
Annexin V SPECT imaging of phosphatidylserine expression in
patients with dementia. Neurology 2006;66:1253-4.
149. Tait JF, Brown DS, Gibson DF, Blankenberg FG, Strauss HW.
Development and characterization of annexin V mutants with
endogenous chelation sites for (99m)Tc. Bioconjug Chem
2000;11:918-25.
150. Jin M, Smith C, Hsieh HY, Gibson DF, Tait JF. Essential role of Bhelix calcium binding sites in annexin V-membrane binding. J Biol
Chem 2004;279:40351-7.
151. Tait JF, Smith C, Blankenberg FG. Structural requirements for in
vivo detection of cell death with 99mTc-annexin V. J Nucl Med.
2005;46: 807-15.
152. Grierson JR, Yagle KJ, Eary JF, Tait JF, Gibson DF, Lewellen B et
al. Production of [F-18]fluoroannexin for imaging apoptosis with
PET. Bioconjug Chem 2004;15:373-9.
153. Murakami Y, Takamatsu H, Taki J, Tatsumi M, Noda A, Ichise R
et al. 18F-labelled annexin V: a PET tracer for apoptosis imaging.
Eur J Nucl Med Mol Imaging 2004;31:469-74.
154. Biancone L, Schillaci O, Capoccetti F, Bozzi RM, Fina D,
Petruzziello C et al. Technetium-99m-HMPAO labeled leukocyte
single photon emission computerized tomography (SPECT) for
assessing Crohn’s disease extent and intestinal infiltration. Am J
Gastroenterol 2005;100:344-54.
155. Bunyaviroch T, Aggarwal A, Oates ME. Optimized scintigraphic
evaluation of infection and inflammation: role of single-photon
emission computed tomography/computed tomography fusion
imaging. Semin Nucl Med 2006;36:295-311.
156. Buscombe J, Signore A. FDG-PET in infectious and inflammatory disease. Eur J Nucl Med Mol Imaging 2003;30:1571-3.
157. Chen DL, Schuster DP. Imaging pulmonary inflammation with
positron emission tomography: a biomarker for drug development. Mol Pharm 2006;3:488-95.
158. Bar-Shalom R, Yefremov N, Guralnik L, Keidar Z, Engel A, Nitecki
S et al. SPECT/CT using 67Ga and 111In-labeled leukocyte scintigraphy for diagnosis of infection. J Nucl Med 2006;47:587-94.
159. Pellegrino D, Bonab AA, Dragotakes SC, Pitman JT, Mariani G,
Carter EA. Inflammation and infection: imaging properties of
18F-FDG-labeled white blood cells versus 18F-FDG. J Nucl Med
2005;46:1522-30.
160. Dumarey N, Egrise D, Blocklet D, Stallenberg B, Remmelink M,
del Marmol V et al. Imaging infection with 18F-FDG-labeled
leukocyte PET/CT: initial experience in 21 patients. J Nucl Med
2006;47:625-32.
161. Signore A, Chianelli M, D’Alessandria C, Annovazzi A. Receptor
targeting agents for imaging inflammation/infection: where are we
now? Q J Nucl Med Mol Imaging 2006;50:236-42.
162. Love C, Tronco GG, Palestro CJ. Imaging of infection and inflammation with 99mTc-Fanolesomab. Q J Nucl Med Mol Imaging
2006;50:113-20.
163. Gross MD, Shapiro B, Fig LM, Steventon R, Skinner RW, Hay RV.
Imaging of human infection with (131)I-labeled recombinant
human interleukin-8. J Nucl Med 2001;42:1656-9.
164. Amartey JK, Esguerra C, Al-Otaibi B, Al-Jammaz I, Al-Qahtani
M, Parhar RS. Prosthetic radioiodination of interleukin-8
([(123/131)I]-IL-8): biological behavior in a mouse infection model. Appl Radiat Isot 2005;62:39-47.
165. van der Laken CJ, Boerman OC, Oyen WJ, van de Ven MT, van
der Meer JW, Corstens FH. Imaging of infection in rabbits with
radioiodinated interleukin-1 (alpha and beta), its receptor antagonist and a chemotactic peptide: a comparative study. Eur J Nucl
Med 1998;25:347-52.
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
Vol. 51 - No. 2
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
109
BLANKENBERG
NUCLEAR MEDICINE APPLICATIONS IN MOLECULAR IMAGING
166. Barrera P, van der Laken CJ, Boerman OC, Oyen WJ, van de Ven
MT, van Lent PL et al. Radiolabelled interleukin-1 receptor antagonist for detection of synovitis in patients with rheumatoid arthritis. Rheumatology (Oxford) 2000;39:870-4.
167. Signore A, Chianelli M, Annovazzi A, Bonanno E, Spagnoli LG,
Pozzilli P et al. 123I-interleukin-2 scintigraphy for in vivo assessment of intestinal mononuclear cell infiltration in Crohn’s disease. J Nucl Med 2000;41:242-9.
168. Moyer BR, Vallabhajosula S, Lister-James J, Bush LR, Cyr JE, Snow
DA et al. Technetium-99m-white blood cell-specific imaging
agent developed from platelet factor 4 to detect infection. J Nucl
Med 1996;37:673-9.
169. Rennen HJ, Frielink C, Brandt E, Zaat SA, Boerman OC, Oyen WJ
et al. Relationship between neutrophil-binding affinity and suitability for infection imaging: comparison of (99m)Tc-labeled
NAP-2 (CXCL-7) and 3 C-terminally truncated isoforms. J Nucl Med
2004;45:1217-23.
170. van Eerd JE, Broekema M, Harris TD, Edwards DS, Oyen WJ,
Corstens FH et al. Imaging of infection and inflammation with an
improved 99mTc-labeled LTB4 antagonist. J Nucl Med
2005;46:1546-51.
171. Signore A, Chianelli M, Annovazzi A, Rossi M, Maiuri L, Greco M
et al. Imaging active lymphocytic infiltration in coeliac disease with
iodine-123-interleukin-2 and the response to diet. Eur J Nucl
Med 2000;27:18-24.
172. Annovazzi A, Biancone L, Caviglia R, Chianelli M, Capriotti G,
Mather SJ et al. 99mTc-interleukin-2 and (99m)Tc-HMPAO granulocyte scintigraphy in patients with inactive Crohn’s disease.
Eur J Nucl Med Mol Imaging 2003;30:374-82.
173. Signore A, Picarelli A, Annovazzi A, Britton KE, Grossman AB,
Bonanno E et al. 123I-Interleukin-2: biochemical characterization and in vivo use for imaging autoimmune diseases. Nucl Med
Commun 2003;24:305-16.
174. Annovazzi A, Bonanno E, Arca M, D’Alessandria C, Marcoccia A,
Spagnoli LG et al. 99mTc-interleukin-2 scintigraphy for the in
vivo imaging of vulnerable atherosclerotic plaques. Eur J Nucl Med
Mol Imaging 2006;33:117-26.
175. Rennen HJ, van Eerd JE, Oyen WJ, Corstens FH, Edwards DS,
Boerman OC. Effects of coligand variation on the in vivo characteristics of Tc-99m-labeled interleukin-8 in detection of infection. Bioconjug Chem 2002;13:370-7.
176. Rennen HJ, Boerman OC, Oyen WJ, Corstens FH. Kinetics of
99mTc-labeled interleukin-8 in experimental inflammation and
infection. J Nucl Med 2003;44:1502-9.
177. Rennen HJ, Bleeker-Rovers CP, van Eerd JE, Frielink C, Oyen WJ,
Corstens FH et al. 99mTc-labeled interleukin-8 for scintigraphic
detection of pulmonary infections. Chest 2004;126:1954-61.
178. Rubello D, Casara D, Maran A, Avogaro A, Tiengo A, Muzzio
PC. Role of anti-granulocyte Fab’ fragment antibody scintigraphy (LeukoScan) in evaluating bone infection: acquisition protocol, interpretation criteria and clinical results. Nucl Med Commun
2004;25:39-47.
179. Kapsoritakis AN, Koutroubakis IE, Kouroumalis EA, Koukouraki
SI, Karkavitsas N. (99m)Tc-Leucoscan in the evaluation of inflammatory bowel disease. Eur J Nucl Med Mol Imaging 2002;29:1098.
180. Rollins BJ. Monocyte chemoattractant protein-1:a potential regulator of monocyte recruitment in inflammatory disease. Mol
Med Today 1996;2:198-204.
181. Ohtsuki K, Hayase M, Akashi K, Kopiwoda S, Strauss HW.
Detection of monocyte chemoattractant protein-1 receptor expression in experimental atherosclerotic lesions: an autoradiographic study. Circulation 2001;104:203-8.
182. Blankenberg FG, Tait JF, Blankenberg TA, Post AM, Strauss HW.
Imaging macrophages and the apoptosis of granulocytes in a
rodent model of subacute and chronic abscesses with radiolabeled
monocyte chemotactic peptide-1 and annexin V. Eur J Nucl Med
2001;28:1384-93.
183. Lupetti A, Nibbering PH, Welling MM, Pauwels EK. Radiopharmaceuticals: new antimicrobial agents. Trends Biotechnol 2003;21:
70-3.
184. Akhtar MS, Qaisar A, Irfanullah J, Iqbal J, Khan B, Jehangir M et
al. Antimicrobial peptide 99mTc-ubiquicidin 29-41 as human
infection-imaging agent: clinical trial. J Nucl Med 2005;46:567-73.
185. Siaens R, Eijsink VG, Vaaje-Kolstad G, Vandenbulcke K,
Cornelissen B, Cuvelier C et al. Synthesis and evaluation of a
99mTechnetium labeled chitin-binding protein as potential specific radioligand for the detection of fungal infections in mice. Q
J Nucl Med Mol Imaging 2006;50:155-66.
186. Benitez A, Roca M, Martin-Comin J. Labeling of antibiotics for
infection diagnosis. Q J Nucl Med Mol Imaging 2006;50:147-52.
187. Savill J. Apoptosis in resolution of inflammation. J Leukoc Biol
1997;61:375-80.
188. Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RAB,
Henson PM. A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature 2000;405:85-90.
189. Brown SB, Savill J. Phagocytosis triggers macrophage release of
Fas ligand and induces apoptosis of bystander leukocytes. J
Immunol 1999;162:480-5.
190. Blankenberg FG, Tait JF, Blankenberg TA, Post AM, Strauss HW.
Imaging macrophages and the apoptosis of granulocytes in a
rodent model of subacute and chronic abscesses with radiolabeled
monocyte chemotactic peptide-1 and annexin V. Eur J Nucl Med
2001;28:1384-93.
191. Kietselaer BL, Reutelingsperger CP, Heidendal GA, Daemen MJ,
Mess WH, Hofstra L et al. Noninvasive detection of plaque instability with use of radiolabeled annexin A5 in patients with carotidartery atherosclerosis. N Engl J Med 2004;350:1472-3.
192. Hartung D, Sarai M, Petrov A, Kolodgie F, Narula N, Verjans J et
al. Resolution of apoptosis in atherosclerotic plaque by dietary
modification and statin therapy. J Nucl Med 2005;46:2051-6.
193. Isobe S, Tsimikas S, Zhou J, Fujimoto S, Sarai M, Branks MJ et al.
Noninvasive imaging of atherosclerotic lesions in apolipoprotein E-deficient and low-density-lipoprotein receptor-deficient
mice with annexin A5. J Nucl Med 2006;47:1497-505.
A
C
I
D
E
M ®
T
A
V
H
R
G
I
E R
N
I
Y
M OP
C
110
THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING
June 2007