Molecular imaging for visualization and quantification of ... - Philips

Molecular imaging for visualization and quantification of ... - Philips Molecular imaging for visualization and quantification of ... - Philips

incenter.medical.philips.com
from incenter.medical.philips.com More from this publisher
03.08.2013 Views

Investigations and research Molecular imaging for visualization and quantification of individualized targeted cancer therapy M. Lubberink S.N.F. Rizvi O.S. Hoekstra G.A.M.S. van Dongen Targeted therapy and imaging Recent advances in molecular biology have facilitated the identification of molecular targets on tumor cells, such as those involved in proliferation, differentiation, apoptosis, and angiogenesis. This has boosted the design of targeted pharmaceuticals, with monoclonal antibodies (MAbs) forming the largest category. Examples of MAbs used in targeted therapy include: • Trastuzumab (Herceptin ® ) directed against the HER-2 receptor and approved for treatment of metastatic breast cancer • Cetuximab (Erbitux ® ) and panitumumab (Vectibix ® ) directed against the epidermal growth factor receptor (EGFR) and approved for treatment of e.g. colorectal cancer • Bevacizumab (avastin ) directed against the vascular endothelial growth factor (VEGF) and approved for treatment of colorectal cancer and non-small cell lung cancer • Rituximab (Rituxan ® ) and 90 Y-ibritumomab tiuxetan (Zevalin ® ; see Figure 1) directed against CD20 and approved for treatment of non-Hodgkin’s lymphoma. These MAbs may be used as monotherapy, but more often in combination with chemo- or radiotherapy. Hundreds of new MAbs are under development. The global market for MAbs is expected to triple between 2005 and 2010, to 20-30 billion US dollars. Targeted therapies are highly expensive and will generally be beneficial to only a subgroup of patients, depending on such factors as over-expression of the target, differences in metabolism, and variability and heterogeneity of tumor uptake. In order to understand the efficacy of a certain targeted drug in an individual patient, its uptake in the tumor and normal tissues should be assessed during a scouting procedure prior to the start of therapy or immediately upon the start of a course of therapy. Response to therapy is usually addressed by anatomical (CT) imaging using RECIST criteria. Department of Nuclear Medicine and PET Research, VU Medical Center, Amsterdam, the Netherlands. Departments of Nuclear Medicine and PET Research, and Otolaryngology/ Head and Neck Surgery, VU Medical Center, Amsterdam, the Netherlands. However, anatomical changes to treatment are usually not visible during the earliest stages of therapy, and several cycles of therapy are given before response can be assessed. Targeted therapeutic agents have compromised the association of volume-based measures and patient outcomes even further. There is increasing evidence that using PET with a metabolic tracer such as 18 F-fluorodeoxyglucose (FDG) shows changes in tumor glucose metabolism before anatomical changes are visible. However, even the use of FDG-PET for response monitoring still requires one or two cycles of therapy before the efficacy of the therapy can be evaluated. Measurement of tumor uptake of the targeted drug, labeled with a positron-emitting nuclide, may provide a predictor of treatment efficacy even before the start of therapy. A PET study with the labeled drug could then be used to select in advance those patients who will benefit from a certain targeted therapy, avoiding unnecessary administration of expensive targeted drugs and avoiding delay in effective treatment for the patient. This, of course, assumes that the kinetics of a tracer amount of the drug are similar to those of pharmacological dosages, so that a tracer study can predict pharmacological distribution of the tracer, and, equally important, that the uptake of the drug in the tumor is an accurate measure of treatment efficacy. Quantitative imaging of targeted drugs is also of value in drug development. Information on the optimal dosage, the uptake in critical organs, and interpatient variations in kinetics and targeting can be obtained at early stages during drug development, thereby allowing for early selection of promising drug candidates and reducing development costs [1]. Labeling of the targeted drug with a positronemitting isotope allows for quantitative PET imaging of the distribution and kinetics of the drug both prior to and during therapy. The radioactive half-lives of the most commonly used PET isotopes, 18 F and 11 C, are 110 and 20.4 min, E E Recent advances in molecular biology have facilitated the identification of tumor cells. Quantitative imaging of targeted drugs is also of value in drug development. MEDICAMUNDI 54/2 2010 41

Investigations <strong>and</strong> research<br />

<strong>Molecular</strong> <strong>imaging</strong> <strong>for</strong> <strong>visualization</strong> <strong>and</strong> <strong>quantification</strong><br />

<strong>of</strong> individualized targeted cancer therapy<br />

M. Lubberink<br />

S.N.F. Rizvi<br />

O.S. Hoekstra<br />

G.A.M.S. van Dongen<br />

Targeted therapy <strong>and</strong> <strong>imaging</strong><br />

Recent advances in molecular biology have<br />

facilitated the identification <strong>of</strong> molecular targets<br />

on tumor cells, such as those involved in<br />

proliferation, differentiation, apoptosis, <strong>and</strong><br />

angiogenesis. This has boosted the design <strong>of</strong><br />

targeted pharmaceuticals, with monoclonal<br />

antibodies (MAbs) <strong>for</strong>ming the largest category.<br />

Examples <strong>of</strong> MAbs used in targeted therapy<br />

include:<br />

• Trastuzumab (Herceptin ® ) directed against<br />

the HER-2 receptor <strong>and</strong> approved <strong>for</strong> treatment<br />

<strong>of</strong> metastatic breast cancer<br />

• Cetuximab (Erbitux ® ) <strong>and</strong> panitumumab<br />

(Vectibix ® ) directed against the epidermal<br />

growth factor receptor (EGFR) <strong>and</strong> approved<br />

<strong>for</strong> treatment <strong>of</strong> e.g. colorectal cancer<br />

• Bevacizumab (avastin ) directed against the<br />

vascular endothelial growth factor (VEGF)<br />

<strong>and</strong> approved <strong>for</strong> treatment <strong>of</strong> colorectal cancer<br />

<strong>and</strong> non-small cell lung cancer<br />

• Rituximab (Rituxan ® ) <strong>and</strong> 90 Y-ibritumomab<br />

tiuxetan (Zevalin ® ; see Figure 1) directed<br />

against CD20 <strong>and</strong> approved <strong>for</strong> treatment <strong>of</strong><br />

non-Hodgkin’s lymphoma.<br />

These MAbs may be used as monotherapy, but<br />

more <strong>of</strong>ten in combination with chemo- or<br />

radiotherapy. Hundreds <strong>of</strong> new MAbs are under<br />

development. The global market <strong>for</strong> MAbs is<br />

expected to triple between 2005 <strong>and</strong> 2010, to<br />

20-30 billion US dollars. Targeted therapies are<br />

highly expensive <strong>and</strong> will generally be beneficial<br />

to only a subgroup <strong>of</strong> patients, depending on<br />

such factors as over-expression <strong>of</strong> the target,<br />

differences in metabolism, <strong>and</strong> variability <strong>and</strong><br />

heterogeneity <strong>of</strong> tumor uptake. In order to<br />

underst<strong>and</strong> the efficacy <strong>of</strong> a certain targeted drug<br />

in an individual patient, its uptake in the tumor<br />

<strong>and</strong> normal tissues should be assessed during a<br />

scouting procedure prior to the start <strong>of</strong> therapy or<br />

immediately upon the start <strong>of</strong> a course <strong>of</strong> therapy.<br />

Response to therapy is usually addressed by<br />

anatomical (CT) <strong>imaging</strong> using RECIST criteria.<br />

Department <strong>of</strong> Nuclear Medicine <strong>and</strong> PET Research, VU Medical Center,<br />

Amsterdam, the Netherl<strong>and</strong>s.<br />

Departments <strong>of</strong> Nuclear Medicine <strong>and</strong> PET Research, <strong>and</strong> Otolaryngology/<br />

Head <strong>and</strong> Neck Surgery, VU Medical Center, Amsterdam, the Netherl<strong>and</strong>s.<br />

However, anatomical changes to treatment are<br />

usually not visible during the earliest stages <strong>of</strong><br />

therapy, <strong>and</strong> several cycles <strong>of</strong> therapy are given<br />

be<strong>for</strong>e response can be assessed. Targeted<br />

therapeutic agents have compromised the<br />

association <strong>of</strong> volume-based measures <strong>and</strong> patient<br />

outcomes even further. There is increasing<br />

evidence that using PET with a metabolic tracer<br />

such as 18 F-fluorodeoxyglucose (FDG) shows<br />

changes in tumor glucose metabolism be<strong>for</strong>e<br />

anatomical changes are visible. However, even<br />

the use <strong>of</strong> FDG-PET <strong>for</strong> response monitoring<br />

still requires one or two cycles <strong>of</strong> therapy be<strong>for</strong>e<br />

the efficacy <strong>of</strong> the therapy can be evaluated.<br />

Measurement <strong>of</strong> tumor uptake <strong>of</strong> the targeted<br />

drug, labeled with a positron-emitting nuclide,<br />

may provide a predictor <strong>of</strong> treatment efficacy<br />

even be<strong>for</strong>e the start <strong>of</strong> therapy. A PET study<br />

with the labeled drug could then be used to<br />

select in advance those patients who will benefit<br />

from a certain targeted therapy, avoiding<br />

unnecessary administration <strong>of</strong> expensive targeted<br />

drugs <strong>and</strong> avoiding delay in effective treatment<br />

<strong>for</strong> the patient. This, <strong>of</strong> course, assumes that<br />

the kinetics <strong>of</strong> a tracer amount <strong>of</strong> the drug are<br />

similar to those <strong>of</strong> pharmacological dosages, so<br />

that a tracer study can predict pharmacological<br />

distribution <strong>of</strong> the tracer, <strong>and</strong>, equally important,<br />

that the uptake <strong>of</strong> the drug in the tumor is an<br />

accurate measure <strong>of</strong> treatment efficacy.<br />

Quantitative <strong>imaging</strong> <strong>of</strong> targeted drugs is also <strong>of</strong><br />

value in drug development. In<strong>for</strong>mation on the<br />

optimal dosage, the uptake in critical organs, <strong>and</strong><br />

interpatient variations in kinetics <strong>and</strong> targeting<br />

can be obtained at early stages during drug<br />

development, thereby allowing <strong>for</strong> early selection<br />

<strong>of</strong> promising drug c<strong>and</strong>idates <strong>and</strong> reducing<br />

development costs [1].<br />

Labeling <strong>of</strong> the targeted drug with a positronemitting<br />

isotope allows <strong>for</strong> quantitative PET<br />

<strong>imaging</strong> <strong>of</strong> the distribution <strong>and</strong> kinetics <strong>of</strong> the<br />

drug both prior to <strong>and</strong> during therapy. The<br />

radioactive half-lives <strong>of</strong> the most commonly used<br />

PET isotopes, 18 F <strong>and</strong> 11 C, are 110 <strong>and</strong> 20.4 min,<br />

E<br />

E<br />

Recent advances in<br />

molecular biology<br />

have facilitated the<br />

identification <strong>of</strong> tumor<br />

cells.<br />

Quantitative <strong>imaging</strong><br />

<strong>of</strong> targeted drugs is<br />

also <strong>of</strong> value in drug<br />

development.<br />

MEDICAMUNDI 54/2 2010 41


E<br />

Table 1. Positron-emitting isotopes<br />

<strong>for</strong> labeling <strong>of</strong> monoclonal<br />

antibodies<br />

1<br />

G<br />

Figure 1. [ 18F]FDG (left) <strong>and</strong> 6 days<br />

p.i. [ 89Zr]ibritumomab tiuxetan<br />

([ 89Zr]Zevalin) PET image <strong>of</strong> a patient<br />

with Non-Hodgkin lymphoma showing<br />

high Zevalin uptake in parailiac lymph<br />

nodes. FDG uptake in the lymph<br />

nodes is only moderate (SUV 2).<br />

Images were acquired on a Gemini<br />

TF-64 PET-CT scanner (<strong>Philips</strong><br />

Healthcare).<br />

42 MEDICAMUNDI 54/2 2010<br />

respectively. This is too short to measure the<br />

kinetics <strong>of</strong> intact MAbs.<br />

The longer half-life positron emitters such as<br />

66 Ga , 64 Cu [2], 86 Y, 76 Br [3], 89 Zr [4-6] <strong>and</strong> 124 I<br />

[7, 8] have been used <strong>for</strong> labeling <strong>of</strong> MAbs,<br />

MAb fragments <strong>and</strong> peptides [4-6, 9-15] <strong>and</strong> a<br />

number <strong>of</strong> mainly 124 I-labeled tracers have been<br />

suggested <strong>for</strong> measuring apoptosis [16, 17], insulin<br />

receptors [18], hypoxia [19], <strong>and</strong> proliferation<br />

[20]. The half lives <strong>of</strong> these isotopes are shown<br />

in Table 1.<br />

Isotope Half-life<br />

66 Ga 9.5 h<br />

64 Cu 12.7 h<br />

86 Y 14.7 h<br />

76 Br 16.7 h<br />

89 Zr 78 h<br />

124 I 100 h<br />

Targeted therapy with<br />

radionuclides<br />

Several radiopharmaceuticals have gained routine<br />

acceptance <strong>for</strong> radionuclide therapy, with 131 I-iodide<br />

<strong>for</strong> the treatment <strong>for</strong> thyroid cancer being the<br />

best known example [21].<br />

Other examples are the use <strong>of</strong> 131 I-labeled MIBG<br />

(meta-iodobenzylguanidine) <strong>for</strong> treatment <strong>of</strong><br />

neuroblastoma, 90 Y, 177 Lu or 111 In-labeled<br />

somatostatin analogues <strong>for</strong> treatment <strong>of</strong><br />

neuroendocrine tumors [22], <strong>and</strong> palliative or<br />

adjuvant treatment <strong>of</strong> skeletal metastases, <strong>for</strong><br />

example using 89 Sr-chloride [23]. Several MAbs<br />

labeled with 90 Y or 131 I, such as 90 Y-labeled<br />

ibritumomab <strong>and</strong> 131 I-labeled tositumomab<br />

(Bexxar ® ) have been approved <strong>for</strong> (radio-)<br />

immunotherapy [24].<br />

The biological effect <strong>of</strong> therapy with radionuclides<br />

is due to the deposition <strong>of</strong> energy <strong>of</strong> ionizing<br />

radiation per unit mass <strong>of</strong> tissue, i.e. the absorbed<br />

dose. This is a well-defined physical quantity,<br />

unlike those <strong>of</strong> other systemic treatments such<br />

as chemotherapy [25]. Although absorbed dose<br />

alone might not be sufficient to fully predict<br />

response to radionuclide therapy, with dose rate,<br />

type <strong>of</strong> radiation, <strong>and</strong> biological characteristics<br />

<strong>of</strong> targeted drug, tumor <strong>and</strong> normal organs being<br />

additional factors that can affect response,<br />

knowledge <strong>of</strong> absorbed doses is required <strong>for</strong> an<br />

optimal application <strong>of</strong> radionuclide therapy.<br />

The amount <strong>of</strong> radioactivity injected should be<br />

chosen such that toxicity to the dose-limiting<br />

organ (usually red marrow in the case <strong>of</strong><br />

antibodies, <strong>and</strong> the kidneys in the case <strong>of</strong> smaller<br />

molecules such as peptides) is limited, while<br />

maximizing tumor absorbed dose.<br />

Internal emitter dosimetry<br />

In current clinical practice in targeted<br />

radiotherapy, radioactivity is usually administered<br />

as a fixed amount, or at best as a function <strong>of</strong><br />

patient size. Absorbed dose calculations, if done<br />

at all, are generally per<strong>for</strong>med on a total body or<br />

region <strong>of</strong> interest basis. The necessary parameters<br />

<strong>for</strong> internal emitter dosimetry are the number<br />

<strong>of</strong> decays in each tissue <strong>of</strong> interest or voxel<br />

(cumulative activity) <strong>and</strong> the geometry <strong>of</strong> the<br />

patient which defines the transport <strong>of</strong> radiation.<br />

Generally three to four measurements <strong>of</strong> the<br />

radioactivity distribution can be made (Figure 2),<br />

after which a (dual-)exponential fit through these<br />

few data points is used to calculate the cumulative<br />

activity in each tissue <strong>of</strong> interest, <strong>and</strong> absorbed<br />

dose calculations are made using the MIRD<br />

(Medical Internal Radiation Dose Committee)<br />

approach applying a st<strong>and</strong>ardized geometry<br />

[26, 27].<br />

In more advanced methods, the geometry used<br />

<strong>for</strong> absorbed dose calculation is based on a CT<br />

image <strong>of</strong> the patient involved. The absorbed dose<br />

in organ or tumor due to radiation originating<br />

from another organ or tumor is then calculated<br />

using voxel S-values [28], dose point kernel<br />

methods [29] or Monte-Carlo simulations [30-34]<br />

(Figure 3).<br />

The main error sources in absorbed dose<br />

calculations are the use <strong>of</strong> a st<strong>and</strong>ardized<br />

geometry, the computation <strong>of</strong> the cumulative<br />

activity, <strong>and</strong> the inaccuracy <strong>of</strong> tissue radioactivity<br />

measurements with single photon <strong>imaging</strong> [35].


2a<br />

2b<br />

The distribution <strong>of</strong> most radionuclides used to<br />

deliver the absorbed dose in targeted therapy<br />

with radionuclides can either not be imaged at<br />

all ( 90 Y, alpha-emitters) or can only be imaged<br />

with limited accuracy using single photon<br />

scintigraphy ( 177 Lu, 131 I). Here, a positronemitting<br />

analogue <strong>of</strong> the therapeutic nuclide can<br />

be used, if available, such as 86 Y <strong>for</strong> 90 Y [13],<br />

124 I <strong>for</strong> 131 I, 110m In <strong>for</strong> 111 In [37], or 83 Sr <strong>for</strong> 89 Sr<br />

[23], but otherwise an isotope with similar<br />

chemical properties may be used. An example<br />

<strong>of</strong> this is the use <strong>of</strong> 89 Zr as surrogate <strong>for</strong> 90 Y, which<br />

may be preferred to 86 Y since 89 Zr has more<br />

favorable decay characteristics <strong>for</strong> <strong>imaging</strong> <strong>and</strong>,<br />

due to its longer half-life, covers more <strong>of</strong> the<br />

kinetics <strong>of</strong> 90 Y [6]. The most straight<strong>for</strong>ward<br />

example here is the use <strong>of</strong> 124 I-iodide as an<br />

analogue <strong>of</strong> 131 I-iodide in thyroid cancer <strong>imaging</strong>.<br />

In the case <strong>of</strong> thyroid cancer therapy dosimetry,<br />

which is a relatively straight<strong>for</strong>ward example<br />

because only a small part <strong>of</strong> the body needs to be<br />

imaged <strong>and</strong> used <strong>for</strong> dose calculations, this has<br />

even been done on a voxel-by-voxel basis yielding<br />

absorbed dose images [38-41] (Figure 3).<br />

Quantitative PET with long-lived<br />

isotopes<br />

Figure 4 shows simplified decay schemes <strong>of</strong> 124 I,<br />

89 Zr <strong>and</strong>, <strong>for</strong> comparison, 18 F. In contrast with<br />

the pure positron emitters 18 F <strong>and</strong> 11 C, the<br />

longer-lived isotopes mentioned above all emit<br />

gamma radiation in addition to positrons. This<br />

gamma radiation is <strong>of</strong>ten emitted simultaneously<br />

with positrons, referred to as “prompt gamma<br />

radiation”. In the decay <strong>of</strong> 124I, <strong>for</strong> example,<br />

about 50% <strong>of</strong> all positrons (β + in Figure 4) are<br />

1<br />

emitted simultaneously with a 603 keV gamma<br />

photon (γ 1 ). This additional gamma radiation<br />

challenges quantitative image acquisition <strong>and</strong><br />

image quality in a number <strong>of</strong> ways.<br />

3<br />

Detection <strong>of</strong> essentially true coincidences <strong>of</strong><br />

these prompt gamma photons with each other<br />

or with annihilation photons introduces a bias<br />

in the images which is not corrected <strong>for</strong> by the<br />

st<strong>and</strong>ard PET corrections [42-45] (Figure 5).<br />

This bias also results in degraded image contrast<br />

[43, 46]. Crude correction methods <strong>for</strong> this effect<br />

have been suggested, in the <strong>for</strong>m <strong>of</strong> a uni<strong>for</strong>m<br />

background subtraction [43, 47], subtraction <strong>of</strong><br />

a fit to the sinogram data outside the object<br />

[43, 48], a convolution subtraction algorithm<br />

[45], or a point-spread function subtraction [44],<br />

but all <strong>of</strong> these methods were developed <strong>for</strong><br />

conventional PET scanners <strong>and</strong> they cannot be<br />

readily used in list-mode image reconstruction<br />

with state-<strong>of</strong>-the-art PET-CT scanners.<br />

The single-scatter simulation scatter correction<br />

applied on all the latest generation PET <strong>and</strong><br />

PET-CT scanners [49] usually includes a scaling<br />

F<br />

Figure 2. Serial 89Zr-cmAb U36 PET<br />

images <strong>of</strong> a patient with oropharyngeal<br />

tumor (indicated by arrows),<br />

arranged (left to right) from 1, 24,<br />

72, <strong>and</strong> 144 h after injection. Gray<br />

scale settings were set <strong>for</strong> each<br />

image independently, <strong>for</strong> clarity.<br />

Adapted from Börjesson et al. [36].<br />

Figure 2a. Increased uptake in time<br />

<strong>of</strong> 89Zr-cmAb U36 in tumor<br />

(indicated by arrows).<br />

Figure 2b. Circulating 89Zr-cmAb U36 in heart <strong>and</strong> uptake in organs.<br />

G<br />

Figure 3. Example <strong>of</strong> 3D 131I absorbed<br />

dose calculations in a thyroid cancer<br />

patient based on serial PET images<br />

with 124I <strong>and</strong> created using the<br />

STRATOS internal radiation<br />

dosimetry package on an Imalytics*<br />

workstation (<strong>Philips</strong> Research,<br />

Aachen, Germany). Image courtesy<br />

<strong>of</strong> Dr. Bernd Schweizer, <strong>Philips</strong><br />

Research, <strong>and</strong> Dr. Walter Jentzen,<br />

University Hospital Essen.<br />

*This product is not licensed or<br />

intended <strong>for</strong> human diagnostic or<br />

therapeutic use.<br />

MEDICAMUNDI 54/2 2010 43


E<br />

Figure 4. Simplified decay schemes<br />

<strong>of</strong> 124I, 89Zr, <strong>and</strong>, <strong>for</strong> comparison,<br />

the “st<strong>and</strong>ard” PET isotope 18F. Only radiation with abundance >1%<br />

is shown.<br />

5<br />

G<br />

Figure 5. Degrading effects in PET,<br />

from left to right: r<strong>and</strong>om<br />

coincidences, scattered radiation,<br />

<strong>and</strong> prompt gamma coincidences<br />

where one <strong>of</strong> the annihilation photons<br />

is detected in coincidence with a<br />

prompt gamma photon.<br />

Adapted from Lubberink et al. [43].<br />

44 MEDICAMUNDI 54/2 2010<br />

4<br />

to match the estimated scatter contribution to<br />

the actual events measured just outside the body.<br />

If this scaling includes both a multiplicative as<br />

well as an additive factor, it implicitly per<strong>for</strong>ms<br />

a crude correction <strong>for</strong> a uni<strong>for</strong>m bias caused by<br />

prompt gamma coincidences as well [50, 51].<br />

Finally, is has been shown that the distribution<br />

<strong>of</strong> prompt gamma coincidences matches the<br />

distribution <strong>of</strong> r<strong>and</strong>om coincidences rather well<br />

[52]. There<strong>for</strong>e, a correction method involving<br />

subtraction <strong>of</strong> a scaled r<strong>and</strong>oms sinogram could<br />

be an accurate correction <strong>for</strong> prompt gamma<br />

coincidences [52], possibly incorporated into<br />

the single scatter simulation [53].<br />

The increased singles rate due to gamma radiation<br />

leads to increased r<strong>and</strong>om coincidence rates.<br />

This can be accurately corrected <strong>for</strong> using the<br />

st<strong>and</strong>ard delayed window method, but correction<br />

<strong>for</strong> a larger r<strong>and</strong>om fraction increases image<br />

noise. One option to improve image quality may<br />

be the use <strong>of</strong> a narrower energy window, which<br />

reduces r<strong>and</strong>om coincidence rates involving<br />

higher-energy photons, such as the 603 keV<br />

photon emitted by 124 I (Figures 6 <strong>and</strong> 7). The<br />

<strong>Philips</strong> Gemini TF-64 system in use in our<br />

research department allows the energy window<br />

to be adjusted while working in the research<br />

mode, but this is not a capability present in the<br />

normal, clinical mode.<br />

The fraction <strong>of</strong> detected photons with energy<br />

outside the scanner’s energy window increases<br />

considerably compared to positron-only emitters.<br />

Rejection <strong>of</strong> photons outside the energy window<br />

does contribute to dead time, but these photons<br />

are not counted in the singles rate. Since the dead<br />

time correction is usually implemented as a<br />

function <strong>of</strong> singles rate, it may become<br />

inaccurate [42, 54].<br />

As can be concluded from the decay schemes in<br />

Figure 4, all <strong>of</strong> these problems affecting image<br />

quality <strong>and</strong> quantitative accuracy occur with 124 I.<br />

For 89 Zr, however, the 909 keV photon is not<br />

emitted simultaneously with positrons. There<strong>for</strong>e,<br />

with 89 Zr, an increased r<strong>and</strong>om coincidence rate<br />

can be expected, but no quantitative bias.<br />

Hence, in terms <strong>of</strong> decay radiation, 89 Zr may be<br />

considered the optimal PET isotope <strong>for</strong><br />

labeling MAbs.<br />

Future developments<br />

An inquiry among nuclear medicine departments<br />

in Europe revealed a high dem<strong>and</strong> <strong>for</strong> new<br />

radionuclides, especially 124 I [55]. Methods <strong>for</strong><br />

large-scale production <strong>of</strong> highly pure 89 Zr <strong>and</strong><br />

124 I, <strong>and</strong> <strong>for</strong> facile <strong>and</strong> stable coupling <strong>of</strong> these<br />

positron emitters to MAbs, have been developed<br />

at VU University Medical Center, Amsterdam<br />

[10, 56, 57]. In addition, a GMP facility has been<br />

established <strong>for</strong> large-scale production <strong>of</strong> these<br />

isotopes in quantities exceeding 3 GBq/day, so<br />

that these isotopes can be supplied worldwide.<br />

Furthermore, the availability <strong>of</strong> the PET-CT<br />

technique is increasing rapidly. For example, in<br />

2002 there were only two PET sites in the<br />

Netherl<strong>and</strong>s, but in 2010 there are more than 30,<br />

almost all <strong>of</strong> which are PET-CT. Similar increases<br />

can be seen worldwide. This allows <strong>for</strong> rapid<br />

clinical introduction <strong>of</strong> labeled targeted drugs<br />

<strong>for</strong> individualized therapy, <strong>and</strong> <strong>for</strong> development<br />

<strong>of</strong> the related techniques.


6<br />

G<br />

Figure 6. Effect <strong>of</strong> using a narrower energy window.<br />

Figure 6a. Noise equivalent count rates, which are a measure <strong>of</strong> signal-to-noise ratio, <strong>of</strong> the Gemini TF-64 PET/CT<br />

with 11C using the st<strong>and</strong>ard 440-665 keV energy window (black), <strong>for</strong> 124I using this same window (blue) <strong>and</strong> <strong>for</strong> 124I using a narrow 440-560 keV window (red). NEC rates were normalized <strong>for</strong> positron abundance.<br />

Figure 6b. Improvement in recovery <strong>and</strong> image contrast <strong>of</strong> 124 I using the narrower energy window.<br />

We <strong>for</strong>esee a very important role <strong>for</strong> PET in the<br />

development <strong>and</strong> application <strong>of</strong> targeted drugs,<br />

as recently described by Van Dongen et al. <strong>for</strong><br />

Mabs [58]. However, despite clinical optimism,<br />

it is fair to state that the efficacy <strong>of</strong> current<br />

targeted drugs is still quite limited, with benefits<br />

<strong>for</strong> only a proportion <strong>of</strong> patients. Moreover,<br />

costs <strong>of</strong> these novel drugs are high, <strong>and</strong> this item<br />

became the subject <strong>of</strong> national discussions<br />

about the right to cancer care (e.g. trastuzumab)<br />

in the Netherl<strong>and</strong>s.<br />

Important questions are how to improve the<br />

efficacy <strong>of</strong> targeted therapy <strong>and</strong> how to identify<br />

patients with the greatest chance <strong>of</strong> benefit. In<br />

other words: when, how, <strong>and</strong> <strong>for</strong> whom should<br />

targeted therapy be reserved?<br />

7a 7b<br />

Quantitative <strong>imaging</strong> <strong>of</strong> targeted drugs can also<br />

be a valuable tool at several stages <strong>of</strong> drug G<br />

development <strong>and</strong> application. From first-in-man Figure 7. PET images <strong>of</strong> a patient with metastatic thyroid<br />

clinical trials with new drugs it is important to cancer at 24 h after administration <strong>of</strong> 37 MBq<br />

learn about the ideal drug dosage <strong>for</strong> optimal<br />

tumor targeting (e.g. saturation <strong>of</strong> receptors),<br />

the uptake in critical normal organs to anticipate<br />

toxicity, <strong>and</strong> the inter-patient variations in<br />

pharmacokinetics <strong>and</strong> tumor targeting. Drug<br />

<strong>imaging</strong> might provide this in<strong>for</strong>mation in an<br />

efficient <strong>and</strong> safe way, with fewer patients treated<br />

at suboptimal dose. This approach is especially<br />

attractive when the drug <strong>of</strong> interest is directed<br />

against a novel tumor target that has not been<br />

previously validated in clinical trials.<br />

Quantitative drug <strong>imaging</strong> might also be <strong>of</strong> value<br />

to guide optimal use <strong>of</strong> FDA-approved drugs, MEDICAMUNDI 54/2 2010 45<br />

124I acquired<br />

on a Gemini TF-64 PET-CT scanner (<strong>Philips</strong> Healthcare).<br />

The narrower energy window (Figure 7b) results in a<br />

15% improvement in image contrast in the largest<br />

metastasis (arrow) due to the decreased image background.<br />

Figure 7a. 440-665 keV energy window.<br />

Figure 7b. 440-560 keV energy window.<br />

including selection <strong>of</strong> patients with the highest<br />

chance <strong>of</strong> benefit from such drugs.<br />

To make this happen, s<strong>of</strong>tware tools <strong>for</strong> optimal<br />

<strong>imaging</strong> <strong>and</strong> improved <strong>quantification</strong> <strong>of</strong> long-lived<br />

positron emitters are urgently needed L


References<br />

[1] Bergstrom M, Grahnen A, Langstrom B. Positron Emission<br />

Tomography Microdosing: A New Concept with Application in Tracer<br />

<strong>and</strong> Early Clinical Drug Development. Eur J Clin Pharmacol.<br />

2003; 59(5-6): 357-366.<br />

[2] Williams HA, Robinson S, Julyan P, Zweit J, Hastings D. A<br />

Comparison <strong>of</strong> PET Imaging Characteristics <strong>of</strong> Various Copper<br />

Radioisotopes. Eur J Nucl Med Mol Imaging. 2005; 32(12): 1473-1480.<br />

[3] Orlova A, Höglund J, Lubberink M, Lebeda O, Gedda L,<br />

Lundqvist H, et al. Comparative Biodistribution <strong>of</strong> the<br />

Radiohalogenated (Br, I <strong>and</strong> At) Antibody A33. Implications <strong>for</strong> In<br />

Vivo Dosimetry. Cancer Biother Radiopharm. 2002; 17(4): 385-396.<br />

[4] Verel I, Visser GWM, Boerman OC, Eerd JEM, Finn R,<br />

Boellaard, et al. Long-Lived Positron Emitters Zirconium-89 <strong>and</strong><br />

Iodine-124 <strong>for</strong> Scouting <strong>of</strong> Therapeutic Radioimmunoconjugates with<br />

PET. Cancer Biother Radiopharm. 2003; 18(4): 655-661.<br />

[5] Verel I, Visser GW, Boellaard R, Stigter-van Walsum M, Snow GB,<br />

van Dongen GAMS, et al. Quantitative 89 Zr Immuno-PET <strong>for</strong> In<br />

Vivo Scouting <strong>of</strong> 90 Y-Labeled Monoclonal Antibodies in Xenograft-<br />

Bearing Nude Mice. J Nucl Med. 2003; 44(10): 1663-1670.<br />

[6] Perk LR, Visser GW, Vosjan MJ, Stigter-van Walsum M, Tijink BM,<br />

Leemans CR, et al. (89)Zr as a PET Surrogate Radioisotope <strong>for</strong><br />

Scouting Biodistribution <strong>of</strong> the Therapeutic Radiometals (90)Y <strong>and</strong><br />

(177)Lu in Tumor-Bearing Nude Mice after Coupling to the<br />

Internalizing Antibody Cetuximab. J Nucl Med. 2005; 46(11):<br />

1898-1906.<br />

[7] Eschmann SM, Reischl G, Bilger K, Kupferschläger J, Thelen MH,<br />

Dohmen BM, et al. Evaluation <strong>of</strong> Dosimetry <strong>of</strong> Radioiodine Therapy<br />

in Benign <strong>and</strong> Malignant Thyroid Disorders by Means <strong>of</strong> Iodine-124<br />

<strong>and</strong> PET. Eur J Nucl Med Mol Imaging. 2002; 29(6): 760-767.<br />

[8] Erdi YE, Macapinlac H, Larson SM, Erdi AK, Yeung H, Furhang EE,<br />

et al. Radiation Dose Assessment <strong>for</strong> I-131 Therapy <strong>of</strong> Thyroid Cancer<br />

Using I-124 PET Imaging. Clin Positron Imaging. 1999; 2(1):<br />

41-46.<br />

[9] Lovqvist A, Lundqvist H, Lubberink M, Tolmachev V, Carlsson J,<br />

Sundin A. Kinetics <strong>of</strong> 76 Br-Labeled Anti-CEA Antibodies in Pigs;<br />

Aspects <strong>of</strong> Dosimetry <strong>and</strong> PET Imaging Properties. Med Phys. 1999;<br />

26(2): 249-258.<br />

[10] Perk LR, Vosjan MJ, Visser GW, Budde M, Jurek P, Kiefer GE, et<br />

al. P-Isothiocyanatobenzyl-Desferrioxamine: A New Bifunctional<br />

Chelate <strong>for</strong> Facile Radiolabeling <strong>of</strong> Monoclonal Antibodies with<br />

Zirconium-89 <strong>for</strong> Immuno-PET Imaging. Eur J Nucl Med Mol<br />

Imaging. 2010; 37(2): 250-259.<br />

46 MEDICAMUNDI 54/2 2010<br />

[11] Perk LR, Stigter-Van Walsum, Visser GW, Kloet RW, Vosjan MJWD,<br />

Leemans CR, et al. Quantitative PET Imaging <strong>of</strong> Met-Expressing<br />

Human Cancer Xenografts with 89 Zr-Labeled Monoclonal Antibody<br />

DN30. Eur J Nucl Med Mol Imaging. 2008; 35(10): 1857-1867.<br />

[12] Perk LR, Visser OJ, Stigter-Van Walsum WM, Vosjan MJWD,<br />

Visser GWM, Zijlstra JM, et al. Preparation <strong>and</strong> Evaluation <strong>of</strong><br />

(89)Zr-Zevalin <strong>for</strong> Monitoring <strong>of</strong> (90)Y-Zevalin Biodistribution with<br />

Positron Emission Tomography. Eur J Nucl Med Mol Imaging.<br />

2006; 33(11): 1337-1345.<br />

[13] Herzog H, Rosch F, Stocklin G, Lueders C, Qaim SM,<br />

Feinendegen LE. Measurement <strong>of</strong> Pharmacokinetics <strong>of</strong> Yttrium-86<br />

Radiopharmaceuticals with PET <strong>and</strong> Radiation Dose Calculation <strong>of</strong><br />

Analogous Yttrium-90 Radiotherapeutics. J Nucl Med. 1993;<br />

34(12): 2222-2226.<br />

[14] Rösch F, Herzog H, Stolz B, Brockmann J, Köhle M,<br />

Mühlensiepen H, et al. Uptake Kinetics <strong>of</strong> the Somatostatin Receptor<br />

Lig<strong>and</strong> [ 86 Y]DOTA-Dphe1- Tyr3-Octreotide ([86Y]SMT487) using<br />

Positron Emission Tomography in Non-Human Primates <strong>and</strong><br />

Calculation <strong>of</strong> Radiation Doses <strong>of</strong> the 90 Y-Labeled Analogue. Eur J<br />

Nucl Med. 1999; 26(4): 358-366.<br />

[15] Graham MC, Pentlow KS, Mawlawi O, Finn RD, Daghighian F,<br />

Larson SM. An Investigation <strong>of</strong> the Physical Characteristics <strong>of</strong> 66Ga<br />

as an Isotope <strong>for</strong> PET Imaging <strong>and</strong> Quantification. Med Phys. 1997;<br />

24(2): 317-326.<br />

[16] Keen HG, Dekker BA, Disley L, Hastings D, Lyons S, Reader AJ,<br />

et al. Imaging Apoptosis in Vivo Using 124 I-Annexin V <strong>and</strong> PET.<br />

Nucl Med Biol. 2005; 32(4): 395-402.<br />

[17] Dekker B, Keen H, Lyons S, Disley L, Hastings D, Reader A, et<br />

al. MBP-Annexin V Radiolabeled Directly with Iodine-124 can be<br />

used to Image Apoptosis In Vivo Using PET. Nucl Med Biol. 2005;<br />

32(3): 241-252.<br />

[18] Iozzo P, Osman S, Glaser M, Knickmeier M, Ferrannini E, Pike VW,<br />

et al. In Vivo Imaging <strong>of</strong> Insulin Receptors by PET: Preclinical<br />

Evaluation <strong>of</strong> Iodine-125 <strong>and</strong> Iodine-124 Labeled Human Insulin.<br />

Nucl Med Biol. 2002; 29(1): 73-82.<br />

[19] Zanzonico P, O’Donoghue J, Chapman JD, Schneider R, Cai S,<br />

Larson S, et al. Iodine-124-Labeled Iodo-Azomycin-Galactoside<br />

Imaging <strong>of</strong> Tumor Hypoxia in Mice with Serial Micropet Scanning.<br />

Eur J Nucl Med Mol Imaging. 2004; 31(1): 117-128.<br />

[20] Roelcke U, Hausmann O, Merlo A, Missimer J, Maguire RP,<br />

Freitag P, et al. PET Imaging Drug Distribution After Intratumoral<br />

Injection: The Case <strong>for</strong> (124)I-Iododeoxyuridine in Malignant Gliomas.<br />

J Nucl Med. 2002; 43(11): 1444-1451.


[21] Sawka AM, Thephamongkhol K, Brouwers M, Thabane L,<br />

Browman G, Gerstein HC. Clinical Review 170: A Systematic<br />

Review <strong>and</strong> Metaanalysis <strong>of</strong> the Effectiveness <strong>of</strong> Radioactive Iodine<br />

Remnant Ablation <strong>for</strong> Well-Differentiated Thyroid Cancer. J Clin<br />

Endocrinol Metab. 2004; 89(8): 3668-3676.<br />

[22] Van Essen M, Krenning EP, De Jong M, Valkema R,<br />

Kwekkeboom DJ. Peptide Receptor Radionuclide Therapy with<br />

Radiolabelled Somatostatin Analogues in Patients with Somatostatin<br />

Receptor Positive Tumours. Acta Oncol. 2007; 46(6): 723-734.<br />

[23] Lundqvist H, Lubberink M, Tolmachev V, Lövqvist A, Sundin A,<br />

Beshara S, et al. Positron Emission Tomography <strong>and</strong><br />

Radioimmunotargeting - General Aspects. Acta Oncol. 1999; 38(3):<br />

335-341.<br />

[24] Wiseman GA, White CA, Sparks RB, Erwin WD, Podol<strong>of</strong>f DA,<br />

Lamonica D, et al. Biodistribution <strong>and</strong> Dosimetry Results from a<br />

Phase III Prospectively R<strong>and</strong>omized Controlled Trial <strong>of</strong> Zevalin<br />

Radioimmunotherapy <strong>for</strong> Low-Grade, Follicular, or Trans<strong>for</strong>med<br />

B-Cell Non-Hodgkin’s Lymphoma. Crit Rev Oncol Hematol. 2001;<br />

39(1-2): 181-194.<br />

[25] Sgouros G. Dosimetry <strong>of</strong> Internal Emitters. J Nucl Med. 2005; 46<br />

Suppl 1: 18S-27S.<br />

[26] Siegel JA, Thomas SR, Stubbs JB, Stabin MG, Hays MT, Koral<br />

KF, et al. MIRD Pamphlet No. 16: Techniques <strong>for</strong> Quantitative<br />

Radiopharmaceutical Biodistribution Data Acquisition <strong>and</strong> Analysis<br />

<strong>for</strong> Use in Human Radiation Dose Estimates. Journal <strong>of</strong> Nuclear<br />

Medicine. 1999; 40(2): 37S-61S.<br />

[27] Stabin MG, Sparks RB, Crowe E. OLINDA/EXM: The Second-<br />

Generation Personal Computer S<strong>of</strong>tware <strong>for</strong> Internal Dose Assessment<br />

in Nuclear Medicine. J Nucl Med. 2005; 46(6): 1023-1027.<br />

[28] Bolch WE, Bouchet LG, Robertson JS, Wessels BW, Siegel JA,<br />

Howell RW, et al. MIRD Pamphlet No. 17: the Dosimetry <strong>of</strong><br />

Nonuni<strong>for</strong>m Activity Distributions--Radionuclide S Values at the<br />

Voxel Level. Medical Internal Radiation Dose Committee. J Nucl<br />

Med. 1999; 40(1): 11S-36S.<br />

[29] Furhang EE, Sgouros G, Chui CS. Radionuclide Photon Dose Kernels<br />

<strong>for</strong> Internal Emitter Dosimetry. Med Phys. 1996; 23(5): 759-764.<br />

[30] Str<strong>and</strong> SE, Jonsson BA, Ljungberg M, Tennvall J.<br />

Radioimmunotherapy Dosimetry - A Review. Acta Oncologica. 1993;<br />

32(7-8): 807-817.<br />

[31] Ljungberg M, Frey E, Sjogreen K, Liu X, Dewaraja Y, Str<strong>and</strong> SE.<br />

3D Absorbed Dose Calculations Based On SPECT: Evaluation <strong>for</strong><br />

111-In/90-Y Therapy Using Monte Carlo Simulations. Cancer<br />

Biother Radiopharm. 2003; 18(1): 99-107.<br />

[32] Furhang EE, Chui CS, Sgouros G. A Monte Carlo Approach to<br />

Patient-Specific Dosimetry. Med Phys. 1996; 23(9): 1523-1529.<br />

[33] Furhang EE, Chui CS, Kolbert KS, Larson SM, Sgouros G.<br />

Implementation <strong>of</strong> a Monte Carlo Dosimetry Method <strong>for</strong> Patient-<br />

Specific Internal Emitter Therapy. Med Phys. 1997; 24(7):<br />

1163-1172.<br />

[34] Song H, Du Y, Sgouros G, Prideaux A, Frey E, Wahl RL.<br />

Therapeutic Potential <strong>of</strong> 90 Y- <strong>and</strong> 131 I-Labeled Anti-CD20<br />

Monoclonal Antibody in Treating Non-Hodgkin’s Lymphoma with<br />

Pulmonary Involvement: A Monte Carlo-Based Dosimetric Analysis.<br />

J Nucl Med. 2007; 48(1): 150-157.<br />

[35] Norrgren K, Svegborn SL, Areberg J, Mattsson S. Accuracy <strong>of</strong> the<br />

Quantification <strong>of</strong> Organ Activity from Planar Gamma Camera Images.<br />

Cancer Biother Radiopharm. 2003; 18(1): 125-131.<br />

[36] Borjesson PK, Jauw YW, de Bree R, Roos JC, Castelijns JA,<br />

Leemans CR, van Dongen GA, Boellaard R, et al. Radiation<br />

Dosimetry <strong>of</strong> 89 Zr-Labeled Chimeric Monoclonal Antibody U36 as<br />

used <strong>for</strong> Immuno-PET in Head <strong>and</strong> Neck Cancer Patients. J Nucl<br />

Med. 2009; 50(11): 1828-1836.<br />

[37] Lubberink M, Tolmachev V, Widström C, Bruskin A, Lundqvist H,<br />

Westlin JE. 110m In-DTPA-D-Phe1-Octreotide <strong>for</strong> Imaging <strong>of</strong><br />

Neuroendocrine Tumors with PET. J Nucl Med. 2002; 43(10):<br />

1391-1397.<br />

[38] Kolbert KS, Sgouros G, Scott AM, Bronstein JE, Malane RA,<br />

Zhang J, et al. Implementation <strong>and</strong> Evaluation <strong>of</strong> Patient-Specific<br />

Three-Dimensional Internal Dosimetry. J Nucl Med. 1997; 38(2):<br />

301-308.<br />

[39] Prideaux AR, Song H, Hobbs RF, He B, Frey EC, Ladenson PW,<br />

et al. Three-Dimensional Radiobiologic Dosimetry: Application <strong>of</strong><br />

Radiobiologic Modeling to Patient-Specific 3-Dimensional Imaging-<br />

Based Internal Dosimetry. J Nucl Med. 2007; 48(6): 1008-1016.<br />

[40] Sgouros G, Kolbert KS, Sheikh A, Pentlow KS, Mun EF, Barth A,<br />

et al. Patient-Specific Dosimetry <strong>for</strong> 131I Thyroid Cancer Therapy<br />

Using 124I PET <strong>and</strong> 3-Dimensional-Internal Dosimetry (3D-ID)<br />

S<strong>of</strong>tware. J Nucl Med. 2004; 45(8): 1366-1372.<br />

[41] Kolbert KS, Pentlow KS, Pearson JR, Sheikh A, Finn RD, Humm JL,<br />

et al. Prediction <strong>of</strong> Absorbed Dose To Normal Organs in Thyroid<br />

Cancer Patients Treated with 131 I by Use <strong>of</strong> 124 I PET <strong>and</strong><br />

3-Dimensional Internal Dosimetry S<strong>of</strong>tware. J Nucl Med. 2007;<br />

48(1): 143-149.<br />

[42] Martin CC, Christian BT, Satter MR, Nickerson LDH, Nickles RJ.<br />

Quantitative PET with Positron Emitters That Emit Prompt Gamma<br />

Rays. IEEE Trans Med Imaging. 1995; 14(4): 681-687.<br />

MEDICAMUNDI 54/2 2010 47


[43] Lubberink M, Schneider H, Bergström M, Lundqvist H.<br />

Quantitative Imaging <strong>and</strong> Correction <strong>for</strong> Cascade Gamma Radiation<br />

<strong>of</strong> 76 Br with 2D <strong>and</strong> 3D PET. Phys Med Biol. 2002; 47(19):<br />

3519-3534.<br />

[44] Walr<strong>and</strong> S, Jamar F, Mathieu I, De Camps J, Lonneux M,<br />

Sibomana M, et al. Quantitation in PET using Isotopes Emitting<br />

Prompt Single Gammas: Application To Yttrium-86. Eur J Nucl<br />

Med Mol Imaging. 2003; 30(3): 354-361.<br />

[45] Beattie BJ, Finn RD, Rowl<strong>and</strong> DJ, Pentlow KS. Quantitative<br />

Imaging <strong>of</strong> Bromine-76 <strong>and</strong> Yttrium-86 with PET: A Method <strong>for</strong> the<br />

Removal <strong>of</strong> Spurious Activity Introduced by Cascade Gamma Rays.<br />

Med Phys. 2003; 30(9): 2410-2423.<br />

[46] Lubberink M, Van Schie A, De Jong HW, Van Dongen GA,<br />

Teule GJ. Acquisition Settings <strong>for</strong> PET <strong>of</strong> 124 I Administered<br />

Simultaneously with Therapeutic Amounts <strong>of</strong> 131 I. J Nucl Med.<br />

2006; 47(8): 1375-1381.<br />

[47] Pentlow KS, Finn RD, Larson SM, Erdi YE, Beattie BJ, Humm JL.<br />

Quantitative Imaging <strong>of</strong> Yttrium-86 with PET. The Occurrence<br />

<strong>and</strong> Correction <strong>of</strong> Anomalous Apparent Activity in High Density<br />

Regions. Clin Positron Imaging. 2000; 3(3): 85-90.<br />

[48] Kull T, Ruckgaber J, Weller R, Reske S, Glatting G. Quantitative<br />

Imaging <strong>of</strong> Yttrium-86 PET with the ECAT EXACT HR+ in 2D<br />

Mode. Cancer Biother Radiopharm. 2004; 19(4): 482-490.<br />

[49] Watson CC. New, Faster, Image-Based Scatter Correction <strong>for</strong> 3D<br />

PET. IEEE Trans Nucl Sci. 2000; 47(4): 1587-1594.<br />

[50] Gregory R, Partridge M, Flux GD. Assessment <strong>of</strong> Cascade Correction<br />

Methods <strong>for</strong> Quantitative 124 I PET Imaging. Eur J Nucl Med Mol<br />

Imaging. 2009; 36: OP066.<br />

[51] Surti S, Scheuermann R, Karp JS. Correction Technique <strong>for</strong> Cascade<br />

Gammas in I-124 Imaging on a Fully-3D, Time-Of-Flight PET Scanner.<br />

IEEE Trans Nucl Sci. 2009; 56(3): 653-660.<br />

[52] Lubberink M. Quantitative Imaging with PET - Per<strong>for</strong>mance <strong>and</strong><br />

Applications <strong>of</strong> 76 Br, 52 Fe, 110m In <strong>and</strong> 134 La. Uppsala: Acta Universitatis<br />

Upsaliensis. Comprehensive Summaries <strong>of</strong> Uppsala Dissertations<br />

from the Faculty <strong>of</strong> Medicine 1034; 2001.<br />

[53] Watson CC, Hayden C, Casey ME, Hamill J, Bendriem B.<br />

Prompt Gamma Correction <strong>for</strong> Improved Quantification in 82 Rb PET.<br />

J Nucl Med. 2008; 49: 64P.<br />

[54] Lubberink M, Schneider H, Bergström M, Lundqvist H. Count<br />

Rate Characteristics <strong>of</strong> PET with Bromine-76. European Journal <strong>of</strong><br />

Nuclear Medicine. 2001; 28(8): 1008.<br />

[55] Bockisch A, Freudenberg L, Rosenbaum S, Jentzen W. (124)I in<br />

PET Imaging: Impact On Quantification, Radiopharmaceutical<br />

Development <strong>and</strong> Distribution. Eur J Nucl Med Mol Imaging.<br />

2006; 33(11): 1247-1248.<br />

48 MEDICAMUNDI 54/2 2010<br />

[56] Tijink BM, Perk LR, Budde M, Stigter-van Walsum M, Visser GW,<br />

Kloet RW, et al. (124)I-L19-SIP <strong>for</strong> Immuno-PET Imaging <strong>of</strong> Tumor<br />

Vasculature <strong>and</strong> Guidance <strong>of</strong> (131)I-L19-SIP Radioimmunotherapy.<br />

Eur J Nucl Med Mol Imaging. 2009; 36(8): 1235-1244.<br />

[57] Vosjan MJ, Perk LR, Visser GW, Budde M, Jurek P, Kiefer GE, et<br />

al. Conjugation <strong>and</strong> Radiolabeling <strong>of</strong> Monoclonal Antibodies with<br />

Zirconium-89 <strong>for</strong> PET Imaging Using the Bifunctional Chelate<br />

P-Isothiocyanatobenzyl-Desferrioxamine. Nature Protocols. 2010;<br />

5: in Press.<br />

[58] Van Dongen GA, Visser GW, Lub-De Hooge MN, De Vries EG,<br />

Perk LR. Immuno-PET: A Navigator in Monoclonal Antibody<br />

Development <strong>and</strong> Applications. Oncologist. 2007; 12(12):<br />

1379-1389.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!