Trends in Radiopharmaceuticals Pub1294_vol2_web
Trends in Radiopharmaceuticals Pub1294_vol2_web
Trends in Radiopharmaceuticals Pub1294_vol2_web
RADIOPHARMACEUTICALS
(ISTR-2005)
VOLUME 2
The following States are Members of the International Atomic Energy Agency:
The Agency’s Statute was approved on 23 October 1956 by the Conference on the Statute of
the IAEA held at United Nations Headquarters, New York; it entered into force on 29 July 1957.
The Headquarters of the Agency are situated in Vienna. Its principal objective is “to accelerate and
enlarge the contribution of atomic energy to peace, health and prosperity throughout the world’’.
PROCEEDINGS SERIES
TRENDS IN
RADIOPHARMACEUTICALS
(ISTR-2005)
In two volumes
VOLUME 2
All IAEA scientific and technical publications are protected by the terms
of the Universal Copyright Convention as adopted in 1952 (Berne) and as
revised in 1972 (Paris). The copyright has since been extended by the World
Intellectual Property Organization (Geneva) to include electronic and virtual
intellectual property. Permission to use whole or parts of texts contained in
IAEA publications in printed or electronic form must be obtained and is
usually subject to royalty agreements. Proposals for non-commercial
reproductions and translations are welcomed and considered on a case-by-case
basis. Enquiries should be addressed to the IAEA Publishing Section at:
© IAEA, 2007
Printed by the IAEA in Austria
November 2007
STI/PUB/1294
IAEAL 07–00497
FOREWORD
The Proceedings have been edited by the editorial staff of the IAEA to the extent
considered necessary for the reader’s assistance. The views expressed remain, however, the
responsibility of the named authors or participants. In addition, the views are not
necessarily those of the governments of the nominating Member States or of the
nominating organizations.
Although great care has been taken to maintain the accuracy of information
contained in this publication, neither the IAEA nor its Member States assume any
responsibility for consequences which may arise from its use.
The use of particular designations of countries or territories does not imply any
judgement by the publisher, the IAEA, as to the legal status of such countries or territories,
of their authorities and institutions or of the delimitation of their boundaries.
The mention of names of specific companies or products (whether or not indicated
as registered) does not imply any intention to infringe proprietary rights, nor should it be
construed as an endorsement or recommendation on the part of the IAEA.
The authors are responsible for having obtained the necessary permission for the
IAEA to reproduce, translate or use material from sources already protected by
copyrights.
CONTENTS OF VOLUME 2
177
Lu labelled nitroimadzoles and nitrotriazoles for possible use
in targetedtherapy of hypoxic tumours . . . . . . . . . . . . . . . . . . . . . . . . . 51
T. Das, S. Chakraborty, A. Mukherjee, S. Banerjee, G. Samuel,
H.D. Sarma, M. Venkatesh
Iodine whole body scan, thyroglobulin levels, 99mTc MIBI scan and
computed tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
N.Ö. Küçük, S.S. Gültekin, G. Aras, E. İbiş
99m
Tc-MIBI and 131I scintigraphy in the follow-up of differentiated
thyroid carcinoma (DTC) patients after surgery . . . . . . . . . . . . . . . . . 123
S. Sergieva, T. Hadjieva, V. Botev, A. Dudov
18
F RADIOPHARMACEUTICALS AND AUTOMATION OF
SYNTHESIS (SESSION 12)
18
F based radiopharmaceuticals and automation of synthesis . . . . . . . . . . . 265
P.K. Garg, S. Garg
1-[18F]fluoroethyleneglycol-2-nitroimidazoles:
A novel class of potential hypoxia PET markers . . . . . . . . . . . . . . . . . 295
R.J. Abdel-Jalil, M. Übele, W. Ehrlichmann, W. Voelter,
H.-J. Machulla
(Session 8)
Chairpersons
C. DECRISTOFORO
Austria
K.K. SOLANKI
IAEA
.
BIOLOGICAL AND CHEMICAL EVALUATION OF
VARIOUS RADIOCOLLOIDS USED FOR CLINICAL
RADIOSYNOVECTOMY
Abstract
1. INTRODUCTION
3
JÁNOKI et al.
198
Au colloid was first used for radiocolloid synovectomy and this isotope has
been continously investigated. The main drawbacks of an 198Au colloid are the
411 keV gamma emission which creates an unnecessary radiation hazard, the
small particle size, which results in excessive loss from the joint space by
lymphatic drainage, and the high radiation doses to the proximal lymph nodes.
Other radiopharmaceuticals [1–17] that have been developed, studied
and registered for human use are listed in Table 1. The different radiation
properties of each therapeutic isotope determine their respective use regarding
the joint size. The authors studied the preparation and biological use of colloids
labelled with isotopes 90Y, 166Ho, 169Er and 188Re which are applicable for
treating the various joints for radiosynoviorthesis.
4
SESSION 8
In the case of the 90Y-citrate-silicate colloid, the TLC method was used to
determine colloid bound activity. Merck Silicagel 5553 layer, solvent containing
64 mL n-Propanol, 16 mL water, 20 mL methanol and 1 g of tartaric acid were
used and 1–5 µL volume of sample was deposited.
The Rf value of radiocolloids was zero. For in vivo stability study 100 µL
of radiocolloid was inoculated in 3 mL of rabbit synovial fluid at 37ºC for 24 h
and 5 d. After inoculation, colloid bound activity ratios were determined by
using the same TLC method used earlier.
5
JÁNOKI et al.
6
SESSION 8
an activity in the range 105–500 µCi (5.6–18.5 MBq)/100 µL of the test article
into their knee joint randomly selected.
As previous experiments in the NRIRR have shown, in the case of
gamma camera (Nucline X-Ring/R, Mediso Ltd, Hungary) imaging of 90Y
bremsstrahlung in a fully open energy window centred at 140 keV, a vial with
0.55 MBq and a vial containing 25 MBq of activity placed at the same detector
can be distinguished visually by the operator. Thus, correctness of the injections
was controlled with the use of a gamma camera, the exclusion criterion being
the appearance of a second spot of activity near the injection site. Immediately
after dosing, the animal was placed in the ventrodorsal position above the
detector and the scintigraphic image was checked. In the case of two or more
spots appearing on screen, the fact was recorded and the animal was discarded
from the experiment. If the activity spot was unique and in the region of the
knee, the injection was deemed to be correct. Figure 1 shows an example of
correct injection.
At the end of the study, the rabbits were anaesthetized again by intra-
muscular administration of 100 mg ketamine/kg b.w. (SBH-Ketamin, SBH Kft.,
Hungary) and 2 mg xylazine/kg b.w. (Primazin, Alfasav B.V., Netherlands). The
animals were sacrificed 5–10 min after anaesthesia induction by intracardial
injection of 0.5 mL/kg b.w. of a specific veterinary drug for euthanasia, T 61® ad
FIG. 1. Ventrodorsal whole body scintillation image of a correctly injected rabbit. The
only spot appearing corresponds to the injection site at the knee.
7
JÁNOKI et al.
us. vet. (Intervet B.V., Netherlands). All the animal handling methods used in
the study were in compliance with all applicable sections of the Hungarian
Laws No. XXVIII/1998 and LXVII/2002 on the protection and welfare of
animals.
Animals were sacrificed at 6, 24 and 48 h and 5 d post-injection. The
samples of the injected knees (with 1 cm of femoral and tibial bone outside the
capsule) were collected. The samples were washed in physiological saline, dried
with absorbent paper and weighed after collection. Samples were collected in
containers labelled according to animal identification, organ, collection time
points and date.
Samples were placed in individually identified porcelain crucibles. The
wet net weight of the samples was calculated from weight measurements made
on the empty crucible and containing the sample. Samples were desiccated in a
thermodryer at 200°C for 24 h and ashed in a laboratory furnace (OH-63,
OMSZOV, Hungary) at 600°C for 12 h. The net weight of each ash sample was
determined by measuring the crucibles with and without ashes with an
analytical balance to four digits.
Concentrated HNO3 was added to the ash samples and the solution of
each sample was brought to a fixed 10 mL volume. This solution was incubated
for at least 12 h at room temperature and homogenized by a Vortex mixer
several times during dissolution. Immediately after the last homogenization,
1 mL aliquots of the sample solutions were transferred to plastic measurement
test tubes in triplicate.
After the dose administration and a 30 min totally immobile period due
to general anaesthesia, the injected knee of the rabbits in the immobilized
group was held by a PVC plastic splint formed individually for the animals and
fixed by tape and bandage to hold the knee at semi-flexion, to facilitate the
keeping of a natural seated position of the rabbit in the cage. This immobili-
zation splint was kept on the knees for 24 h to ensure proper and indulgent
immobilization of the joint, as demonstrated by preliminary experiments at the
NRIRR.
8
SESSION 8
triplicate sample solutions. It was found that the device measurements are
linear in the range of 750–2694 kBq for 1 mL sample volumes and 60 s
measurements.
Standard deviations (SD) of data were calculated using n-1 as the degree
of freedom for multiplying n in the divider of the appropriate formula.
Comparison of the groups was performed using the two-tailed Student’s t-test
and the Mann-Whitney U-test. ANOVA calculations were made to compare
the different organ uptake values at the different time points. On the basis of
biodistribution data, retention times, pharmacokinetic curves and the absorbed
doses were calculated using Prism 4.0 and MIRDOSE 3.1 softwares.
3. RESULTS
9
JÁNOKI et al.
10
SESSION 8
4. BIODISTRIBUTION STUDY
90
4.1. Y-citrate/silicate colloid
Biodistribution of these two 90Y labelled colloids are seen in Tables 3 and 4.
167
4.2. Er-citrate colloid
In the present study, the remaining activity in rabbit knee after intra-
articularly injected 167Er-citrate colloid suspension was determined. At three
different time points (6 h, 2d, 8 d) knee, bone and blood were sampled and
measured by liquid scintillation techniques. Radiochemical purity of the
filtered batch used in the present work was high throughout the study (>99%).
Activity values retained in the knee as injected dose per cent were as
follows:
Time
6h 24 h 5d 8d 13 d
point
Dose %
in whole Ave. ± SD Ave. ± SD Ave. ± SD Ave. ± SD Ave. ± SD
organ
Blood 0.158 0.386 0.083 0.120 1.454 2.606 0.885 2.168 0.018 0.030
Liver 0.318 0.307 0.301 0.437 0.749 1.009 0.272 0.196 0.082 0.183
Spleen 0.014 0.017 0.029 0.030 0.050 0.043 0.099 0.051 0.046 0.048
Kidney 0.120 0.106 0.061 0.047 0.205 0.095 0.248 0.372 0.082 0.077
Testes 0.006 0.010 0.029 0.032 0.008 0.008 0.055 0.069 0.000 0.000
Ing. 0.006 0.008 0.015 0.016 0.000 0.001 0.075 0.064 0.059 0.081
lymph
node
Whole 1.396 2.495 0.050 0.050 3.252 1.023 6.300 2.969 0.150 0.196
skeleton
Knee 95.984 2.479 94.265 1.733 91.282 2.721 88.116 4.851 87.600 2.074
11
JÁNOKI et al.
Time point 6h 24 h 48 h 5d
Dose %
in whole Ave. ± SD Ave. ± SD Ave. ± SD Ave. ± SD
organ
Low level leakage was documented also with the very low blood activity
which was always less than 0.01%ID and with the low whole skeleton uptake
which ranged 1.7–4.3%ID.
None of the rabbits used in this experiment showed any chemical or
radiation toxicity signs, including any local intolerance.
188
4.3. Re-tin colloid
12
SESSION 8
FIG. 4. Static images of the knee of a rabbit taken at 3, 24, 48 and 72 h after injection.
Differences in background are due to the automatic cut-off level setting of the camera
software. Rabbit contours were generated from the acquisition data by using the Inter-
View© software.
demonstrated a very fast excretion of perrhenate via the kidneys and bladder
48 and 72 h post-injection; only background activity could be observed in
healthy rabbits receiving perrhenate eluate only.
166
4.4. Ho-phytate biodistribution
13
JÁNOKI et al.
Knee: 92%
Liver: 2.8%
Kidney: 1.5%
Cummulative activity
In urine: 1.19%
In faeces: 2.23%
166
FIG. 6. Ho-phytate distribution in rabbit 48 h after intra-articulation injection.
14
SESSION 8
Observation 13 d 5d 5d 19 d 3d 7d
time
T1/2 2.67 d 2.67 d 2.67 d 9.4 d 0.71 d 1.12 d
Residence time (h)
Knee 282.072 112.081 103.152 395.042 10.647 157.380
15
JÁNOKI et al.
16
SESSION 8
5. DISCUSSION
17
JÁNOKI et al.
The radiochemical purity and stability studies (in vitro and in synovial
fluid) of different radiocolloids showed very uniform results. In all cases, the
rate of colloid bound activity always exceeded 99% throughout the whole
experiment. The mean particle size of this colloid was 0.6–3.1 µm and after
intra-articular injection into 149 rabbits the animals showed no sign of chemical
or radiation toxicity. No local intolerance was reported. The injection
technique developed and practiced was controlled by XR, US and scintigraphy
and allowed personnel to control any leakage relating to the incorrectly
positioned needle in the knee joint cavity.
The distribution of injected activity in various organs outside the knee
(this value is equivalent to leakage activity) such as the liver, spleen, kidneys,
testes, blood, lymph nodes and skeleton was very low; in most cases it was
around 1% ID during the whole study.
The long stability of radiocolloids is exemplified by 90Y-citrate colloid
where applied activity values retained in the knee between 6 h and 13 d ranged
from 93.61 ± 1.67% to 86.80 ± 1.59% of injected activity in the non-
immobilized group and 95.10 ± 0.91% to 87.68 ± 2.26% in the immobilized
18
SESSION 8
19
JÁNOKI et al.
6. CONCLUSION
(a) The energy of the ß– particles must be high enough to penetrate the whole
depth of the inflamed synovial membrane.
(b) The colloid particle size should be between 1 and 5 µm.
(c) The hardness of particles (resistance to in vivo dissolution, enzymatic
action or mechanical stress) are also important.
ACKNOWLEDGEMENTS
20
SESSION 8
REFERENCES
[1] WEBB, F.W.S., LOWE, J., BLUESTONE, R., Uptake of colloidal radioactive
Yttrium by synovial membrane, Ann. Rheum. Dis., 28: p. 300, (1969).
[2] HUŠÁK, V., WIEDERMANN, M., KRÁL, M., Absorbed dose due to beta-rays
from radioactive colloids in radiation synovectomy, Phys. Med. Biol.,. Vol. 18, No.
6, pp. 848-854, (1973).
[3] OKA, M., Radiation synovectomy of the rheumatoid knee with Yttrium-90,
Annals of Clinical Research 7: pp. 205 – 210, (1975).
[4] BOWRING, C.S., KEELING, D.H., Absorbed radiation dose in radiation
synovectomy, British Journal of Radiology, 51, pp. 836 – 837, (1978).
[5] McCOY, J.M., Radioactive Yttrium-90: intra-articularly for the treatment of rheu-
matoid arthritis, Journal of MAG Vol. 67, pp. 361 – 365, May (1978).
[6] DUNSCOMBE, P.B., RAMSEY, N.W., Radioactivity studies on 2 synovial
specimens after radiation synovectomy with Yttrium-90 Silicate, Annals of the
Rheumatic Diseases, 39: pp. 87 – 89, (1980).
[7] BEATSON, T.R., Radiation synovectomy (synoviorthesis) for rheumatoid
arthritis in the Isle of Man, Gerontology 28: pp. 258 – 264, (1982).
[8] NOBLE, J., et al., Leakage of radioactive particle systems from a synovial joint
studied with a gamma camera, The Journal of Bone and Joint Surgery, Incorpo-
rated, Vol. 65-A, No. 3, March, (1983).
[9] ZALUTSKY, M.R., et al., Radiation synovectomy with 165DY-FHMA: lymph
node uptake and radiation dosimetry calculations, Int. J. Nucl. Med. Biol. Vol. 12,
No. 6. pp. 457 – 465, (1986).
[10] ZALUTSKY, M.R., NOSKA, M.A., GALLAGHER, P.W., SHORTKROFF, S.,
SLEDGE, C.B., Use of liposomes as carriers for radiation synovectomy, Nucl.
Med. Biol. Vol. 15, No. 2. pp. 151 – 156, (1988).
[11] DAVIS, M.A., CHINOL, M., Radiopharmaceuticals for radiation synovectomy:
evaluation of two Yttrium-90 particulate agents, J. Nucl. Med. Vol. 30: No. 6, pp.
1047 – 1055, June (1989).
[12] MYERS, S.L., SLOWMAN, S.D., BRANDT, K.D., Radiation synovectomy stimu-
lates glicosaminoglycan synthesis by normal articular cartilage, J. Lab. Clinik Meg.
pp. 28 – 35, (1989).
[13] CHINOL, M., VALLABHAJOSULA, S., ZUCKERMAN, J.D., GOLDSMITH, S.J.,
In vivo stability of ferric hydroxide macroaggregates (FHMA). Is it a suitable
carrier for radionuclides used in synovectomy?, Nucl. Med. Biol. Vol. 17, No. 5.
pp. 479 – 486, (1990).
[14] JOHNSON, L.S., YANCH, J.C., Absorbed dose profiles for radionuclides of
frequent use in radiation synovectomy, arthritis and rheumatism, Vol. 34, No.12,
December, (1991).
[15] VAN KASTEREN, M.E.E., NOVÁKOVÁ, I.R.O., BOERBOOMS, A.M.T.,
LEMMENS, J.A.M., Long term follow up of radiosynovectomy with Yttrium-90
silicate in haemophilic haemarthrosis, Ann Rheum Dis 52: 548 – 550, (1993).
21
JÁNOKI et al.
[16] STUCKI, G., BOZZONE, P., TREUER, E., WASSMER, P., FELKER, M.,
Efficacy and safety of radiation synovectomy with Yttrium-90: a retrospective
long-term analysis of 164 applications in 82 patients, British Journal of Rheuma-
tology 32: 383 – 386, (1993).
[17] EDMONDS, J., et al., A comparative study of the safety and efficacy of dyspro-
sium-165 hydroxide macro-aggregate and Yttrium-90 silicate colloid in radiation
synovectomy–a multicentre double blind clinical trial, British Journal of Rheuma-
tology 33: pp. 947 – 953, (1994).
[18] FELLINGER, K., SCHMIDT, J., Wien Z. Inn. Med., 33, 351 (1952).
[19] CLUNIE, G., PETER, J., A survey of radiation synovectomy in Europe, 1991 –
1993, European Journal of Nuclear Medicine Vol.22, No. 9, September, (1995).
[20] OHASHI, F., et al., The production os arthritis in beagles by an immunological
reaction to bovine serum albumin, Exp. Anim. 45 (4), pp. 299 – 307, (1996).
[21] JAHANGIER, Z.N., JACOBS, J.W.G., VAN ISSELT, J.W., BIJLSMA, W.J.,
Persistent synovitis treated with radiation synovectomy using Yttrium-90: a retro-
spective evaluation of 83 procedures for 45 patients, British Journal of Rheuma-
tology 36: pp. 861 – 869, (1997).
[22] WUNDERLICH, G., et al., Preparation and biodistribution of Rhenium-188
labeled albumin microspheres b 20: a promising new agent for radiotherapy,
Applied Radiation and Isotopes 52: pp. 63 – 68, (2000).
[23] HEUFT-DORENBOSCH, L.L.J., DE VET, H.C.W., VAN DER LINDEN, S.,
Yttrium radiosynoviortheses in the treatment of knee arthritis in rheumatoid
arthritis: A systematic review, Ann. Rheum. Diss. 59 (2000) 583–586.
[24] KAMPEN, W.U., BRENNER, W., CZECH, N., HENZE, E., Intraarticular appli-
cation of unsealed beta-emitting radionuclides in the treatment course of inflam-
matory joint diseases, Curr. Med. Chem. Anti-inflamatory & Anti-allergy Agents
1 (2002) 77–87.
[25] VUORELA, J., SOKKA, T., PUKKALA, E., HANNONEN, P., Does Yttrium
radiosynovectomy increase the risk of cancer in patients with rheumatoid
arthritis?, Ann. Rheum. Dis. 62: pp. 251 – 253, (2003).
[26] EUROPEAN ASSOCIATION OF NUCLEAR MEDICINE, EANM procedure
guidelines for radiosynovectomy, Eur. J. Nucl. Med. BP12 – PB16 Vol . 30, No. 3,
March (2003).
[27] ERSELCAN, T., et al., Lypoma arborescens; successfully treated by Yttrium-90
radiosynovectomy, Annals of Nuclear Medicine, Vol. 17, No. 7, pp. 593 – 596,
(2003).
[28] SILVA, M., LUCK, J.V., LLINÁS, A., Chronic hemophilic synovitis: the role of
radiosynovectomy, Treatment of Hemophilia, 33, April (2004).
[29] MÄKELÄLA, O., et al., Experimental radiation synovectomy in rabbit knee with
holmium-166 ferric hydroxide macroaggregate, Nucl. Med. Biol. 29 (2002) 593–
598.
[30] WANG, S.-J., et al., Histologic study of effects of radiation synovectomy with
rhenium-188 microsphere, Nucl. Med. Biol. 28 (2001) 727–732.
22
SESSION 8
23
.
TARGETED RADIOTHERAPY WITH ALPHA
PARTICLE EMITTING RADIONUCLIDES
Abstract
1. INTRODUCTION
25
ZALUTSKY et al.
26
SESSION 8
energy transfer (LET) quality to this radiation. Yttrium-90 emits high energy
b particles that have a mean LET of about 0.2 keV/mm; in comparison, the LET
of clinically relevant a particle emitters is several orders of magnitude higher,
about 100 keV/mm. The radiobiological implications of high LET radiation are
perhaps the most compelling rationale for pursuing a particle emitters for
therapy [6]. Of primary importance is the fact that the relative biological effec-
tiveness of high LET a particles is considerably higher than b particles or
standard external beam radiation. Radiation at 100 keV/mm is particularly
cytotoxic because the distance between ionizing events at this LET is nearly
identical to that between DNA strands, increasing the probability of creating
highly cytotoxic double DNA strand breaks. The predicted exquisite cytotox-
icity of targeted a particle emitting radiopharmaceuticals has been validated
experimentally with a variety of radionuclides, carrier molecules and human
cancer cell lines [7]. In addition, the conditions under which high LET radiation
is maximally effective are relatively wide ranging, not being compromised by a
lack of oxygen, cell cycle stage or dose rate.
27
ZALUTSKY et al.
28
SESSION 8
4. TERBIUM-149
29
ZALUTSKY et al.
5.55 MBq 149Tb, or saline control [11]. At 120 d, tumour free survival was
observed in all but one animal receiving the labelled mAb but none of the
animals receiving cold mAb or vehicle alone. Although these results at first
glance appear to be encouraging, this interpretation must be made with
caution. Treatment only two days after injection of tumour cells presents a
rather favourable model in which problems related to homogeneous delivery of
radionuclide are minimized. Additionally, particularly for a model of this type,
the most critical control was not performed, namely, a 149Tb labelled non-
specific control mAb.
In summary, the principal attractions of 149Tb as an a particle emitter for
targeted radiotherapy are its 4.15 h half-life, which could be a good match for
smaller molecular weight tumour avid compounds, as well as its adaptability to
radiochemistries already developed for other radiometals such as 90Y and
177
Lu. On the other hand, the limited availablity of 149Tb and its low a particle
abundance are signficant disadvantages. Finally, the lack of compelling
evidence that specific and selective tumour cell kill can be achieved with a 149Tb
labelled molecule is also of concern.
5. RADIUM-223
30
SESSION 8
peutic efforts with 223Ra has involved exploiting the natural affinity of radium
for bone for the treatment of skeletal metastases. An a emitter could be ideal
for this type of clinical application because the shorter particle range,
compared with b emitters, should greatly increase the capability of delivering
high radiation doses to bone metastases while minimizing excessive dose to
radiation sensitive bone marrow, which otherwise could be dose limiting.
Because 89Sr was used routinely in the past as a bone seeking radiophar-
maceutical, the biodistribution and radiation dosimetry of 223Ra has been
compared to that of 89Sr, with both being administered as their chloride salts
[14]. The uptake of 223Ra in the bone was significantly higher than that of 89Sr,
reaching a level of 40% injected dose per gram in the femur at 24 h post-
injection. Importantly, no significant decline in bone retention was seen over
the course of the 14 d experiment and redistribution of progeny from the bone
was quite small. Accumulation of 223Ra was also observed in soft tissues, partic-
ularly the spleen and the kidneys. However, femur:soft tissue radiation dose
ratios were extremely favourable, generally greater than 1000:1. Dosimetry
calculations modelled dose to bone and bone marrow and indicated that 223Ra
should offer significant advantages compared with 89Sr with regard to
minimizing bone marrow radiation dose.
Preclinical evaluation of 223Ra has also been performed in rats including
those with skeletal metastases [15]. Initital studies confirmed the bone seeking
properties of the radiopharmaceutical, with femur:kidney, femur:liver and
femur:spleen ratios of 590:1, 620:1 and 810:1 observed 24 h after injection. The
femur:bone marrow ratio increased from 6.5:1 at 24 h to >15:1 at 14 d. For the
therapy studies, rats were injected with 1 × 106 MT-1 human breast cancer cells
into the left ventricle and then treated one week later. Control animals had to
be killed 20–30 d later because of tumour induced paralysis while 36 or 40% of
rats receiving 10 or 11 kBq 223Ra had significantly longer symptom free survival
over the 67 d follow-up period. No sign of bone marrow toxicity was observed,
indicating that this radiopharmaceutical warranted evaluation as an endoradio-
therapeutic in cancer patients.
Recently, the results of the first clinical trial of 223Ra in patients with
skeletal metastases were reported [16]. In this phase I trial, 25 patients
(10 breast cancer, 15 prostate cancer) were treated in cohorts of five patients at
doses of 46, 93, 163, 213 or 250 kBq per kg body weight. The labelled
compound was well tolerated with myelosuppression being mild and reversible.
No dose limiting toxicites were observed at any dose level. Median survival for
the 25 patients was greater than 20 months which is encouraging; however, the
lack of a control group makes it difficult to draw meaningful conclusions about
the survival benefit of this treatment. Such a comparison will be done in due
31
ZALUTSKY et al.
6. BISMUTH-213
32
SESSION 8
7. ACTINIUM-225
33
ZALUTSKY et al.
8. ASTATINE-211
34
SESSION 8
residues. The most common tactic for labelling biomolecules with 211At is via an
astatodemetallation reaction and a wide variety of 211At labelled molecules
have been synthesized and evaluated as potential targeted radiotherapeutics
[7]. The authors’ own efforts in the small molecule arena have included 211At
labelled meta-iodobenzylguanidine analogues, biotin conjugates, peptides,
bisphosphonates and thymidine analogues [7]. By far the most active area of
research both at the authors’ institution and elsewhere has been in evaluation
of 211At labelled mAbs and mAb fragments as a particle emitting targeted
radiotherapeutics. Promising results have been obtained with 211At labelled
immunoconjugates directed at many types of cancer including brain, ovarian,
osteosarcoma, melanoma and non-Hodgkins lymphoma [42–48]. A clinical trial
of 211At labelled chimeric 81C6 mAb administered directly into surgically
created tumour resection cavities is currently under way at the authors’
institution and the results thus far have been very encouraging [49].
9. CONCLUSIONS
ACKNOWLEDGEMENTS
REFERENCES
35
ZALUTSKY et al.
[3] SAUTTER-BIHL, M.-L., et al., Minimal residual disease: a target for radioimmu-
notherapy with 131I-labeled monoclonal antibodies? Some dosimetric considera-
tions. Recent Results Cancer Res. 141 (1996) 67
[4] HUMM, J.L., Dosimetric aspects of radiolabeled antibodies for tumor therapy. J.
Nucl. Med. 27 (1986) 1490
[5] HUMM, J.L., A microdosimetric model of astatine-211 labeled antibodies for
radioimmunotherapy. Int. J. Radiat. Oncol. Biol. Phys. 13 (1987) 1767.
[6] HALL, E.J., Radiobiology for the Radiologist 5th edition. J. B. Lippincott,
Williams, and Wilkins, Philadelphia (2000).
[7] ZALUTSKY, M.R., VAIDYANATHAN, G. Astatine-211-labeled radiotherapeu-
tics: an emerging approach to targeted alpha particle therapy. Current Pharm.
Design 6 (2000) 1433.
[8] ALLEN, B.J., BLAGOJEVIC, N., Alpha and beta emitting radiolanthanides in
targeted cancer therapy: the potential tole of Terbium-149. Nucl. Med. Comm. 17
(1996) 40.
[9] BEYER, G.J., Radioactive ion beams for biomedical research and application.
Hyperfine Interact. 129 (2000) 529.
[10] MIEDERER, M., et al., Comparison of the radiotoxicity of two alpha-particle-
emitting immunoconjugates, terbium-149 and bismuth-214, directed against a
tumor-specific, exon 9 deleted (d9) E-cadherin adhesion protein. Radiation Res.
159 (2003) 612.
[11] BEYER, G.J., et al., Targeted alpha particle therapy in vivo: direct evidence for
single cancer cell kill using 149Tb-rituximab. Eur. J. Nucl. Med. Mol. Imaging 31
(2004) 547.
[12] HENRIKSEN, G., et al., 223Ra for endoradiotherapeutic applications prepared
from an immobilized 227Ac/227Th source. Radiochim. Acta 89 (2001) 66.
[13] HENRIKSEN, G., et al., Sterically stabilized liposomes as a carrier for α-emitting
radium and actinium radionuclides. Nucl. Med. Biol. 31 (2004) 441.
[14] HENRIKSEN, G., et al., Targeting of osseous sites with α-emitting 223Ra: compar-
ison with the β-emitter 89Sr in mice. J. Nucl. Med. 44 (2003) 252.
[15] HENRIKSEN, G., et al., Significant antitumor effect from bone-seeking
α-particle-emitting 223Ra demonstrated in an experimental skeletal metastases
model. Cancer Res. 62 (2002) 3120.
[16] NILSSON, S., et al., First clinical experience with α-emitting radium-223 in the
treatment of skeletal metastases. Clin. Cancer Res. 11 (2005) 4451.
[17] MCDEVITT, M.R., et al., Preparation of α-emitting 213Bi-labeled antibody
constructs for clinical use. J. Nucl. Med. 40 (1999) 1722.
[18] MA, D., et al., Breakthrough of 225Ac and its radionuclide daughters from an
225
Ac/213Bi generator: development of new methods, quantitative characteriza-
tion, and implications for clinical use. Appl. Radiat. Isotop. 55 (2001) 667.
[19] MCDEVITT, M.R., et al., An alpha-particle emitting antibody ([213Bi]J591) for
radioimmunotherapy of prostate cancer. Cancer Res. 60 (2000) 6095.
36
SESSION 8
37
ZALUTSKY et al.
[36] MIEDERER, M., et al., Pharmacokinetics, dosimetry, and toxicity of the targ-
etable atomic generator, 225Ac-HuM195, in nonhuman primates. J. Nucl. Med. 45
(2004) 129.
[37] TURKINGTON, T.G., et al., Measuring astatine-211 distributions with SPECT.
Phys. Med. Biol. 38 (1993) 1121.
[38] JOHNSON, E.L., et al., Quantitation of 211At in small volumes for evaluation of
targeted radiotherapy in animal models. Nucl. Med. Biol. 22 (1995) 45.
[39] FRIEDMAN, A.M., et al., Preparation of a biologically stable and immuno-
genically competent astatinated protein. Int. J. Nucl. Med. Biol. 4 (1977) 219.
[40] LARSEN, R.H., et al., Evaluation of an internal cyclotron target for the produc-
tion of astatine-211 via the 209Bi(α,2n)211At reaction. Appl. Radiat. Isotop. 47
(1996) 135.
[41] ZALUTSKY, M.R., et al., High-level production of alpha-particle-emitting 211At
and preparation of 211At-labeled antibodies for clinical use. J. Nucl. Med. 42
(2001) 1508.
[42] YORDANOV, A.T., et al., Preparation and in vivo evaluation of linkers for 211At
labeling of humanized anti-Tac. Nucl. Med. Biol. 28 (2001) 845.
[43] AURLIEN, E., et al., Radiation doses to non-Hodgkin's lymphoma cells and
normal bone marrow exposed in vitro. Comparison of an alpha-emitting radioim-
munoconjugate and external gamma-irradiation. Int. J. Radiat. Biol. 78 (2002)
133.
[44] AURLIEN, E., et al., Exposure of human osteosarcoma and bone marrow cells to
tumour-targeted alpha-particles and gamma-irradiation: analysis of cell survival
and microdosimetry. Int. J. Radiat. Biol. 76 (2000) 1129.
[45] LARSEN, R.H., et al., The cytotoxicity and microdosimetry of astatine-211-
labeled chimeric monoclonal antibodies in human glioma and melanoma cells in
vitro. Radiat. Res. 149 (1998) 155.
[46] ANDERSSON, H., et al., Astatine-211-labeled antibodies for treatment of
disseminated ovarian cancer: an overview of results in an ovarian tumor model.
Clin. Cancer Res. 9 (suppl) (2003) 3914S.
[47] KENNEL, S.J., et al., Vascular-targeted radioimmunotherapy with the alpha-
particle emitter 211At. Radiat. Res. 157 (2002) 633.
[48] AKABANI, G., et al., Vascular targeted endoradiotherapy using alpha-particle
emitting compounds: theoretical analysis. Int. J. Radiat. Oncol. Biol. Phys. 54
(2002) 1259.
[49] ZALUTSKY, M.R., et al., Astatine-211 labeled human/mouse chimeric anti-
tenascin monoclonal antibody via surgically created resection cavities for patients
with recurrent glioma: Phase I study. Neuro-Oncol. 4 (2002) S103 (abstract).
38
DOTA-TYR3-OCTREOTATE LABELLED WITH 177Lu
AND 131I
A comparative evaluation
Bucharest, Romania
Abstract
1. INTRODUCTION
39
LUNGU et al.
2.1. Materials
All chemicals were purchased from Fluka Chemical and Sigma Aldrich.
The 177LuCl3 and Na131I, with specific activities of 45 Ci/mg and
1600 Ci/mg respectively, were purchased from Nordion Canada.
DOTA-Tyr3-TATE and Sandostatin were obtained from the IAEA and
from Pichem Austria.
Wistar rats (150–200 g) were used for the preparation of cortex membranes
and Lewis rats were used for biodistribution studies. The HRS1 tumour bearing
rats were prepared in the Institute of Oncology Bucharest, Romania.
Radioactive samples were counted on Robotron and Spectroscaler
gamma counters.
2.2. Methods
40
SESSION 8
The competition binding assay was performed using rat brain cortex
membrane (50 mg protein). 35 000–40 000 counts/min of 125I-Tyr3-octreotide
(970 Ci/mM) were added in each test tube in the presence of increasing concen-
tration of DOTA-natI-Tyr3-TATE (synthesized in the same conditions as
DOTA-131I-Tyr3-TATE): 0.06; 0.13; 0.34; 0.68; 1.00; 2.72; 10.00; 57.80; 100nM in
total volume of 300 mL 50mM HEPES (pH7.6, 0.3% BSA, 5mM MgCl2, 10mM
bacitracin). The samples were incubated for 2 h at room temperature; the
incubation was stopped by addition of ice cold buffer (1 mL, 10mM HEPES,
150mM NaCl, pH7.6).
The suspension was rapidly filtered over glass fibre filters (Whatman GF/B)
presoaked in binding buffer using a Millipore multifiltration apparatus. The
filters were rinsed with buffer (4 × 2 mL) and filter activity was measured on a
NaI(Tl) gamma counter.
41
LUNGU et al.
A similar method (Section 2.2.6) was used. For the experiments regarding
saturation binding assay of 177Lu-DOTA-Tyr3-TATE, the following concentra-
tions of 177Lu-DOTA-TATE were prepared: 0.05; 0.15; 4.5; 13.5; 40.5; 120.5;
361.5nM for total and non-specific binding assay. A method similar to that
described in Section 2.2.6 was used for the processing of the obtained samples
and acquisition of data.
42
SESSION 8
43
LUNGU et al.
Best-fit values
35000 BOTTOM 239.5
TOP 32796
30000
LOGEC50 -8.893
25000 EC50 1.280e-009
Mean cpm
Std. Error
20000
BOTTOM 1815
15000 TOP 3338
LOGEC50 0.1711
10000
5000
0
-10 -9 -8 -7
log[somatostatin](M)
14000
12000
10000
Mean (CPM)
8000
6000
4000
2000
0
-10 -9 -8 -7 -6
Log([somatostatin])
44
SESSION 8
Best-fit values
0.35
Slope -0.006346 ± 0.0006098
0.30 Y-intercept when X=0.0 0.3568 ± 0.02275
X-intercept when Y=0.0 56.22
0.25
1/slope -157.6
Bound/Free
0.20
0.15
0.10
0.05
0.00
10 20 30 40 50 60 70
-0.05
Bound (pM)
FIG. 3. Saturation binding curve of DOTA-131I-Tyr3-TATE.
0.6
0.5
0.4
Bound/Free
0.3
0.2
0.1
0.0
0 20 40 60
Bound (pM)
177
FIG. 4. Saturation binding curve of Lu-DOTA-Tyr3-TATE. Fit line: y = A + Bx:
A = 0.48 ± 0.03; B = -0.007 ± 0.0008 pM.
45
LUNGU et al.
Organ 3h 24 h 48 h 72 h 168 h
Blood 0.72 ± 0.47 0.04 ± 0.03 0.04 ± 0.01 0.03 ± 0.01 0.03 ± 0.00
Liver 0.68 ± 0.15 0.44 ± 0.11 0.40 ± 0.05 0.28 ± 0.13 0.18 ± 0.01
Spleen 0.20 ± 0.11 0.14 ± 0.05 0.07 ± 0.01 0.03 ± 0.05 0.03 ± 0.00
Kidney 2.12 ± 0.23 1.90 ± 0.78 1.55 ± 0.82 1.56 ± 0.02 1.23 ± 0.05
Stomach 1.95 ± 0.42 1.25 ± 0.03 1.08 ± 0.32 0.94 ± 0.08 0.79 ± 0.01
Small intestine 0.92 ± 0.37 0.89 ± 0.13 0.57 ± 0.22 0.30 ± 0.10 0.17 ± 0.01
Large intestine 2.66 ± 0.14 2.02 ± 0.10 1.73 ± 0.10 1.99 ± 0.21 0.73 ± 0.02
Adrenal 0.71 ± 0.32 0.76 ± 0.51 0.75 ± 0.16 0.50 ± 0.11 0.35 ± 0.01
Pancreas 7.79 ± 0.08 4.10 ± 0.92 3.67 ± 0.67 2.01 ± 0.31 1.50 ± 0.02
Thyroid 1.92 ± 0.21 2.09 ± 0.14 0.82 ± 0.13 0.97 ± 0.08 0.73 ± 0.10
Pituitary 0.37 ± 0.13 0.57 ± 0.17 0.39 ± 0.11 0.34 ± 0.02 0.12 ± 0.01
Bone 9.54 ± 0.27 5.40 ± 0.72 2.79 ± 0.53 1.72 ± 0.31 1.05 ± 0.15
Lung 0.24 ± 0.11 0.17 ± 0.08 0.12 ± 0.05 0.05 ± 0.01 0.02 ± 0.01
Tumour 3.22 ± 2.15 6.31 ± 3.02 4.18 ± 3.42 4.07 ± 2.58 0.61 ± 0.29
46
SESSION 8
9
3h 24 h 48 h 72 h 168 h
8
7
ID (%) / organ (g)
0
Blood Liver Kidney Adrenals Pancreas Tumor
organ
bone was tested. The results (see Fig. 6.) show a progressive accumulation of
177
Lu, with the maximum value in the 4–24 h range. The high level of radio-
activity was maintained for the duration of the experiment; the kinetics of
177
LuCl3 appear different from those obtained for 177Lu-DOTA-Tyr3-TATE.
Lu chloride Lu peptide
18
16
14
ID (%) / organ (g)
12
10
0
3h 24 h 48 h 72 h 168 h
Time
47
LUNGU et al.
Because the uptake in the non-tumour tissues and the clearance of 177Lu-
131
and I-DOTA-TATE were similar, the present study examines whether one
radioproduct is more stable and has more uptake than the other in tumour
bearing rats co-injected with 177Lu-DOTA-TATE and 131I-DOTA-TATE.
The competitive localization index (CLI) was defined:
177
The results (see Table 2 and Fig. 7) show that Lu-CLI is higher than
131
I-CLI (~3 times).
177
TABLE 2. COMPETITIVE LOCALIZATION INDEX OF Lu-DOTA-
Tyr3-TATE AND DOTA-131I-Tyr3-TATE IN TUMOUR
CLI 3h 24 h 48 h 72 h 168 h
177
Lu-CLI 4.87 157.75 104.50 69 20.38
131
I-CLI 1.42 103.00 29.81 6.38 3.08
48
SESSION 8
160
140
120
100
80
60
40
20
0
3h 24 h 48 h 72 h 168 h
Time (h)
4. CONCLUSIONS
49
LUNGU et al.
ACKNOWLEDGEMENTS
REFERENCES
[1] WEN, PING LI, et al., DOTA-D-Tyr – Octreotate: A Somatostatine analogue for
labeling with metal and halogen radionuclides for cancer imaging and therapy,
Bioconjugate Chem. 13 (2002) 721-728.
[2] BAKKER, W.H., et al., Iodine-131 labelled octreotide: not an option for somato-
statine receptor therapy, Eur. J. Nucl. Med. 23 7 (1996) 775-781.
[3] MAINA, T., et al., [99mTc] Demotate, a new 99mTc-based [Tyr3] octreotate
analogue for the detection of somatostatine receptor – positive tumours: synthesis
and preclinical results, Eur. J. Nucl. Med. 29 6 (2002) 742-753.
[4] KWEKKEBOOM, D.J., et al., [177Lu-DOTA0,Tyr3] octreotate: comparison with
[111In-DTPA0]octreotide in patients, Eur. J. Nucl. Med. 28 9(2001) 1319-1325.
[5] LEWIS, J.S., et al., Toxicity and dosimetry of 177Lu-DOTA-Y3-octreotate in a rat
model, Int. J. Cancer 94 (2001) 873-877.
[6] LEWIS, J.S., et al., Radiotherapy and dosimetry of 64Cu-TETA-Tyr3 -Octreotate
in a somatostative receptor-positive, tumor-bearing rat model, Clinical Cancer
Research 5 (1999) 3608-3616.
[7] VALKEMA, R., et al., Long-term folow-up of renal function after peptide
receptor radiation therapy with 90Y-DOTA0, Tyr3 – Octreotide and 177Lu-DOTA0,
Tyr3 -Octreotate, J. Nucl. Med. 46 1 (2005) 995-1065.
[8] KWEKKEBOOM, D.J., et al., Overview of results of peptide receptor radionu-
clide therapy with 3 radiolabeled somatostatin analogs, J. Nucl. Med. 45 11 (2004)
1-5.
[9] RAYNOR, K., REISINE, T., Analogs of somatostatin selectively label distinct
serbtypes of somatostatin receptor in ratbrain, J. Pharmacol Exp. Ther. 251 (1989)
510-517.
50
177
Lu LABELLED NITROIMADZOLES AND
NITROTRIAZOLES FOR POSSIBLE USE IN
TARGETED THERAPY OF HYPOXIC TUMOURS
*Radiopharmaceuticals Division
Email: meerav@apsara.barc.ernet.in
Abstract
1. INTRODUCTION
51
DAS et al.
as potent tumour avid substrates, are chosen as the carrier molecules [4, 5].
Lutetium-177, which is fast emerging as a promising radionuclide for targeted
therapy owing to its suitable decay characteristics (T1/2 = 6.73 d, Eβ(max) =
497 keV and Eγ = 113 keV (6.4%), 208 keV (11%)) and favourable production
logistics (σ = 2100 b for 176Lu(n,γ)177Lu), was identified as the radioisotope of
choice [6]. The relatively longer half-life of 177Lu provides logistical advantages
for facilitating supply to places far away from the reactors and assumes signifi-
cance for those countries having limited reactor facilities for isotope
production. Moreover, 177Lu can be produced in adequate specific activity for
in vivo targeted therapy applications by a simple (n,γ) process using an
enriched 176Lu target due to its very high neutron capture cross-section (σ =
2100 b) [6, 7]. Since direct incorporation of 177Lu in either of the aforemen-
tioned nitroimidazole or nitrotriazole moiety is not feasible, indirect incorpo-
ration of 177Lu through a suitable bifunctional chelating agent (BFCA) was
envisaged. As it is well documented that the lanthanide complexes of poly-
azamacrocycles exhibit very high thermodynamic stability and excellent kinetic
inertness, it is quite logical to choose those ligands as the BFCAs of choice [8].
For the present study, metronidazole was coupled with para-aminobenzyl-
1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (p-amino-benzyl-
DOTA) while the sanazole derivative [(N-2’(carboxyethyl)-2-(3’-nitro-1’-tri-
azolyl)acetamide] was conjugated with 1,4,7,10-tetraaza-1-(4’-aminobenzyl-
acetamido)-cyclododecane-4,7,10-triacetic acid (p-amino-DOTA-anilide). The
present paper describes the radiolabelling of both the conjugates with 177Lu in
high yields and the pharmacokinetic behaviour of the developed radiochemical
agents in suitable tumour bearing animal models.
2. EXPERIMENTAL
2.1. Materials
52
SESSION 8
2.3.1. Metronidazole-p-amino-benzyl-DOTA
53
DAS et al.
left overnight at room temperature (25°C). The reaction mixture was filtered
and the filtrate was acidified and extracted in diethyl ether. The oxidized
product, 2-[N-(2’-methyl-5’-nitro)imidazolyl]ethanoic acid, was obtained by
evaporating the solvent under vacuum. In the second step, 2-[N-(2’-methyl-5’-
nitro)imidazolyl]ethanoic acid (3 mg, 0.0162mM), p-NH2-benzyl-DOTA.4HCl
(10 mg, 0.0152mM) and DCC (3 mg, 0.0146mM) were stirred in dioxan for 20 h
at room temperature to obtain the desired conjugate. After the completion of
reaction, the reaction mixture was filtered and the filtrate was evaporated. The
crude product thus obtained was purified by preparative TLC using 3%
ammonium hydroxide in methanol as the eluting solvent.
2.3.2. Sanazole-p-amino-DOTA-anilide
54
SESSION 8
55
DAS et al.
177m
Lu, the activity of 177Lu was allowed to decay completely (45–65 d, ~8–10
half-lives of 177Lu) and the gamma ray spectrum was recorded. The average
level of radionuclidic impurity burden was found to be 150 nCi of 177mLu /1 mCi
of 177Lu (5.5 kBq /37 MBq) at end of bombardment.
3.2.1. Metronidazole-p-amino-benzyl-DOTA
FT-IR (KBr, ν cm-1): 3425, 2925, 2849, 1733, 1692, 1514, 1469, 1385.
1
H-NMR, CD3OD (δ ppm): 1.29 [3H, s, metronidazole CH3], 3.56-3.63
[15H, m, DOTA cyclic CH2], 3.67-3.74 [8H, m, N(DOTA)-CH2-COOH], 4.08-
4.20 [2H, m, CH(DOTA)-CH2-Ph], 4.94 [2H, s, N(metronidazole)-CH2-CO],
7.44-7.49 [2H, m, p-amino-benzyl-DOTA ArH], 7.58-7.62 [2H, m, p-amino-
benzyl-DOTA ArH], 8.03 [1H, s, metronidazole H].
The peak positions and integrations observed in the 1H-NMR spectrum
of the conjugate were consistent with the structure of the expected compound.
In addition to the protons assignable to the DOTA moiety, the presence of the
peaks at δ = 1.29 and 4.94 corresponding to the protons of the metronidazole
moiety and the deshielded proton of the imidazole ring of metronidazole
observable at δ = 8.03 provided further evidence towards the desired
derivatization.
3.2.2. Sanazole-p-amino-DOTA-anilide
FT-IR (KBr, ν cm-1): 3443, 3096, 2966, 2853, 1682, 1631, 1555.
1
H-NMR (CD3OD, δ ppm): 3.40-3.78 [16H, m, cyclic CH2], 4.11-4.24 [6H,
m, N(DOTA)-CH2-COOH] 4.30-4.38 [2H, m, N(DOTA)-CH2-CO-NH], 4.76
[2H, dd, J = 2.2 Hz & 14.3 Hz, sanazole-N-CH2-CO], 5.11 [4H, dd, J = 9.9 Hz &
23 Hz, -CO-CH2-CH2-CO], 8.07 [2H, bs, CONH-aromatic H], 8.09 [1H, s,
sanazole H], 8.11-8.13 [2H, m, CONH-aromatic H].
The peak positions, multiplicities and integrations observed in the high
resolution 1H-NMR spectrum of the conjugate were consistent with the
structure of the expected compound. Appearance of the peaks at δ = 5.11 and
4.76 corresponding to the protons of the sanazole moiety indicates the desired
conjugation. The deshielded proton of the triazole ring of sanazole observable
at δ = 8.09 provides further evidence towards the desired derivatization.
56
SESSION 8
FIG. 1. (a) HPLC pattern of 177Lu labelled metronidazole-BFCA conjugate, (b) HPLC
pattern of 177Lu labelled sanazole-BFCA conjugate.
57
DAS et al.
58
SESSION 8
%ID/g
Organ
1h 3h 24 h
Note: Figures in parentheses show standard deviations. Three animals were used for
each time point studied.
pertinent to draw a comparison with respect to the tumour uptake and target to
non-target ratios of the radiolabelled conjugates described in the present paper
with a few standard agents intended for use in hypoxia imaging, such as 99mTc-
BMS181321 [10] and 99mTc-BRU59-21 [11], which indicates that the developed
agents exhibit superior tumour to blood and tumour to muscle ratios at the
time points studied.
4. CONCLUSION
59
DAS et al.
Organ %ID/g
1h 3h 24 h
Note: Figures in parentheses show standard deviations. Three animals were used for
each time point studied.
ACKNOWLEDGEMENTS
60
SESSION 8
REFERENCES
[1] NUNN, A., LINDER, K., STRAUSS, H.W., Nitroimidazoles and imaging
hypoxia, Eur. J. Nucl. Med. 22 (1995) 265.
[2] BROWN, J.M., Hypoxic cell radiosensitizers: What next?, Int. J. Radiat. Oncol.
Biol. Phys. 16 (1989), 987.
[3] CHAPMAN, J.D., Hypoxic sensitizers: Implications for radiation therapy, N.
Engl. J. Med. 301 (1979) 1429.
[4] DAS, T., et al., 99mTc-labeled modified metronidazole: A potential agent for
imaging hypoxia, Nucl. Med. Biol. 30 (2003) 127.
[5] SHIBAMOTO, Y., et al., Evaluation of various types of new hypoxic cell sensi-
tizers using the EMT6 single cell-spheroid-solid tumor system. Int. J. Radiat. Biol.
52 (1987) 347.
[6] FIRESTONE, R., Table of isotopes (Shirley, V.S., Ed.), John Wiley and Sons, New
York (1996).
[7] PILLAI, M.R.A., CHAKRABORTY, S., DAS, T., VENKATESH, M., RAMA-
MOORTHY, N., Production logistics of 177Lu for radionuclide therapy. Appl.
Radiat. Isot. 59 (2003)109.
[8] LIU, S., EDWARDS, D.S., Bifunctional chelators for therapeutic lanthanide radi-
opharmaceuticals, Bioconj. Chem. 12 (2001) 7.
[9] VOGEL, A.I., Textbook of practical organic chemistry, Longman Scientific
Group, London (1994).
[10] BALLINGER, J.R., MIN-KEE, J.W., RAUTH, A.M., In vitro and in vivo evalua-
tion of a technetium-99m labeled 2-nitroimidazole (BMS 181321) as marker of
tumor hypoxia, J. Nucl. Med. 37 (1996) 1023.
[11] MELO, T., DUNCAN, J., BALLINGER, J.R., RAUTH, A.M., BRU59-21, a
second-generation 99mTc-labeled 2-nitroimidazole for imaging hypoxia in tumors,
J. Nucl. Med. 41 (2000)169.
61
.
LABELLING AND BIOLOGICAL EVALUATION OF
ANTI-CD20 FOR TREATMENT OF NON-HODGKIN’S
LYMPHOMA
Abstract
A radiopharmaceutical based on the use of Mab anti-CD20 labelled with 131I and
188
Re is proposed for the treatment of non-Hodgkin’s lymphoma. The antibody has been
successfully used alone as well as associated with cytotoxic drugs, therefore encouraging
the present research. The radionuclides were chosen on the basis of their decay proper-
ties and availability. Labelling techniques employed were previously used with other
monoclonals. Oxidation with chloramine-T was used for 131I labelling and SnF2·2H2O as
the reducing agent for 188Re. Non-specific precipitation and Sephadex purification were
used for primary control and extraction. Quality control procedures including thin layer
as well as high performance liquid chromatography were undertaken. Biodistribution in
mice as well as affinity characteristics in a source rich in CD20 antigens were also
studied. Stability over time was estimated. It was concluded that the labelling of anti-
CD20 with ß emitters of therapeutic interest, in this case 131I and 188Re, gave reliable
results by simple and efficient methodologies, yielding products compatible with clinical
radioimmunotherapy. Quality control methods for the evaluation of radiochemical
purity showed good reproducibility with short bench time. Immunoaffinity studies
showed binding dependency on membrane antigen concentration and good specificity
of binding was demonstrated by inhibition with unlabelled anti-CD20. Use of
membranes, stable at -80ºC for more than 6 months instead of concentrated short lived
leucocytes, has shown excellent reproducibility and therefore they are convenient at
production centres distant from blood banks. Biodistributions were useful to determine
the normal pattern and kinetics of uptake and excretion.
1. INTRODUCTION
63
OLIVER et al.
2. MATERIALS
64
SESSION 8
3. METHODOLOGY
The 131I- was introduced in one tyrosyl residue of the protein chain by
adding 28 MBq to 20 µg of anti-CD20 at pH7.4 and 10 µL of chloramine-T
(0.13 µg/µL). After 1 min reaction time at room temperature, the yield was
determined by protein precipitation with trichloroacetic acid solution (10%)
and purification was done by gel permeation with Sephadex G-25 eluting with
phosphate saline buffer 50mM, 0.2% BSA. Specific activity and iodine incorpo-
ration were determined.
For the labelling with 188Re, anti-CD20 was first reduced by incubation
with 2-mercaptoethanol to expose sulphydril groups and then purified by gel
permeation over a PD10 column. Fractions of reduced antibody were pooled
and formulated as kit for instant labelling. Each kit contained 1 mg anti-CD20;
82.8 mg of sodium tartrate; 1.67 mg of stannous fluoride and 0.25 mg gentisic
acid. For the labelling, sodium perrhenate (1.5–1.9 GBq), previously acidified,
was added to the kit and then incubated for 1 h at room temperature. The
radiochemical purity of 188Re-anti-CD20 was evaluated by ITLC-SG using
MEK and saline as solvents and by saturated ITLC-SG strips (BSA 5%) using
EtOH-NH4OH-H2O (2:1:5). It was also evaluated by HPLC using an SW300
protein Pak column and eluting with phosphate buffer 0.01M, pH7.4 at
1.0 mL/min. Specific activity was determined.
65
OLIVER et al.
4. RESULTS
66
SESSION 8
45
40
35
30
act (%)
25
20
15
10
0
5 2 5 5 5 8 5 11 .5 14
0. 3. 6. 9. 12
eluate (mL)
FIG. 1. Purification profile of mab anti-CD20 labelled with 131I on PD10 columns.
2000
1500
B (cpm)
1000
500
0
0 4 8 12 16
67
OLIVER et al.
4000
3000
B (cpm)
2000
1000
0
0.00 0.25 0.50 0.75 1.00 1.25 1.50 1.75 2.00
Anti-CD20 (mg/mL)
100
80
60
% Binding
40
20
0
0 5 10 15 20 25 30 35
CD20 Membranes (mg/mL)
68
SESSION 8
8000
6000
B (cpm)
4000
2000
0
0 1 2 3 4
Anti-CD20 (mg/mL)
69
OLIVER et al.
5. CONCLUSIONS
ACKNOWLEDGEMENTS
REFERENCES
70
THERAPEUTIC RADIOPHARMACEUTICALS
(Session 9)
Chairpersons
J. HARVEY TURNER
Australia
M. DONDI
IAEA
.
177
Lu-DOTA-J591 MONOCLONAL ANTIBODY:
CHEMISTRY, TOXICITY, DOSIMETRY AND
CLINICAL EFFICACY
RIT of prostate cancer using 177Lu-J591 anti-PSMA antibody
Abstract
73
GOLDSMITH et al.
1. INTRODUCTION
177
1.1. Lu for therapy
74
SESSION 9
75
GOLDSMITH et al.
177
1.4. Lu-DOTA-J591
The clinical grade de-immunized J591 mAb was produced under GMP
conditions at Lonza Biologics (Slough, United Kingdom) and supplied in 5 mL
of phosphate buffer, pH7.0 containing 5 mg/mL of antibody. The J591 antibody
was covalently linked with the chelating agent, 1,4,7,10-tetraazacyclododecane-
N,N’,N’’,N’’’-tetraacetic acid (DOTA) (Goodwin Biotech, Plantation, United
States of America) as previously reported [22]. The sterile pyrogen free clinical
material, DOTA-J591 mAb in 0.3M ammonium acetate buffer, pH7.0 (8 mg/mL)
was provided by BZL Biologics (Framingham, USA).
The DOTA-J591 mAb was labelled by incubating the antibody in an
ammonium acetate buffer (1M, pH7.0) with 177Lu chloride as previously
described [23]. 177Lu-DOTA-J591 mAb (177Lu-J591) was purified by gel
filtration and sterilized by membrane (0.2 μm) filtration prior to administration
into patients. Labelling efficiency and radiochemical purity were determined
using ITLC SG and 5mM DTPA solution as solvent. Immunoreactivity of
177
Lu-J591 was determined by the Lindmo method using PSMA positive
LNCaP tumour cells [24, 25]. Immunoreactivity was well preserved, even at
0.5–0.7 GBq/mg of specific activity.
2. CLINICAL STUDIES
76
SESSION 9
2.2.1. Retreatment
77
GOLDSMITH et al.
78
SESSION 9
177
4. Lu-J591 TUMOUR TARGETING
5. HAEMATOLOGICAL TOXICITY
Anterior Posterior
FIG. 1. Tumour localization of 177Lu-J591 (on day 6) in a patient with prostate cancer.
79
GOLDSMITH et al.
370 3 3 3
555 3 2 1 3
1110 5 2 2 1 2 2 1
1665 5 1 3 1 2 1 2
2220 3 1 1 1 1 2
2590 6 1 4 1 1 2 1 2
2775 3 2 1 2 1
Most of the remaining patients at these two dose levels experienced grade 3
platelet toxicity. With the 177Lu-J591 antibody, the 2590 MBq/m2 dose level was
determined to be the MTD. Post-treatment platelet counts decline generally at
2.5–3 weeks with platelet nadirs occurring at 4–5 weeks thereafter followed by a
recovery phase. In all the three subjects, at 2775 MBq/m2, the fractional decrease
in platelets as a function of time (days) is shown in Fig. 2. The mean platelet
counts returned to 80–90% of their pre-treatment values over 2–3 weeks.
80
SESSION 9
81
GOLDSMITH et al.
177
Lu dose (r = 0.88) and the bone marrow radiation dose based on blood
radioactivity (Fig. 4 (a) and (b)).
7. ANTI-TUMOUR RESPONSE
All 35 patients in this trial had abnormal, rising PSAs and 7 patients had
measurable disease. None of the 7 patients with measurable disease had an
objective tumour response nor a ≥50% PSA decline. On the basis of PSA, 14
patients demonstrated progressive disease (PSA increase of >25%) after
82
SESSION 9
FIG. 4(a). Correlation of fractional decrease in platelets with the total treatment dose
(MBq) of the 177Lu-J591 antibody.
FIG. 4(b). Correlation of fractional decrease in platelets with bone marrow radiation
absorbed dose (cGy) following treatment with the 177Lu-J591 antibody.
83
GOLDSMITH et al.
8. DISCUSSION
84
SESSION 9
177
8.1. Lu-J591 tumour targeting
177
8.2. Lu-J591 toxicity
85
GOLDSMITH et al.
characteristics, use of the latter is compromised in this antigenic system as, after
internalization, 131I labelled antibody is rapidly dehalogenated and diffuses out
of tumour cells while radiometals such as 177Lu or 90Y remain sequestered
within the targeted tumour cells.
In this study, as in the authors’ 90Y-J591 trial, no clear relationship was
found between toxicity and a history of prior chemotherapy treatment.
Similarly, no correlation was found between toxicity and prior radiotherapy nor
between toxicity and the extent of bone marrow involvement by cancer [30].
Similar observations in prostate cancer were made by Knox et al. [33] and
O’Donnell et al. [34]. The non-haematological toxicity in RIT trials in general
and in this 177Lu-J591 trial was minimal and not dose limiting. Radiation
dosimetry calculations indicate a radiation dose to liver is approximately
1088 cGy at the MTD of 2590 MBq/m2. In addition, in this 177Lu-J591 trial, no
significant hepatotoxicity was seen in patients who received multiple doses,
including 8 patients who received cumulative doses between 3330–4440 MBq/m2.
Radiation doses to kidney and spleen were also well within acceptable limits
and no related organ toxicity was noted [31].
In patients who were treated with 177Lu-J591, a strong correlation was
observed between myelotoxicity and the amount of administered 177Lu dose or
the bone marrow absorbed radiation dose [35]. In contrast, the correlation
between myelotoxicity and marrow radiation absorbed dose or the adminis-
tered total MBq dose has been poor with 90Y-J591. The similarly weak
association between myelotoxicity and bone marrow radiation absorbed doses
based on blood has been previously reported for both 131I and 90Y labelled
mAbs. The authors believe that the demonstration of the predictability of
myelotoxicity based on bone marrow radiation dose with 177Lu, but not with
the 90Y radionuclide, may help us understand the importance of the energy of
radiation and the relative in vivo stability of the radionuclide–antibody
complex in the overall assessment of radiation dose and myelotoxicity.
177
8.3. Lu-J591 retreatment
Sixteen of the 35 patients in 177Lu trial received multiple doses. Two doses
of 1665 or 2220 mCi/m2, totalling 3330–4440 MBq/m2, proved to be quite toxic,
with 3 of 5 patients experiencing prolonged and incomplete platelet recovery.
Two or more doses of 1110 MBq/m2, however, were well tolerated and
4 patients received cumulative doses of 3330 MBq/m2, almost 30% higher than
the single dose MTD [30]. In this study, each dose was administered after
allowing for haematological recovery from the prior dose. It therefore took
3–4 months to administer the three doses, resulting in a higher cumulative dose
but lower dose rate. While there may be advantages to the higher cumulative
86
SESSION 9
dose, the time required to deliver this dose using this regimen may be more
than offset by unrelenting tumour progression. Given the kinetics of platelet
decline and recovery, a dose interval of 14–17 d may allow a two dose regimen
that might result in a higher cumulative dose than a single dose regimen to be
given over a shorter period of time than attempted in this trial. Such a schedule
would result in the onset of platelet recovery from the first dose coinciding with
platelet decline from the second dose thereby resulting in a longer but
shallower nadir than with a single MTD dose. Such a dose schedule remains to
be explored.
9. CONCLUSION
ACKNOWLEDGEMENTS
87
GOLDSMITH et al.
REFERENCES
[1] SURFACE, D., Promising isotopes - looking at lutetium-177 and other targeted
radiotherapy isotopes, Radiology Today 5 (2004) 20.
[2] LEONARD, J.P., et al., Comparative physical and pharmacologic characteristics
of iodine-131 and yttrium-90: Implications for radioimmunotherapy for patients
with non-Hodgkin’s lymphoma, Cancer Investigation 21 (2003) 241.
[3] KNAPP, F.F., et al., "Direct" production of lutetium-177 (Lu-177) from enriched
Lu-176 in a high flux reactor - the only practical route to provide high multi curie
levels of high specific activity Lu-177 required for routine clinical use. 5th Interna-
tion Conference of Isotopes, Brussels, Belgium, April 2005. Abstract book, page
11.
[4] CUTLER, C.S., Personal communication regarding specific activity of Lu-177,
2005.
[5] ISRAELI, R.S., et al., Expression of the prostate-specific membrane antigen,
Cancer Res. 54 (1994) 1807.
[6] BANDER, N.H., et al., Targeted systemic therapy of prostate cancer with a mono-
clonal antibody to prostate specific membrane antigen (PSMA), Semin. Oncol. 30
(2003) 667.
[7] BOSTWICK, D.G., et al., Prostate specific membrane antigen expression in
prostatic intra-epithelial neoplasia and adenocarcinoma: A study of 184 cases,
Cancer 82 (1998) 2256.
[8] SILVER, D.A., et al., Prostate-specific membrane antigen expression in normal
and malignant human tissues, Clin. Cancer. Res. 3 (1997) 81.
[9] WRIGHT, G.L., et al., Upregulation of prostate-specific membrane antigen after
androgen-deprivation therapy, Urology 48 (1996) 326.
[10] SWEAT, S.D., et al., PSMA expression is greatest in prostate adenocarcinoma and
lymph node metastases, Urology 52, (1998) 637.
[11] TROYER, J.K., et al., Detection and characterization of the prostate-specific
membrane antigen (PSMA) in tissue extracts and body fluids, Int. J. Cancer 62
(1995) 552.
[12] LIU, H., et al., Monoclonal antibodies to the extracellular domain of prostate
specific membrane antigen also react with tumor endothelium, Cancer Res. 57
(1997) 3629.
[13] CHANG, S.S., et al., Five different anti-prostate-specific membrane antigen
(PSMA) antibodies confirm PSMA expression in tumor-associated neovascula-
ture, Cancer Res. 59 (1999) 3192.
[14] NANUS, D., et al., Clinical use of monoclonal antibody huJ591 therapy:
Targeting prostate specific membrane ntigen, J.Urol. 170 (2003) S84.
[15] O’KEEFE, D.S., et al., Mapping, genomic organization and promotor analysis of
the human prostate-specific membrane antigen gene, Biochim. Biophys. Acta. 144
(1998) 113.
88
SESSION 9
89
GOLDSMITH et al.
90
RADIOLABELLED SOMATOSTATIN ANALOGUES
FOR RADIONUCLIDE THERAPY OF TUMOURS
Abstract
Molecular imaging and therapy are rapidly developing and will become important
topics in medicine in the 21st century. Radiolabelled peptides that bind to receptors
form an important class of radiopharmaceuticals for tumour diagnosis and therapy. The
specific receptor binding property of the peptide can be exploited by labelling with
radionuclide and using the radiolabelled peptide as a vehicle to guide the radioactivity
to tumours expressing a particular receptor. The high affinity of the peptide for the
peptide–receptor complex facilitates high uptake of the radiolabel in receptor
expressing tumours, while its relatively small size facilitates rapid clearance from blood,
resulting in low background radioactivity. The use of radiolabelled peptides is growing
rapidly due to these favourable characteristics, their low antigenicity and ease of
production. Receptor binding peptides labelled with gamma radiation emitters or
positron emitters enable non-evasive, whole body visualization. This process is referred
to as peptide receptor scintigraphy and is being used to detect, stage and plan the
therapy of receptor expressing tumours and also to follow tumours after therapy. In
addition, labelled with a therapeutic beta emitter these peptide molecules have the
potential to destroy receptor expressing tumours, an approach referred to as peptide
receptor radionuclide therapy (PRRT). To date, five somatostatin receptor subtypes
(sst1–sst5) have been identified and cloned. The diagnostic accuracy of 111In labelled
octreotide to visualize tumour lesions after intravenous injection has been determined
in a large series of patients with sst2 positive, mostly neuroendocrine tumours. Most
interesting is the successful application of somatostatin analogues in PET, after labelling
with positron emitters. The next logical step was to try to label these with therapeutic
radionuclides and to treat receptor positive tumors with peptide receptor radionuclides.
So far, 90Y and 177Lu are the most frequently used radionuclides in PRRT. The second
generation somatostatin analogue DOTA-Tyr3-octreotide can form a stable complex
with 90Y. In rats with subcutaneous, CA20948 pancreatic tumours, 90Y-DOTA-Tyr3-
octreotide, effectively controlled tumour growth. Studies to determine the therapeutic
efficacy of 90Y-DOTA-Tyr3-octreotide in cancer patients are ongoing at various institu-
tions and show most promising rates of complete plus partial remission. With the devel-
opment of new somatostatin analogues that bind with high affinity receptors on
91
DE JONG et al.
tumours, the available tools for radionuclide imaging and therapy of these tumours have
increased significantly.
1. INTRODUCTION
92
SESSION 9
2. [111In-DTPA]OCTREOTIDE
93
DE JONG et al.
94
SESSION 9
disease when entering the study. Although in both series favourable effects on
symptomatology were reported, CT assessed tumour regression was only
observed in rare cases.
Stokkel et al. [22] recently reported on a study that determined the effect
of [111In-DTPA]octreotide therapy in patients with progressive radioiodine
non-responsive thyroid cancer. Therapeutic effects were determined in relation
to [111In-DTPA]octreotide uptake by tumour localizations assessed on pre-
treatment diagnostic [111In-DTPA]octreotide scans. Eleven patients, selected
on positive pretreatment diagnostic scans, were treated with up to four fixed
doses of 7400 MBq [111In-DTPA]octreotide with an interval of 2–3 weeks
between the doses. In 44% of the patients, stable disease was achieved up to 6
months after the first treatment. These four patients had relatively low
pretreatment thyroglobulin values, representing limited metastasized disease.
It was therefore concluded that treatment with high doses of [111In-
DTPA]octreotide in differentiated thyroid cancer can result in stable disease in
a subgroup of patients, whereas low pretreatment thyroglobulin value, repre-
senting a small tumour load, might be a selection criterion for treatment.
It was concluded that 111In coupled peptides are not ideal for PRRT
because of the small particle range and therefore short tissue penetration.
Consequently, various research groups have aimed to develop somatostatin
analogues that can be linked via a chelator to a therapeutic radionuclide.
DOTA is a universal chelator capable of forming stable complexes with such
metals as 111In, 67Ga, 68Ga, 86Y and 64Cu for imaging as well as with 90Y and with
radiolanthanides such as 177Lu for receptor mediated radionuclide therapy
[23–25]. In addition, new somatostatin analogues were synthesized to improve
receptor affinity [4, 26].
After 111In, the next radionuclide investigated for PRRT was 90Y, emitting
ß particles with a high maximum energy (2.27 MeV) and a long maximum
particle range. The first somatostatin analogue radiolabelled with 90Y and
applied for PRRT in animals and patients was [90Y-DOTA,Tyr3]octreotide, in
which, in comparison with octreotide, the phenylalanine residue at position 3
has been replaced with tyrosine; this makes the compound more hydrophilic
and increases the affinity for sst2, leading to higher uptake in sst2 positive
tumours both in preclinical studies and in patients [27, 28].
The next analogue investigated in preclinical radionuclide therapy studies
was [177Lu-DOTA,Tyr3]octreotate. This somatostatin analogue has a very high
affinity for sst2 [4] and after radiolabelling with 177Lu high tumour uptake was
95
DE JONG et al.
seen, even very small tumours could be visualized (Fig. 1) and very good anti-
tumour effects were found in therapeutic studies in animal models [29–31].
Lutetium-177 emits gamma radiation with a suitable energy for imaging and
therapeutic β particles with low to medium energy (maximum 0.50 MeV), so
the same compound can be used for imaging and dosimetry and radionuclide
therapy, thus obviating the need for a pretherapeutic dosimetric study. The
approximate range of the β particles is 20 cell diameters, whereas the range of
those emitted by 90Y is 150 cell diameters. Less ‘cross-fire’ induced radiation
damage in the radiosensitive renal glomeruli (see below) can therefore be
expected with 177Lu. Also, in comparison with 90Y, a higher percentage of the
177
Lu radiation energy will be absorbed in very small tumours and
(micro)metastases [24, 29, 32].
96
SESSION 9
radiolabelled peptides in the blood plasma are filtered through the glomerular
capillaries in the kidneys and subsequently partly reabsorbed by, and retained
in, the proximal tubular cells, thereby reducing the scintigraphic sensitivity for
detection of small tumours in the perirenal region and the possibilities for
radionuclide therapy. It was shown that the renal uptake of radiolabelled
octreotide in rats could be reduced by positively charged amino acids, such as
lysine and arginine. About a 50% reduction could be obtained by single
intravenous administration of 400 mg/kg of L- or D-lysine [33, 34]. Therefore,
during PRRT, an infusion containing the positively charged amino acids
L-lysine and L-arginine can be given during and after the infusion of the
radiopharmaceutical, in order to reduce the kidney uptake. Various protocols
have been described, resulting in up to a 55% reduction in renal uptake of
radioactivity, thereby allowing a higher administered dose [35–38]. The
authors’ preferred protocol comprises a combination of lysine and arginine.
Patients receive a 1 L infusion (500 mL of L-lysine HCl 5% plus 250 mL of
L-arginine HCl 10% plus 250 mL saline, brought to pH7.4). This infusion lasts
4 h; it starts 30 min prior to the radiopeptide injection and a constant infusion
rate is used throughout the infusion period [35].
97
DE JONG et al.
and two required haemodialysis. Two of the five patients exhibited anaemia
(both grade 3) and thrombocytopenia (grades 2 and 4, respectively). Twenty of
the 29 patients had disease stabilization, two a partial remission, four a
reduction of tumour mass of <50% and three a progression of tumour growth.
Waldherr et al. reported several phase 2 studies in patients with neuro-
endocrine tumours [42–44]. The patients received four or more single doses of
[90Y-DOTA,Tyr3]octreotide with ascending activity at intervals of approxi-
mately 6 weeks. Observed renal or haematological toxicity was ≤grade 2
according to the National Cancer Institute grading criteria. The cumulative
dose was ≤7.4 GBq/m2. Complete and partial responses obtained in different
studies amounted to 24%. In addition, distinct protocols were compared: in the
above mentioned studies, patients received four injections of 1.85 GBq/m2
(at intervals of around 6 weeks), while in another study two injections of
3.7 GBq/m2 were administered at an interval of 8 weeks. Interestingly, the
results from the last study were the most impressive. A higher percentage of
complete responses plus partial remissions (24% after four injections versus
33% after two injections) was found, while side effects were not significantly
different [44]. It should be emphasized, however, that this was not a
randomized trial comparing two dosing schemes.
Forrer et al. recently described the results of a study with [90Y-
DOTA,Tyr3]octreotide in which 116 patients with metastatic neuroendocrine
tumours were included [45]. All patients were pre-therapeutically staged with
morphological imaging procedures and with somatostatin receptor scintig-
raphy. The scintigraphy was positive in all cases. The patients were treated with
6–7.4 GBq/m2 body surface. In this study, complete remissions were found in
4%, partial remissions in 23%, stabilization in 62% and progressive disease in
11%. A significant reduction of symptoms was found in 83%. No serious
adverse event occurred and the toxicity was acceptable [45].
Chinol et al. from the European Institute of Oncology (Milan, Italy)
described dosimetric and dose finding studies with [90Y-DOTA,Tyr3]octreotide
with and without the administration of kidney protecting agents [46]. No major
acute reactions were observed up to an administered dose of 5.6 GBq per cycle.
Reversible grade 3 haematological toxicity was found in patients injected with
5.2 GBq, which was defined as the maximum tolerated dose per cycle. None of
the patients developed acute or delayed kidney nephropathy, although follow-
up was short. Partial and complete remissions were reported by the same group
in 28% of 87 patients with neuroendocrine tumours [47]. In more detailed
publications from the same group, Bodei et al. [36, 38] report the results of a
phase 1 study of 40 patients with somatostatin receptor positive tumours, of
whom 21 had GEP tumours. Cumulative total treatment doses were in the
range 5.9–11.1 GBq, given in two treatment cycles. Six of 21 (29%) patients had
98
SESSION 9
tumour regression. Median duration of the response was 9 months. Bodei et al.
also recently evaluated the efficacy of [90Y-DOTA,Tyr3]octreotide therapy in
metastatic MTC patients with a positive [111In-DTPA]octreotide scintigram,
progressing after conventional treatments. Twenty-one patients were retro-
spectively evaluated after therapy, receiving 7.5–19.2 GBq in 2–8 cycles. Two
patients (10%) obtained a CR, while stable disease was observed in 12 patients
(57%); 7 patients (33%) did not respond to therapy. The duration of the
response was in the range 3–40 months [38].
Another study with [90Y-DOTA,Tyr3]octreotide (OctreoTher, 90Y-SMT-
487) was the phase 1 Novartis study performed in Rotterdam, Brussels and
Tampa, which aimed to define the maximum tolerated single and four cycle
doses of [90Y-DOTA,Tyr3]octreotide and in which patients received escalating
doses up to 14.8 GBq/m2 in 4 cycles or up to a 9.3 GBq/m2 single dose, without
reaching the maximum tolerated single dose [18, 48, 49]. The cumulative
radiation dose to kidneys was limited to 27 Gy. All patients received amino
acids concomitant with [90Y-DOTA,Tyr3]octreotide for kidney protection.
Three patients had dose limiting toxicity: 1 liver toxicity, 1 thrombocytopenia
grade 4 (<25 × 109/L) and 1 myelodysplastic syndrome. Four out of 54 (7%)
patients who had received their maximum allowed dose had partial remission
and 7 (13%) minor remission. The median time to progression in the
44 patients who had either stable disease, minor remission or partial remission
was 30 months. An important observation in this study was a clear dose–
response relation; the percentage reduction in tumour volume increased with
increasing tumour radiation dose (up to about 600 Gy) [50]. Prior chemo-
therapy was predisposed to haematological toxicity. Renal toxicity was mild in
these patients, with individualized dosimetry and amino acid infusion for
kidney protection. Despite the differences in the protocols used, the rate of
complete plus partial responses seen in the various aforementioned [90Y-
DOTA,Tyr3]octreotide studies consistently exceeds that obtained with [111In-
DTPA]octreotide (see above).
99
DE JONG et al.
range of 177Lu when compared with 90Y, which may be especially important for
small tumours [24].
The first treatment effects of [177Lu-DOTA,Tyr3]octreotate therapy were
described in 35 patients with neuroendocrine GEP tumours, who had a follow-
up of 3–6 months after receiving their final dose [51]. Patients were treated with
dosages of 3.7, 5.6, or 7.4 GBq [177Lu-DOTA,Tyr3]octreotate, up to a final
cumulative dose of 22.2–29.6 GBq, with treatment intervals of 6–9 weeks. The
effects of the therapy on tumour size were evaluable in 34 patients. Three
months after the final administration a complete remission was found in one
patient (3%), partial remission in 12 (35%), stable disease in 14 (41%) and
progressive disease in 7 (21%), including 3 patients who died during the
treatment period. The side effects of treatment with [177Lu-DOTA,Tyr3]octre-
otate were few and mostly transient, with mild bone marrow depression the
most common finding. In a more recent update of this treatment in 76 patients
with GEP tumours [52], complete remission was found in 1 patient (1%),
partial remission in 22 (29%), minor remission in 9 (12%), stable disease in
30 (39%), and PD in 14 patients (18%). Six out of 32 patients who had initially
stable disease or tumour regression after the therapy and who were also
evaluated after 12 months (mean 18 months from therapy start) became
progressive; in the other 26 the tumour response was unchanged. Median time
to progression was not reached at 25 months from start of therapy. Serious side
effects in the whole group of patients who had been treated or were being
treated up to that moment consisted of myelodysplastic syndrome in a patient
who had had chemotherapy with alkylating agents two years before entering
the study and renal insufficiency in another patient who had had unexplained
rises in serum creatinine concentrations in the year preceding the start of
therapy and who had a urinary creatinine clearance of 41 mL/min when
entering the study. Tumour regression was positively correlated with a high
uptake on the [111In-DTPA]octreotide scintigram, limited hepatic tumour mass
and high Karnofsky performance score.
In patients with progressive metastatic (or recurrent) differentiated
thyroid carcinoma (DTC) who do not respond to radioiodine therapy or do not
show uptake on radioiodine scintigraphy, treatment options are few. As these
tumours may express somatostatin receptors, PRRT using somatostatin
analogues might be effective and therefore the authors evaluated the
therapeutic efficacy of [177Lu-DOTA,Tyr3]octreotate in patients with DTC. In
addition, the uptake of radioactivity in the tumours was studied in relation to
treatment outcome. Five patients with DTC were treated with 22.4–30.1 GBq
of [177Lu-DOTA,Tyr3]octreotate. Three patients had Hürthle cell thyroid
carcinoma, one patient had papillary thyroid carcinoma and one had follicular
thyroid carcinoma. The uptake on [177Lu-DOTA,Tyr3]octreotate scintigraphy
100
SESSION 9
3.5. [90Y-DOTA]lanreotide
101
DE JONG et al.
that 111In labelled [DOTA]lanreotide bound with high affinity to hsst2, hsst3,
hsst4 and hsst5 and with lower affinity to hsst1 expressed on COS7 cells, making
it a universal receptor binder [58]. However, Reubi et al. found in vitro in cell
lines transfected with the different somatostatin receptor subtypes and that
whereas [90Y-DOTA]lanreotide had a good affinity for the sst5, it had a low
affinity for sst3 (IC50 290nM) and sst4 (IC50 > 10 000nM) [4]. Froidevaux et al.
[59] concluded from their comparison study of, among other things,
[DOTA,Tyr3]octreotide and [DOTA]lanreotide in rats that radiolabelled
[DOTA,Tyr3]octreotide has more potential for clinical application than
[DOTA]lanreotide.
Lanreotide labelled with 90Y was the second analogue used for clinical
PRRT studies at different centres in the MAURITIUS trial [56]. In this study,
cumulative treatment doses of up to 8.58 GBq [90Y-DOTA]lanreotide were
given as a short term intravenous infusion. Treatment results in 154 patients
indicated minor responses in 14%. No severe, acute or chronic haematological
toxicity or changes in renal or liver function parameters due to [90Y-
DOTA]lanreotide were reported. In two-thirds of patients with neuroendo-
crine tumour lesions, [90Y-DOTA,Tyr3]octreotide showed a higher tumour
uptake than [90Y-DOTA]lanreotide, which can be explained by the lower
affinity of [90Y-DOTA]lanreotide for sst2.
102
SESSION 9
kidney function and haematological parameters for study entry and for each
new administration. Lastly, the patients have to meet a minimum performance
score as they are isolated in a nuclear medicine ward for a variable time,
depending on national laws on radiation protection.
5. CONCLUSION
103
DE JONG et al.
REFERENCES
[1] PATEL, Y.C., et al., The somatostatin receptor family. Life Sci, 1995. 57(13): p.
1249-65.
[2] PATEL, Y.C., Somatostatin and its receptor family. Front Neuroendocrinol, 1999.
20(3): p. 157-98.
[3] SCHONBRUNN, A., Somatostatin receptors present knowledge and future
directions. Ann Oncol, 1999. 10(Suppl 2): p. S17-21.
[4] REUBI, J.C., et al., Affinity profiles for human somatostatin receptor subtypes
SST1-SST5 of somatostatin radiotracers selected for scintigraphic and radiothera-
peutic use. Eur J Nucl Med, 2000. 27(3): p. 273-82.
[5] KVOLS, L.K., et al., Treatment of the malignant carcinoid syndrome. Evaluation
of a long- acting somatostatin analogue. N Engl J Med, 1986. 315(11): p. 663-6.
[6] ERIKSSON, B., OBERG, K., Summing up 15 years of somatostatin analog
therapy in neuroendocrine tumors: future outlook. Ann Oncol, 1999. 10(Suppl 2):
p. S31-8.
[7] LAMBERTS, S.W., KRENNING, E.P., REUBI, J.C., The role of somatostatin
and its analogs in the diagnosis and treatment of tumors. Endocr Rev, 1991. 12(4):
p. 450-82.
[8] LAMBERTS, S.W., REUBI, J.C., KRENNING, E.P., Somatostatin analogs in the
treatment of acromegaly. Endocrinol Metab Clin North Am, 1992. 21(3): p. 737-
52.
[9] JANSON, E.T., OBERG, K., Long-term management of the carcinoid syndrome.
Treatment with octreotide alone and in combination with alpha-interferon. Acta
Oncol, 1993. 32(2): p. 225-9.
[10] ANDERSSON, P., et al., Internalization of indium-111 into human neuroendo-
crine tumor cells after incubation with indium-111-DTPA-D-Phe1-octreotide. J
Nucl Med, 1996. 37(12): p. 2002-6.
[11] DE JONG, M., et al., Internalization of radiolabelled [DTPA0]octreotide and
[DOTA0,Tyr3]octreotide: peptides for somatostatin receptor-targeted scintig-
raphy and radionuclide therapy. Nucl Med Commun, 1998. 19(3): p. 283-8.
[12] HOFLAND, L.J., VAN KOETSVELD, P.M., WAAIJERS, M., LAMBERTS, S.W.,
Internalisation of isotope-coupled somatostatin analogues. Digestion, 1996.
57(Suppl 1): p. 2-6.
[13] DUNCAN, J.R., STEPHENSON, M.T., WU, H.P., ANDERSON, C.J., Indium-
111-diethylenetriaminepentaacetic acid-octreotide is delivered in vivo to pancre-
atic, tumor cell, renal, and hepatocyte lysosomes. Cancer Res, 1997. 57(4): p. 659-
71.
[14] HORNIK, C.A., et al., Progressive nuclear translocation of somatostatin analogs.
J Nucl Med, 2000. 41(7): p. 1256-63.
[15] WANG, M., et al., Subcellular localization of radiolabeled somatostatin
analogues: implications for targeted radiotherapy of cancer. Cancer Res, 2003.
63(20): p. 6864-9.
104
SESSION 9
105
DE JONG et al.
[31] SCHMITT, A., et al., Radiation therapy of small cell lung cancer with 177Lu-
DOTA-Tyr3-octreotate in an animal model. J Nucl Med, 2004. 45(9): p. 1542-8.
[32] DE JONG, M., et al., Tumor Response After [(90)Y-DOTA(0),Tyr(3)]Octreotide
Radionuclide Therapy in a Transplantable Rat Tumor Model Is Dependent on
Tumor Size. J Nucl Med, 2001. 42(12): p. 1841-6.
[33] DE JONG, M., et al., Inhibition of renal uptake of indium-111-DTPA-octreotide
in vivo. J Nucl Med, 1996. 37(8): p. 1388-92.
[34] BERNARD, B.F., et al., D-lysine reduction of indium-111 octreotide and yttrium-
90 octreotide renal uptake. J Nucl Med, 1997. 38(12): p. 1929-33.
[35] ROLLEMAN, E.J., VALKEMA, R., DE JONG, M., KOOIJ, P.P., KRENNING, E.P.,
Safe and effective inhibition of renal uptake of radiolabelled octreotide by a
combination of lysine and arginine. Eur J Nucl Med Mol Imaging, 2003. 30(1):
p. 9-15.
[36] BODEI, L., et al., Receptor-mediated radionuclide therapy with 90Y-DOTATOC
in association with amino acid infusion: a phase I study. Eur J Nucl Med Mol
Imaging, 2003. 30(2): p. 207-16.
[37] KWEKKEBOOM, D.J., et al., [177Lu-DOTA0,Tyr3]octreotate: comparison with
[111In-DTPA0]octreotide in patients. Eur J Nucl Med, 2001. 28(9): p. 1319-1325.
[38] BODEI, L., et al., Receptor radionuclide therapy with 90Y-[DOTA]0-Tyr3-octre-
otide (90Y-DOTATOC) in neuroendocrine tumours. Eur J Nucl Med Mol
Imaging, 2004. 31(7): p. 1038-46.
[39] OTTE, A., et al., Yttrium-90 DOTATOC: first clinical results. Eur J Nucl Med,
1999. 26(11): p. 1439-47.
[40] OTTE, A., HERRMANN, R., MAECKE, H.R., MUELLER-BRAND, J.,
[Yttrium 90 DOTATOC: a new somatostatin analog for cancer therapy of
neuroendocrine tumors]. Schweiz Rundsch Med Prax, 1999. 88(31-32): p. 1263-8.
[41] OTTE, A., et al., Yttrium-90-labelled somatostatin-analogue for cancer treatment
[letter]. Lancet, 1998. 351(9100): p. 417-8.
[42] WALDHERR, C., PLESS, M., MAECKE, H.R., HALDEMANN, A.,
MUELLER-BRAND, J., The clinical value of [90Y-DOTA]-D-Phe1-Tyr3-octre-
otide (90Y-DOTATOC) in the treatment of neuroendocrine tumours: a clinical
phase II study. Ann Oncol, 2001. 12(7): p. 941-5.
[43] WALDHERR, C., et al., Tumor response and clinical benefit in neuroendocrine
tumors after 7.4 GBq (90)Y-DOTATOC. J Nucl Med, 2002. 43(5): p. 610-6.
[44] WALDHERR, C., et al., Does tumor response depend on the number of
treatment sessions at constant injected dose using 90Yttrium-DOTATOC in
neuroendocrine tumors? Eur J Nucl Med, 2002. 29: p. S100.
[45] FORRER, F., WALDHERR, C., MAECKE, H.R., MUELLER-BRAND, J.,
Targeted radionuclide therapy with 90Y-DOTATOC in patients with neuroendo-
crine tumors. Anticancer Res, 2006. 26(1B): p. 703-7.
[46] CHINOL, M., BODEI, L., CREMONESI, M., PAGANELLI, G., Receptor-
mediated radiotherapy with Y-DOTA-DPhe-Tyr-octreotide: the experience of the
European Institute of Oncology Group. Semin Nucl Med, 2002. 32(2): p. 141-7.
106
SESSION 9
107
.
IODINE WHOLE BODY SCAN, THYROGLOBULIN
LEVELS, 99mTc MIBI SCAN AND COMPUTED
TOMOGRAPHY
Results in patients with lung metastasis from differentiated
thyroid cancer
Abstract
Correlation of the 131I whole body scan (131I WBS), 99mTc-sestamibi (Tc-MIBI)
scans, computed tomography (CT) and the value of routine follow-up for 131I WBS and
thyroglobulin levels were assessed in patients with differentiated thyroid cancer (DTC)
lung metastasis. Pulmonary metastasis was detected with 131I WBS, increased thyroglob-
ulin levels and/or other positive radiological findings in 32 patients out of 583 with DTC.
Iodine-131 WBS, thyroglobulin level assessment and/or CT were performed in the
diagnosis and follow-up of the patients with lung metastasis. A Tc-MIBI scan was
performed on 19 randomly chosen patients. Nineteen out of 32 patients had lung meta-
stasis before they received the first 131I treatment. Pulmonary metastasis was observed
in the first 131I WBS of all the patients except one; whereas no pulmonary metastasis was
detected in CT in 3/32. The final 131I WBS became normal in 13/32. Thyroglobulin levels
diminished in 21/32 and were elevated in 3/32. Iodine-131 WBS continued to be
abnormal in 2 out of 3 patients with increased thyroglobulin levels but became normal in
1 patient whose CT still demonstrated macronodulary lesions. Thyroglobulin levels did
not change significantly in 8/32. Iodine-131 WBS became normal in 5/8 and 4/5 showed
micronodules in their CT scans. Metastasis was detected in 12/19 patients who had
Tc-MIBI scans, 18/19 showed metastasis in 131I WBS and 17/19 in CT. Of the 7 patients
without the sign of metastasis in Tc-MIBI scintigraphy, 1 was negative in terms of meta-
stasis in 131I WBS and 1 in CT. Fibrosis was observed in 2/32 patients in CT. One patient
developed dedifferentiation decided by negative 131I WBS and positive CT. It was
concluded that the 131I WBS and thyroglobulin levels are the most important parameters
in the evaluation of lung metastasis in DTC. CT is an additional effect to 131I WBS and
thyroglobulin level, on the other hand, MIBI imaging alone may not be enough to detect
these metastases.
109
KÜÇÜK et al.
1. INTRODUCTION
2.1. Patients
110
SESSION 9
2.2. Surgery
Characteristic n = 32 %
Gender
Male 17 53.12
Female 15 46.87
None 1 3.12
Total or near total thyroidectomy 31 96.87
Lymph node dissection 12 37.50
Histology
Papillary 15 46.87
Follicular 13 40.62
Mix type 4 12.50
Yes 19 59.37
No 13 40.62
111
KÜÇÜK et al.
The patients with DTC who had undergone total thyroidectomy received
a fixed dose of 131I for the ablation of thyroid remnants. Post-treatment WBS
(PTWBS) was performed six days after the ablation therapy. Periodical
thyroglobulin measurement and DWBS were used for the follow-up of patients
after successful ablation. Tellurium-201 or Tc-MIBI is used for diagnosis and
follow-up in the case of negative DWBS and elevated thyroglobulin levels. The
authors could not undertake F-18 FDG scanning of the patients owing to the
absence of a PET camera in the department. The algorithm of treatment and
follow-up used is given in Fig. 1.
131
2.4. I therapy and PTWBS
The patients were given a high fixed ablative dose of 131I. L-thyroxin
replacement therapy withdrawal and low iodine diet protocols were applied for
4 weeks before the 131I therapy. TSH levels were increased at least 30 ng/mL.
A total of 100–1450 mCi (3.7–53.65 GBq) 131I was given to each patient. In the
presence of persistent functioning lung metastasis, radioiodine therapy was
repeated periodically (at least 6 months after the therapy).
PTWBS planar and spot images were obtained in anterior and posterior
projections. It was performed on the sixth post-treatment day using a large field
of view gamma camera equipped with a high energy (peak energy centred on
360 keV with a 20% energy window) parallel hole collimator (GE 4000iXC-T/
STARCAM, GE Medical Systems, Milwaukee, United States of America).
PTWBS was used only for the purpose of verification of the metastatic disease.
All the patients were researched about positive uptake on the PTWBS. These
patients with increased pulmonary activity were referred to the Department of
Thoracic Diseases and evaluated by bronchoscopy in order to confirm the
metastasis.
112
SESSION 9
113
KÜÇÜK et al.
energy collimator. Patients with normal DWBS were evaluated with periodic
follow-up, at first once a year and then every two or three years within a ten
year period. However, thyroglobulin levels of all the patients were obtained
every sixth month. The thyroglobulin levels were measured by the immuno-
radiometric method over the period 1985–2000 and by the chemiluminescence
method after 2000. Suppressive hormonal therapy was resumed after each 131I
WBS. All the patients were evaluated regarding positive pulmonary uptake on
the DWBS.
The Tc-MIBI WBS was carried out on 19 patients who were randomly
chosen. The authors have used Tc-MIBI in since 1990 and it took a long time to
start using it routinely for the patient group. Therefore, not all patients in the
group have had Tc-MIBI scans. The authors performed the 201Tl WBS on some
patients but did not include them in this study because their number was small.
A Tc-MIBI WBS was performed on the patients during suppressive hormonal
therapy. Tc-MIBI (15–20 mCi (555–740 MBq), CARDIO-SPECT, Medi-
Radiopharma Ltd, Hungary) was injected intravenously. Images were obtained
20–30 min later. MIBI WBS planar and spot images were obtained in anterior
and posterior projections with a large field of view gamma camera equipped
with a low energy (peak energy centred on 140 keV with a 15% energy
window), high resolution collimator (Siemens ECAM Dual Head Variable
Systems, Siemens Medical Solutions, Illinois, USA). The patients were
examined for the similar positive pulmonary uptake regions in Tc-MIBI and
131
I WBS.
2.7. CT
114
SESSION 9
3. RESULTS
Nineteen out of 32 patients had lung metastasis before the first 131I
treatment. Thirteen out of 32 patients exhibited lung metastasis during the
follow-up period. All patients were given radioiodine therapy. Thyroid
remnant ablation was successful in the first DWBS in 22/32 patients. Six out of
32 patients had other distant organ metastases than the lungs. Four of the 6
patients only had lung and bone metastases. Pulmonary metastasis was
observed in the 131I WBS of all the patients except one (31/32, 96.8%), whereas
CT showed pulmonary metastasis to be absent in 3/32 patients.
The last DWBS was negative in 13/32 patients (40.6%). DWBS was
positive in 19/32 patients. In their initial examination, 27/32 patients had
thyroglobulin levels higher than 30 ng/mL (84.4%) and 5/32 patients had
thyroglobulin lower than 30 ng/mL (15.6%). In their final examination,
20/32 patients had thyroglobulin levels higher than 30 ng/mL (62.5%) and
12/32 patients had thyroglobulin levels lower than 30 ng/mL. The final
thyroglobulin levels began to change in 7/32 patients. In 8/32 patients the
thyroglobulin level was lower than 30 ng/mL plus a negative 131I WBS.
However, one patient had thyroglobulin values higher than 30 ng/mL who
had been negative for 131I WBS. Four out of 32 patients had thyroglobulin
levels less than 30 ng/mL (15.6%) and positive 131I WBS. Thirteen out of 32
patients had negative 131I WBS after a final diagnostic 131I WBS.
Thyroglobulin levels were lower in 21/32 and elevated in 3/32 patients.
The 131I WBS continued to be abnormal in 2 out of 3 patients with increased
thyroglobulin levels but became normal in one patient whose CT still demon-
strated macronodular lesions. Thyroglobulin levels did not change signifi-
cantly in 8/32. Iodine-131 WBS became normal in 5/8 and 4/5 showed
micronodules in their CTs. Fibrosis was observed in 2/32 patients in CT. One
patient developed dedifferentiation with negative 131I WBS and positive CT.
Metastasis was discerned in 12/19 patients who underwent Tc-MIBI
WBS, 18/19 showed metastasis in 131I WBS and 17/19 in CT (Table 2). Of the
Positive 18 12 17
Negative 1 7 2
% 94.7 63.1 89.4
115
KÜÇÜK et al.
4. DISCUSSION
Lungs are the most frequent distant localization of metastasis due to DTC
(1–4%) [4]. The 131I WBS and thyroglobulin levels play an important diagnostic
role in the evaluation of lung metastasis in DTC [9, 10]. Lung metastases were
observed in the 131I WBS of all the patients except one, who had a very high
thyroglobulin level and a negative 131I WBS.
Serum thyroglobulin level measurement is the most sensitive and specific
marker of DTC patients, but increased thyroglobulin concentration alone is not
enough when there is a large thyroid remnant. The thyroglobulin levels used in
the retrospective study were measured by an immunoradiometric method over
the period 1985–2000 and by chemiluminescence after 2000. The cut-off values
were 30 ng/mL in the first period and 5 ng/mL in the second period and the
values above these were considered for the assessment of metastatic disease.
Although it is reported that the chemiluminescence method is more sensitive
compared to the immunoradiometric method [19], a cut-off value of 30 ng/mL
was selected in this retrospective study because both methods were used over
the long duration of the analysis period.
Initial thyroglobulin concentrations determined after total thyroidectomy
or near total thyroidectomy with lung metastasis in 32 patients off thyroxin and
before 131I therapy or scanning showed that 15.6% (5/32) of patients had initial
levels less than 30 ng/mL and, in a contrary manner, 84.4% (27/32) had initial
levels higher than 30 ng/mL. Filesi et al. [11] reported that in their series of
patients, 66.7% of those with metastases had their initial thyroglobulin levels
higher than 60 ng/mL. David et al. [7] reported that 46% of patients with
thyroid remnants or metastases had their initial serum thyroglobulin values
higher than 30 ng/mL. In the authors’ group, 84.4% of patients with lung
metastases had their initial thyroglobulin levels higher than 30 ng/mL. In
addition, Filesi et al. reported metastases were observed in the initial 131I WBS
in 47.8% of patients with thyroglobulin values of less than 60 ng/mL; 61.3% of
patients with the initial 131I WBS negative for metastases had thyroglobulin
levels greater than 60 ng/mL. In contrast to the finding, David et al. determined
116
SESSION 9
only 2.7% of scan negative subjects had initial serum thyroglobulin levels
higher than 30 ng/mL. The authors’ retrospective findings suggested that 3.1%
of patients had both 131I WBS negative and thyroglobulin levels higher than
30 ng/mL. It might be associated with thyroid remnants and/or other distant
metastases. Thyroid remnant ablation was successful in the first DWBS in 22/32
patients and 6/32 patients had other distant organ metastases out of lung in the
study. In addition, the study showed that 16.5% of patients who had initial 131I
scan positive for metastases had thyroglobulin levels of less than 30 ng/mL. The
authors consider that the thyroglobulin cut-off level must be reduced to below
30 ng/mL. David et al. reported that with 131I ablation and long term thyroxin
suppression, the level of thyroglobulin tended to fall. In the study, final
thyroglobulin concentrations were determined in order to measure initial
thyroglobulin concentrations under the same conditions. The authors observed
that 37.5% (12/32) of patients had final thyroglobulin levels of less than 30 ng/mL
and, conversely, 62.5% (20/32) had final thyroglobulin levels higher than 30 ng/mL.
Initial thyroglobulin levels varied from 0.5 to 13 454 (average: 882.6) ng/mL.
Final thyroglobulin levels were measured from 0.5 to 4795 (average: 519).
Consequently, declining thyroglobulin levels were observed in 65.6% (21/32) of
the patients. Furthermore, final thyroglobulin values had less than 30 ng/mL in
37.5% (12/32) of the patients (Fig. 2).
The combination of routine thyroglobulin measurement and 131I WBS
can be used as a gold standard in the diagnosis and follow-up of lung metastasis
in DTC patients without any determined distant organ metastasis and after
successful thyroid remnant ablation. Increased activity in the lung region in the
FIG. 2. Thyroglobulin levels are presented in 32 patients with lung metastases from DTC.
117
KÜÇÜK et al.
118
SESSION 9
Dadparvar et al. [13] found a poor sensitivity (36%) but a high specificity
(89%) for Tc-MIBI WBS compared with 131I WBS. Miyamoto et al. reported
that Tc-MIBI WBS did not discern pulmonary metastases in more patients
than the 131I scan (75% and 85%, respectively). The authors’ findings suggested
that, compared with 131I WBS, Tc-MIBI WBS was less sensitive (94.2% and
63.2%, respectively) in detecting lung metastasis with DTC patients.
It has been reported that the sensitivity of thoracic CT is about 80% [14].
To the contrary, some studies have shown that a CT scan can detect 3 mm
peripheral and 6 mm central nodules, although it still fails to discern the diffuse
interstitial type of lung metastases in patients with DTC [15]. The authors
found that CT detected lung metastases in 29/32 patients (90.6%) in their
series. Nevertheless, their findings suggest that, compared with 131I WBS, CT
was less sensitive in detecting lung metastasis with DTC patients. It may be
difficult to interpret data in an area of the neck already submitted to surgery.
However, the authors also found that fibrosis was observed in 2/32 patients in
CT. Dedifferentiation was observed in one patient decided by negative 131I
WBS and positive CT, owing to the fact that CT was an additive effect to the
131
I WBS and the thyroglobulin level.
FDG PET scanning may be useful in localizing distant metastases,
especially when there is no radioiodine uptake. These metastases may be
located in the mediastinum or other distant areas [16, 17]. FDG uptake was
also detected more frequently in patients with poor DTC, in whom no
detectable 131I uptake could be demonstrated. FDG PET cannot supersede the
131
I scan. However, several investigators reported that FDG PET and 131I WBS
played complementary roles in the detection of recurrent metastatic DTC [17].
The authors were not able to use PET scanning because this modality was not
available in the department.
In their study, the authors chose to evaluate retrospective data with the
help of previous data, instead of statistically assessing various parameters
affecting the survival period. Their aim in doing so was to see if there was a
treatment mode which would result in progress in the patients. Margo et al.
analysed patient, tumour and treatment related factors and their relation to
disease specific survival using statistical tests. They found that at the age of
45 years or more, a site other than lung only or bone only and symptoms at the
time of diagnosis are associated with poorer outcomes [18]. In addition, Ronga
et al. reported that a young age at diagnosis and radioiodine uptake by
metastases are the most important factors positively affecting survival time.
They found that radioiodine therapy, also with high cumulative radioiodine 131I
activity, can lead to longer survival time or complete recovery [4]. The authors
found a prolongation of the disease free period and progress in the parameters
followed in those of their patient group who showed radioactive iodine
119
KÜÇÜK et al.
accumulation and were younger than 45 years old at the first diagnosis. Six of
8/32 patients who showed progress in the parameters followed (DWBS,
thyroglobulin level less than 30 ng/mL) were younger than 45 years old and
needed fewer cumulative doses than the other two patients. The authors
believe that radioactive iodine therapy should continue to be given to the
patients because metastatic disease at least slows down the course of the
disease, although it may not appear to cure the disease in persistent or
recurrent cases.
5. CONCLUSION
For lung metastasis detection and follow-up after total thyroidectomy, the
131
I WBS and thyroglobulin levels were the most important parameters. CT has
an additive effect to 131I WBS. The authors’ findings suggested that, compared
with 131I WBS, Tc-MIBI WBS was less sensitive (94.2% and 63.2%, respec-
tively) in detecting lung metastasis with DTC patients. MIBI imaging alone
might not be enough to detect these metastases.
REFERENCES
120
SESSION 9
121
.
99m
Tc-MIBI AND 131I SCINTIGRAPHY IN THE
FOLLOW-UP OF DIFFERENTIATED THYROID
CARCINOMA (DTC) PATIENTS AFTER SURGERY
Sofia, Bulgaria
Abstract
The MIBI scan has been reported to be a highly sensitive imaging technique for
the detection of differentiated thyroid carcinoma (DTC) metastases that have lost the
capability to uptake 131I. The purpose of this study was to evaluate, retrospectively, the
value of the 99mTc-MIBI scan and 131I whole body scintigraphy using thyroglobulin (Tg)
levels as a basis for comparison. A total of 84 patients with DTC (47 cases with papillary,
18 cases with follicular and 19 cases with papillary–follicular) were assessed. All of them
had undergone total or near total thyroidectomy and received radioiodine treatment for
ablation of post-surgical residual thyroid tissue. They were examined after 4 weeks
L-thyroxin withdrawal in the follow-up of DTC. Planar and whole body images were
acquired at 15 min and 180 min after IV application of 99mTc-MIBI (555–740 MBq) and
at 48 h after post-operative administration of 131I (111–185 MBq). Serum Tg assays were
performed to clarify the presence of residual recurrent malignancy. The 131I scan was
positive in 55 patients, showing thyroid remnants in 31 cases, lymph node metastases in
24 cases, pulmonary metastases in 6 cases and bone lesions in 2 cases. In 18 patients the
131
I scan was negative, Tg was undetectable, and therefore the patients were considered
tumour free. In 11 patients the 131I scan was negative while serum Tg was increased.
These false negative results were observed predominantly in cases with less differenti-
ated metastatic cells, especially after several courses of high dose 131I therapy. The 99mTc-
MIBI scan revealed the presence of lymph node and/or lung metastases (non-func-
tioning metastases) in 9 of them; false negative results were obtained in 2 cases. Serum
Tg was increased in all patients with local lymph node and distant metastases, visualized
by 131I or by 99mTc-MIBI, but also in 18 patients with thyroid remnants only. Considering
the 131I scan as the most specific standard procedure the authors conclude that the
combined 99mTc-MIBI scintigraphy and serum Tg assay appear to be an alternative to
radioiodine diagnostic imaging to demonstrate the extent of the disease in cases with
DTC and elevated Tg.
123
SERGIEVA et al.
1. INTRODUCTION
It is widely accepted that the 131I scan is the most specific diagnostic
procedure, a ‘gold standard’ in follow-up of patients with differentiated thyroid
carcinoma (DTC) after surgery. Depending on the histological type and
tumour stage, local recurrence, lymph node metastases or distant metastases
may be present or may develop during follow-up [1]. The most reliable
parameter for tumour recurrence or metastatic disease is a raised thyroglobulin
(Tg) level. With the introduction of lipophilic cationic tracers such as 99mTc–
sestamibi (MIBI) and 99mTc-tetrofosmin and, especially, fluorine-18 fluorode-
oxyglucose positron emission tomography (FDG PET) it has become obvious
that the use of only whole body 131I scintigraphy (131I-WBS) leads to serious
underestimation of the extent of disease in patients with DTC and elevated Tg
[2–4].
The 99mTc-MIBI scan has been reported to be a highly sensitive imaging
technique for detection of DTC metastases that have lost the capability to
uptake 131I [5–7].
The purpose of the present study was to evaluate, retrospectively, the
value of the 99mTc-MIBI scan and 131I-WBS using Tg levels as a basis for
comparison.
2.1. Patients
2.2. Methods
All of the patients had undergone total or near total thyroidectomy and
received radioiodine treatment for ablation of post-surgical residual thyroid
tissue, in a total activity range of 3.3–7.0 GBq (90–190 mCi).
Routine follow-up examination was conducted 6 months post-ablation
therapy, after 4 weeks of L-thyroxin withdrawal. Serum Tg was measured. A
serum level above 4 ng/mL was considered abnormal when the patients were in
the hypothyroid state. All patients were asked to adhere to a low iodine diet
until the study had been completed.
124
SESSION 9
131
I-WBS images were acquired at 48 h after oral administration of 111–
185 MBq radioiodine on Toshiba GCA gamma camera at a speed of 7.5 cm/min
with a 1.024 × 512 matrix. Additional planar views from the neck, chest and
abdominal regions were also acquired as necessary using a HEAP collimator.
In 15 patients post-therapeutic 131I-WBS was carried out 5 d after treatment
with high dose radioiodine.
In 11 patients with a negative 131I scan and increased level of serum Tg,
additional 99mTc-MIBI scintigraphy was carried out. Planar images were
performed at 15 min and 180 min after IV application of 555–740 MBq 99mTc-
MIBI with a LEHR collimator.
All scintigraphy findings were compared to US, CT or MRT data.
3. RESULTS
(a) (b)
FIG. 1. A female patient (68 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT4pNxM0”; Tg 11.55 ng/mL. (a) 131I-WBS was
positive for a remnant in the region of operative cicatrise. (b) After a second course of
radioiodine therapy 131I-WBS showed a reduced residual thyroid tissue; Tg 2.68 ng/mL.
125
SERGIEVA et al.
(a) (b)
FIG. 2. A female patient (76 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam – pT1pN0M0. Status post-131I therapiam”; Tg 2.05 ng/mL. (a)
Planar and (b) 131I-WBS showed totally ablated remnant after radioiodine treatment.
cases cervical lymph node dissection was performed with histological confir-
mation for metastatic infiltration (Fig. 5).
Six patients had positive 131I-WBS for pulmonary metastases and 2 had
bone lesions, confirmed by CT and/or MRI (Figs 6 and 7). After therapy with a
high dose of radioiodine, a partial therapeutic response and decreased level of
serum Tg were obtained during the follow-up in 5 of them with lung metastases.
FIG. 3. (a) A female patient (33 years) with the clinical diagnosis “Ca gl.thyreoideae.
Status post-thyreoidectomiam et 131I therapiam – pT1pN1M0”; Tg 23.74 ng/mL. 131I-WBS
was positive for a remnant in the region of operative cicatrise and metastatic infiltration of
left cervical lymph nodes. (b) A female patient (23 years) with the clinical diagnosis “Ca
gl.thyreoideae. Status post-thyreoidectomiam et 131I therapiam – pT4pN1M0”; Tg 115.32
ng/mL. 131I-WBS was positive for metastatic process in the region of the left cervical
lymph nodes and upper mediastinum. (c) A female patient (22 years) with the clinical
diagnosis “Ca gl.thyreoideae. Status post-thyreoidectomiam et 131I therapiam –
pT2pN1bM0”; Tg 1548.0 ng/mL. 131I-WBS was positive for a recurrence in the region of
operative cicatrice and metastatic infiltration of left and right cervical lymph nodes.
126
SESSION 9
(a) (b)
FIG. 4. A male patient (38 years) with the clinical diagnosis “Ca gl.thyreoideae. Status post-
thyreoidectomiam et 131I therapiam – pT2pN1M0”; Tg 28.98 ng/mL. (a) 131I-WBS was
positive for a metastatic infiltration of right cervical lymph nodes. (b) After radioiodine
therapy 6 months later 131I-WBS was negative for lymph node metastases; Tg 4.05 ng/mL.
In 1 case with lung lesions and 2 with bone metastases, there was no
answer to the therapy and they showed disease progression (Fig. 8).
In 11 patients, the 131I scan was negative while the serum Tg was
increased. These false negative results were observed predominantly in cases
with lymph node and pulmonary metastases, especially after several courses of
high dose 131I therapy. The 99mTc-MIBI scan revealed the presence of lymph
node and/or lung lesions, which have lost the capability to uptake 131I in 9 of
FIG. 5. A male patient (34 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT4pN1M0”. (a) First 131I-WBS was positive
for a remnant and metastatic infiltration of left cervical lymph nodes; Tg 19.36 ng/mL. (b)
Second 131I-WBS performed 6 months later was positive for a reduced remnant and
persistence of metastatic infiltration of left cervical lymph nodes; Tg 9.23 ng/mL. (c) 131I-
WBS, performed after extirpation of metastatic cervical lymph node in the right and the
third course of radioiodine therapy, showed background radioactivity in the neck.
127
SERGIEVA et al.
FIG. 6. A male patient (50 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT4pN1M1”; Tg 1350 ng/mL. 131I-WBS was
positive for a recurrence and diffuse lung metastases.
these patients (Figs 9–11). In 2 cases with elevated Tg, the 99mTc-MIBI scan and
the 131I-WBS were negative.
Serum Tg was increased in all patients with local lymph node lesions and
distant metastases, visualized either by 131I or by 99mTc-MIBI, but also in 18
patients with thyroid remnants only (Figs 12 and 13).
(a) (b)
FIG. 7. A female patient (50 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT3pN1M1”; Tg 1341 ng/mL. 131I-WBS was
positive for multiple lung and bone metastases, visualized in anterior (a) and posterior (b)
positions.
128
SESSION 9
(a) (b)
FIG. 8. A female patient (70 years) with the clinical diagnosis “Ca gl.thyreoideae. Status post-
thyreoidectomiam et 131I therapiam – pT2pN0M0”; Tg 294 ng/mL. 131I-WBS was positive for
multiple lung metastases, visualized before application of high 131I activity (a); Tg 194 ng/mL.
There was no answer to the radioiodine treatment on the control 131I-WBS (b); Tg 203.4 ng/mL.
4. DISCUSSION
(a) (b)
FIG. 9. A female patient (54 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam – pT3pN1aM0. Status post-131I therapiam”; Tg 139.7 ng/mL. (a)
131
I-WBS showed background radioactivity in the neck. (b) 99mTc-MIBI showed an
intensive uptake of the tracer in the region of left cervical lymph nodes, significant for
metastatic infiltration.
129
SERGIEVA et al.
(d) (e)
FIG. 10. A female patient (72 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et lymphadenectomiam – pT4pN2M0. Status post-131I therapiam
et TGT”; Tg 10.19 ng/mL. (a) 131I scan showed background radioactivity in the neck
region. (b) 99mTc-MIBI showed an intensive uptake of the tracer in the region of left
cervical lymph nodes, significant for metastatic infiltration. (c) Pulmonary metastases,
visualized in the CT. (d) 131I-WBS showed a high level of background radioactivity in the
gastric and intestinal regions. (e) 99mTc-MIBI showed intensive uptake of the tracer in the
left and right lung, visualized in the anterior and posterior positions, significant for an
active proliferation.
130
SESSION 9
(a) (b)
FIG. 11. A female patient (70 years) with clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT1pN0M0. Lymph metastases colli dextra.
Status post-extirpationem”. (a) 131I-WBS showed background radioactivity in the neck.
(b) 99mTc-MIBI showed an intensive uptake of the tracer in the region of the right sub-
clavicular and cervical lymph nodes, significant for metastatic infiltration; Tg 123.2 ng/mL.
many of the lesions as possible and to ascertain whether they have the
capability ability to store iodine [3, 8, 9]. Negative 131I-WBS in patients with
elevated Tg may be explained by methodological difficulties or by the degree
of differentiation of the neoplastic cells. These false negative results were
observed predominantly in cases with less differentiated metastatic cells,
especially after several courses of high dose 131I therapy. The authors’ results
showed that 99mTc-MIBI scintigraphy has a high sensitivity in revealing
metastatic lesions that have lost the capability to uptake radioiodine. Others
reported that the 99mTc-MIBI scan has a higher sensitivity for detection of local
recurrences, lymph nodes and bone metastases, but lower sensitivity in
FIG. 12. A female patient (51 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT4apN0M0”; Tg 14.05 ng/mL. 131I-WBS was
positive for a remnant in the region of operative cicatrice.
131
SERGIEVA et al.
FIG. 13. A female patient (70 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post thyreoidectomiam et 131I therapiam – pT2pN0M0”; Tg 194 ng/mL. 131I-WBS was
positive for multiple lung and liver metastases.
revealing thyroid remnants and diffuse lung metastases as compared to the 131I-
WBS [10, 11].
By using the 99mTc-MIBI scan instead of the 131I-WBS it is not necessary
to stop thyroid replacement treatment. It is very important for patients who
cannot tolerate the withdrawal of replacement therapy.
In three cases, additional metastatic lesions in the cervical region were
visualized on the post-therapeutic 131I-WBS compared to the diagnostic
imaging obtained before radioiodine treatment. These data suggest that a
negative diagnostic 131I-WBS does not necessarily indicate a lesion lacking the
capability to accumulate iodine [3, 12]. There is also evidence that ‘blind’ high
dose administration of radioiodine has a beneficial therapeutic effect in these
cases [8, 13]. Therapeutic effect of ‘blind’ radioiodine treatment is indicated by
a significant reduction in the serum Tg level. On the other hand, this practice
has been questioned by other groups, mainly due to a lack of long term
randomized trials [14].
In conclusion, considering the 131I scan as the most specific standard
procedure, the combined 99mTc-MIBI scintigraphy and serum Tg assay appears
132
SESSION 9
FIG. 14. A male patient (72 years) with the clinical diagnosis “Ca gl.thyreoideae. Status
post-thyreoidectomiam et 131I therapiam – pT4apN0M0”; Tg 75.15 ng/mL. (a) Diagnostic
131
I-WBS was positive for a recurrence in the region of operative cicatrice, visualized on
the CT scan (b). In (c), 131I-WBS performed 5 d after administration of a high dose of
therapeutic radioiodine showed intensive uptake in the region of cervical lymph nodes
significant for metastatic process.
REFERENCES
[1] LIND, P., I-131 whole-body scintigraphy in thyroid cancer patients, Q J Nucl Med
43 (1999) 188-194.
[2] PACINI, F., et al., Diagnostic 131-I whole-body scan may be avoided in thyroid
cancer patients who have undetectable stimulated serum Tg levels after initial
treatment, J Clin Endicrinol Metab 87 (2002) 1499-1501.
[3] LIND, P., Should high hTg levels in the absence of iodine uptake be treated? Eur
J Nucl Med 30 (2003) 157-160.
[4] GROSSO, M., et al., Usefulness of 99mTc-Tetrofosmin scintigraphy and gamma-
probe detection of node metastases of thyroid cancer, Eur J Nucl Med (2000)
27(8): Abstract PS-561, p1151.
[5] MAZZAFERRI, E.L., KLOOS, R.T., Current approaches to primary therapy for
papillary and follicular thyroid cancer, J Clin Endocrinol Metab 89 (2001) 1447-
1463.
[6] MALESEVIC, M., STEFANOVIC, L.J., Value of 99mTc-MIBI scintigraphy in
the follow-up of patients confirmed thyroid carcinoma, Eur J Nucl Med (2000)
27(8): Abstract PS-560, p1150.
[7] CASARA, D., et al., Role of MIBI scan, neck us and s-Tg in clinical staging of
differentiated thyroid carcinoma (DTC) patients after surgery, Eur J Nucl Med
(2000) 27(8): Abstract PS-562, p1151.
133
SERGIEVA et al.
134
RESULTS OF KNEE RADIOSYNOVIORTHESIS IN
HAEMOPHILIC AND RHEUMATOID ARTHRITIC
PATIENTS WITH 32P COLLOID OF LOCAL
PRODUCTION
** Hospital de Clínicas,
University of Buenos Aires
Abstract
The objective of this study was to assess the effects of radioactive treatment in
knee joints with refractory synovitis in haemophilic patients, using a colloidal suspen-
sion of 32P, a pure beta emitter, developed domestically. Results were then compared
with those from chemical synovectomy. A population of rheumatoid arthritis (RA)
patients was treated and compared against intra-articular steroids and systemic drugs.
Fifty-eight male haemophilic patients, aged 4–52 years, were treated. Nine of them had
re-injections (67 procedures). Adults received 37–74 MBq; children of 2–6 years
received one third the adult’s activity; 6–10 years received one half the activity, whereas
10–16 years were injected with three quarters the activity given to adults. Anti-haemo-
philic factors (AHF) therapy, clinical examination as well as a pre-3-phase MDP scan
were registered and followed-up with MDP scans through 9 months. The intra-articular
therapies for either 32P in 44 patients or the antibiotic Rifampicin-99mTc macroaggre-
gates in 14 patients were monitored in the gamma camera with 32P bremsstrahlung
emission, searching for leakage. Twelve RA patients were studied: six received 32P and
the others intra-articular corticoids. Comparison of RoIs in treated knees during soft
tissue scintigraphies in pre- and post-third MDP control shows knee improvement. Joint
motion increased. Bleeding episodes, as well as requirements of AHF in 80% of the
radiosinovectomies, diminished. Intra-articular Rifampicin treatment requires several
135
SOROA et al.
1. INTRODUCTION
136
SESSION 9
Characteristics of 32P: T1/2 14.3 d; max. β energy 1.71 MeV; max. tissue
penetration 7.9 mm (mean penetration 2.2 mm). The authors’ local radio-
pharmacy, BACON Laboratories, supplied sterile 32P enriched gelatine
chromic phosphate colloid sized 100–200 nm, with 97% radiochemical purity
and a specific activity of 20 µCi/mg 32P [4, 6].
Radiosinovectomies were performed using 32P colloid in 58 male
haemophilic patients sent by the Hemophilic Foundation, aged 4–52 years.
Nine of them had retreatments (67 procedures), either in the same or in the
contralateral knee. The doses used were found to be safe and are based on the
results of several trials [5, 7]. Adults were injected with 37–74 MBq; children of
2–6 years of age received one third the activity of the adult; 6–10 year olds
received one half the activity of an adult, whereas 10–16 year olds were injected
with three quarters the activity given to adults. When children were overweight
by more than 20% from normal (for age and height), one quarter more activity
from the adult dose was added. Informed consent from adults or from the
children’s parents was obtained.
Patients were included in this study only if several knee episodes had
occurred. Exclusion criteria: large Bakers cysts, grades IV–V arthropaties, skin
infections of the joint area and bleeding at the time of the radiosinovectomies
[8]. Documentation of patients’ haemophilic history (severity, haemophilia A
or B), AHF therapy, number of bleedings, pain (visual analogue scale in 10
steps) range of articular movement and clinical examination were registered as
well as a pre-3-phase MDP scan in case report forms (CRF). Patients were
followed-up with the 3-phase bone scans through 1, 3, 6, 9 and 12 months. If
required, joint aspiration was carried out. The puncture sites for intra-articular
therapy for either the radiosinovectomies 32P in 44 patients or the antibiotic
Rifampicin–99mTc macroaggregates (4 MBq) in 14 patients were monitored
with the gamma camera.
Twelve RA patients were studied: six received 32P therapy and the other
six intra-articular corticoids. Clinical, blind evaluation (state of joint
involvement, pain, motility, requirements of AHF, corticoids or analgesics) was
registered in follow-up charts. For intra-articular chemical or corticoid injection
therapy, 4 MBq of 99mTc macroaggregates was added in order to obtain gamma
camera images and to blind the evaluating team of which patient received
radiotherapeutic treatment
To quantify the remission of the lesion over time, a remission rate index
(RR) was defined as the rate of severity of the lesion at the baseline and at each
control after treatment (months 3, 6, 9).
137
SOROA et al.
The severity at each control was defined as the product of the extension
of the lesion (area of the lesion expressed in pixels) and the increase in count
density at the lesion relative to a background normal area. This definition for
the relative count density was a means to establish afterwards an RR independ-
ently of the acquisition parameters and the total activity applied at each study:
where for each planar image, SI(t) is the severity index at time t, with t = 0
(baseline), or month 3, or month 6 and so on, after treatment and:
A successful treatment implies RR(t) >1 while RR(t) <1 implies disease
progression.
A clinical criterion for recovery was considered based on the fall of lesion
detectability with time for responders owing to lack of contrast, or in other
words, an increasing standard error (inversely proportional to the square root
of the count statistics at the lesion RoI). As a result, the authors accepted
clinical recovery as RR(t) values >1.10 whereas RR(t) values less than 0.90
define disease progression. Between both limits, patients were considered as
being without clinical improvement or non-responders.
138
SESSION 9
Saline flushing was carried out before the needle was withdrawn. The treated
joint was manipulated through a full range of motion to distribute the radio-
colloid throughout the joint space [1, 6, 8]. 32P bremsstrahlung emission in 201Tl
photopeak settings with a 25% window was used in the gamma camera for
early and late 24–48 h imaging to monitor extra-articular leakage [6]. After the
procedures, immobilization with a plaster and relative rest for 72 h followed
[8, 10]. Twenty-four h urine collections were obtained from 3 haemophilic
patients and counted in a β scintillation counter.
Figure 1 shows an example of a successfully treated haemophilic child
32
with P colloid injection in the left knee followed through 48 h images, to
which 99mTc markers or flood sources were added in order to provide
anatomical landmarks. Acquisition was obtained with bremsstrahlung
emission.
2.2. Follow-up
3. RESULTS
For the haemophilic patients, there were neither local burns, systemic
effects, nor leakage registered during 32P treatment (see the example in Fig. 1).
Intra-articular Rifampicin procedure required frequent injections (5–9) to
obtain a similar outcome as with 32P. Comparison of RoIs in treated knees
during soft tissue scintigraphies in pre- and post-third MDP control showed
knee improvement when the RR was >1. The follow-up evaluation demon-
strated an increase in joint motion, between 10–30%, diminished articular
volume and less requirement and frequency for the use of AHF in 80% of the
139
SOROA et al.
(a)
(b)
FIG. 1. Intra-articular 32P colloid injection in the left knee of a haemophilic patient. (a)
Upper quadrant of the gamma camera image at 4 h, to which a contour of a 99mTc marker
was added around the left leg. The lower quadrant is the 24 h registration, plus a 99mTc
flood source underneath the patient. (b) 48 h images with no leakage in the iliac fossa or
thorax.
140
SESSION 9
Figure 3 shows plots of the RR of the haemophiliac knees treated with 32P
colloidal injection against the results of the Rifampicin injection. It clearly
demonstrates that the RR exceeds 1.5 in more than 80% of the RS.
FIG. 3. Comparable RR of 32P treatment with those from chemical synovitis in haemo-
philic patients.
141
SOROA et al.
4. CONCLUSIONS
ACKNOWLEDGEMENTS
The authors wish to thank L.B. Questa and T. Bonavita providing gamma
camera images, as well as N. Moretti in charge of the kinesic therapy and the
coordination of all the haemophiliac patients received by the Hemophilic
Foundation.
We thank R. Ughetti from the Radiopharmacist Section in BACON
Laboratories S.A.I.C. for the provision of 32P β colloid. This study was
supported by the IAEA (CRP).
REFERENCES
[1] MOLHO, P., et al., A retropspective study on chemical and radioactive synovec-
tomy in severe haemophilia patients with recurrent hemarthrosis, Hemophilia 5
(1999) 115-123.
[2] SCHNEIDER, P., FARAHATI, J., REINERS, C., Radiosynovectomy in Reuma-
tology, Orthopedics, and Hemophilia, J. Nucl. Med. 46 (2005) 11 (Suppl.1) 48 S-54
S.
[3] CLUNIE, G., LUI, D., CULLUM, I., EDWARDS, J.C.W., ELL, P., Samarium-
153-particulate hydroxyapatite radiation synovectomy: Biodistribution data for
chronic knee synovitis, J. Nucl. Med. 36 (1995) 51-57.
142
SESSION 9
[4] ANGHELERI, L.J., Rapid method for obtaining colloidal suspension of phos-
phorous-32 as chromic phosphate, Proc of 2nd Intl. Conf on Peaceful uses of
Atomic Energy, UN, Geneva (1957).
[5] SOROA, V.E., Radiosynoviorthesis with P-32 colloid is still a helpful therapeutic
practice in haemophilia, J. Nucl. Med. 45 (2004) Abstract Book Supplement
abstract) 147.
[6] SOROA, V.E., et al., Effects of radiosynovectomy with P-32 colloid therapy in
haemophilia and rheumatoid ar-thritis, Cancer Biotherapy & Radiopharmaceuti-
cals 20 (2005) 3, 344-348.
[7] RIVARD, G.-E., et al., Synoviorthesis with colloidal Phosphorous-32 Chromic
Phosphate for the Treatment of Hemophilic Arthropathy, JB & JS 76-A (1994) 4,
482-488.
[8] CLUNIE, G., FISCHER, M., EANM Procedure Guidelines for Radiosynovec-
tomy. Eur. J. Nucl. Med. 30 (2003) BP12-BP16.
[9] JOHNSON, L.S., YANCH, J.C., SHORTKOFF, C.L., BARNES, A.L., SLEDGE,
C.B., Beta-particle dosimetry in radiation synovectomy, Eur. J. Nucl. Med. 22
(1995) 977-988.
[10] WINFIELD, J., CRAWLEY, J.C., HUDSON, E.A., FISHER, M., GUMPEL, J.M.,
Evaluation of two regimens to immobilise the knee after injections of 90yttrium,
British Med. J. 1(1979) 986-987.
[11] SIEGEL, H.J., LUCK, J.V., SIEGEL, M., QUINONES, C., The clinical utilization
of 32Pchromic phosphate radiosynovectomy in Hemophilia outcome analysis of
125 procedures, Proc. Orthopedic Surgeon Annual Meeting (2000) Poster PE152
(abs).
143
.
PET RADIOPHARMACEUTICALS
(Session 10)
Chairpersons
H.-J. MACHULLA
Germany
M.C. LEE
Republic of Korea
.
ALTERNATIVE METHODS OF MAKING
[11C]AMIDES: APPLICATION TO THE
PREPARATION OF 5-HT1A RECEPTOR
RADIOLIGANDS
Abstract
Many ligands for brain 5-HT1A receptors contain an amide group that is subject to
hydrolysis in vivo. In the development of radioligands for use with positron emission
tomography (PET), labelling in the carbonyl function of an amide group may be advan-
tageous for avoiding radioactive metabolites that would readily enter the brain to
confound PET receptor measurements. Several methods of labelling secondary and
tertiary amides in their carbonyl functions with 11C (T1/2 = 20.4 min) have been
developed over the past two decades or so. These methods include reaction of a
[carbonyl-11C]acid chloride, [carboxyl-11C]magnesium halide carboxylate or
[carboxyl-11C]acid with an amine or reaction of [11C]carbon monoxide with an amine
plus an aryl halide, alkyl halide or aryl triflate. Some of these processes are successfully
promoted with microwaves, palladium complexes, light or thermally intitated radicals.
These methods are surveyed here and especially exemplified from research on the
development of 5-HT1A receptor radioligands for brain imaging applications with PET.
1. INTRODUCTION
147
PIKE et al.
or other organs to produce the parent amine and carboxylic acid in the blood.
Generally, simple carboxylic acids, because they are ionized at physiological
pH7.4, do not enter the brain to a great extent, whereas the brain penetration
of amines, though subject to many factors, can be very appreciable. Hence, for
a potential PET radioligand containing an amide linkage, introduction of the 11C
label on the carbonyl side of the amide group, and even in the carbonyl entity,
may be advantageous. This is strikingly so [6, 7] for PET imaging of human
brain 5-HT1A receptors with 11C labelled WAY-100635 (1), where labelling in
the carbonyl function has been shown to provide a far more sensitive
radioligand [8] than labelling in the methoxy group [9] (Fig. 1). Labelling in the
methoxy group gives rise to a radioactive amine, 11C labelled WAY-100634 (2)
which readily enters the brain to bind both specifically and non-specifically,
whereas labelling in the carbonyl function avoids this radioactive metabolite
and instead gives rise to [11C]cyclohexanecarboxylic acid which enters the brain
only to a low and transient extent (Fig. 2). Such metabolic considerations create
a need for effective methods of labelling amides in their carbonyl functions
with cyclotron produced 11C, which is nearly always produced from the
14
N(p,α)11C reaction as either [11C]carbon dioxide or [11C]methane [10].
Various methods of labelling secondary and tertiary amides in the
carbonyl function with 11C have been developed over the last two decades or
so, and these methods are surveyed here. Although these methods have very
wide applicability, they are mainly exemplified in this survey through their
previous and ongoing applications in producing antagonist and agonist type
H
R N
O N O
N
N N R'
148
SESSION 10
[methoxy-11C]1 [carbonyl-11C]1
FIG. 2. The metabolic fate of [methoxy-11C]1 (A) and [carbonyl-11C]1 in humans (B).
PET radioligands for 5-HT1A receptors. Key considerations with respect to the
various methods to be discussed here are their isotope efficiency (i.e. radio-
chemical yields), speed and ability to deliver high specific radioactivity. If a
radioligand is to be obtained in adequately high activity and specific radio-
activity for PET investigations, then generally no more than two half-lives of
11
C (i.e. a total of 40 min) may be taken for radiosynthesis, purification and
formulation. Useful labelling methods typically comply with this time
constraint.
i ii iii
11
CO2 R11COOMgX R11COCl R11CONR'R''
149
PIKE et al.
Aubert et al. [26] have reported the rapid one pot synthesis of aliphatic
[carbonyl-11C]amides in moderate RCYs (15–60%) by direct treatment of
150
SESSION 10
i ii
11
CO2 R11COOMgX R11CONR'R''
i ii
11
CO2 Ar11COOMgX Ar11CONR'R''
151
PIKE et al.
11 i,ii iii, iv v
CO2 Ar11COOH Ar11COIm Ar11CONR'R"
152
SESSION 10
11 i
CO Ar11CONR'R"
Rahman et al. [41] have shown that aryl triflates may serve well in place
of aryl halides in the palladium mediated radiosynthesis of [carbonyl-
11
C]amides from [11C]carbon monoxide. Lithium bromide facilitates the
reactions which may be performed in the miniaturized autoclave described by
Synthia AB. A variety of [carbonyl-11C]amides was prepared from aryl triflates
and primary or secondary aliphatic amines or aniline in RCYs of 2–63% from
5 min reaction times (Fig. 8).
This method has been applied successfully to the preparation of several
candidate radioligands for the peripheral benzodiazepine receptor (PBR) in
RCYs ranging from 10 to 55% and with high specific radioactivities (200–900
GBq/μmol) [42].
11 i
CO Ar11CONR'R"
153
PIKE et al.
i
11
CO R11CONR'R''
5.2.1. Photoinitiation
i
11
CO [11C]WAY
FIG. 10. One step preparation of [11C]WAY from [11C]carbon monoxide. Conditions: (i)
WAY-100634 (2), base, c.hexyl iodide, hv, 5 min. Specific radioactivity: no carrier added.
154
SESSION 10
i ii
11
COCl2 N Cl N
11 11
C C
O O
FIG. 11. Synthesis of a [carbonyl-11 C]amide via [11C]phosgene and a [carbonyl-
11
C]carbamoyl chloride. Sample conditions: (i) 2,4-dimethoxybenzyl-tetrahydroisoquino-
line, dichloromethane, 20° C; (ii) Ph2CuMgBr.BrMgCN, THF, -30° C, 5 min then sat. aq.
NH4Cl. Specific radioactivity: no carrier added.
7. CONCLUSIONS
Several useful and alternative methods are now known for the versatile
and efficient labelling of secondary and tertiary amides in their carbonyl
functions with cyclotron produced 11C and these are finding extensive
application in the preparation of PET radioligands for 5-HT1A receptors and
other targets (e.g. opiate receptors [11, 17, 42], α1-adrenoceptors [15], σ1
receptors [42], CK receptors [42], PBR [47] and MAO [42]).
ACKNOWLEDGEMENTS
155
PIKE et al.
REFERENCES
[1] PIKE, V.W., Positron-emitting radioligands for studies in vivo — probes for
human psychopharmacology, J. Psychopharmacology 7 (1993) 139–158.
[2] HALLDIN, C., GULYAS, B., LANGER, O., FARDE, L., Brain radioligands -
state of the art and new trends, Q. J. Nucl. Med. 45 (2001) 139-152.
[3] SEDVALL, G., PET scanning as a tool in clinical psychopharmacology, Triangle
30 (1991) 11-20.
[4] FARDE, L., The advantage of using positron emission tomography in drug
research, Trends Neurosci. 19 (1996) 211-214.
[5] BURNS, H.D., et al., Positron emisison tomography neuroreceptor imaging as a
tool in drug discovery, research and development, Curr. Opinion Chem. Biol. 3
(1999) 388-394.
[6] OSMAN, S., et al., Characterization of the radioactive metabolites of the 5-HT1A
receptor radioligand, [O-methyl-11C]WAY-100635, in monkey and human plasma
by HPLC — comparison of the behaviour of an identified radioactive metabolite
with parent radioligand in monkey using PET, Nucl. Med. Biol. 23 (1996) 627–634.
[7] OSMAN, S., et al., Characterisation of the appearance of radioactive metabolites
in monkey and human plasma from the 5-HT1A receptor radioligand, [carbonyl-
11
C]WAY-100635 — explanation of high signal in PET and an aid to biomathemat-
ical modelling, Nucl. Med. Biol. 25 (1998) 215–223.
[8] PIKE, V.W., et al., Exquisite delineation of 5-HT1A receptors in human brain with
PET and [carbonyl-11C]WAY-100635, Eur. J. Pharmacol. 301 (1996) R5–R7.
[9] PIKE, V.W., et al., First delineation of 5-HT1A receptors in human brain with PET
and [11C]WAY-100635, Eur. J. Pharmacol. 283 (1995) R1–R3.
[10] QAIM, S.M., et al., PET radionuclide production, in Radiopharmaceuticals for
Positron Emission Tomography, Stöcklin G., Pike V.W. (Eds), Kluwer Academic
Publishers, the Nertherlands (1993) pp. 1-43.
[11] LUTHRA, S.K., PIKE, V.W., BRADY, F., The preparation of carbon-11 labelled
diprenorphine: a new radioligand for the study of the opiate receptor system in
vivo, J. Chem. Soc., Chem. Commun. (1985) 1423–1425.
[12] EHRIN, E., LUTHRA, S.K., CROUZEL, C., PIKE, V.W., Preparation of carbon-
11 labeled prazosin, a potent and selective α1–adrenoceptor antagonist, J. Label.
Compd. Radiopharm. 25 (1987) 177-183.
[13] PIKE, V.W., et al., Pre-clinical development of a radioligand for studies of central
5-HT1A receptors in vivo — [11C]WAY-100635, Med. Chem. Res. 5 (1995) 208–
227.
[14] SCRIPKO, J.G., HUANG, C.C., KILBOURN, M.R., Synthesis of [carbonyl-
11C]CI-99, a potent κ-opioid receptor agonist, J. Label. Compd. Radiopharm. 38
(1996) 141 (Abstract).
156
SESSION 10
[15] MCCARRON, J.A., TURTON, D.R., PIKE, V.W., POOLE, K.G., Remotely-
controlled production of the 5-HT1A receptor radioligand, [carbonyl-11C]WAY-
100635, via 11C-carboxylation of an immobilized Grignard reagent, J. Label.
Compd. Radiopharm. 38 (1996) 941–953.
[16] TRUONG, P., KRASIKOVA, R.N., HALLDIN, C., A fully automated produc-
tion of [carbonyl-11C]WAY-1000635 for clinical studies, J. Label Compd. Radiop-
harm. 46 (2003) S244 (Abstract).
[17] HWANG, D.R., SIMPSON, N.R., MONTOYA, J., MANN, J.J., LARUELLE, M.,
An improved one-pot procedure for the preparation of [11C-carbonyl]-
WAY100635. Nucl. Med. Biol. 26 (1999) 815-819.
[18] SHCHUKIN, E.V., KRASIKOVA, R.N., ANDERSSON, J., TRUONG, P.,
HALLDIN, C., A fully automated one-pot synthesis of [carbonyl-11C]WAY-
100635: validation in routine PET studies, J. Label. Compd. Radiopharm. 48
(2005) S208 (Abstract).
[19] PIKE, V.W., European concerted action on “New radiotracers for quality
assurance for nuclear medicine applications” Eur. J. Nucl. Med. 24 (1997) BP15–
BP19.
[20] PIKE, V.W., et al., [carbonyl-11C]Desmethyl-WAY-100635 (DWAY) is a potent
and selective radioligand for central 5-HT1A receptors in vitro and in vivo, Eur. J.
Nucl. Med. 25 (1998) 338–346.
[21] ANDRÉE, B., et al., The PET radioligand [carbonyl-11C]desmethyl-WAY-
1000635 binds to 5-HT1A receptors and provides a higher radioactive signal than
[carbonyl-11C]WAY-1000635 in the human brain, J. Nucl. Med. 43 (2002) 292-303.
[22] PIKE, V.W., et al., Radioligands for the study of brain 5-HT1A receptors in vivo –
development of some new analogues of WAY, Nucl. Med. Biol. (2000) 27, 429–
527.
[23] MCCARRON, J.A., et al., Two C-methyl derivatives of [11C]WAY-100635 –
effects of an amido α-methyl group on metabolism and brain 5-HT1A receptor
radioligand behavior in monkey, Mol. Imaging & Biol. 7 (2005) 209–219.
[24] MCCARRON, J.A., Development of radioligands for the study of brain 5-HT1A
and α2-adrenoceptors with PET. PhD Thesis, University of London, U.K. (1998).
[25] SHIUE, C.Y., et al., p-[18F]-MPPF: a potential radioligand for PET studies of 5-
HT1A receptors in humans, Synapse 25 (1997)147-154.
[26] AUBERT, C., HUARD-PERRIO, C., LASNE, M.-C., Rapid synthesis of
aliphatic amides by reaction of carboxylic acids, Grignard reagent and amines:
application to the preparation of [11C]amides, J. Chem. Soc., Perkin Trans. 1
(1997) 2837-2842.
[27] LU, S.Y., HONG, J.S., PIKE, V.W., Synthesis of NCA [carbonyl-11C]amides by
direct reaction of in situ generated [11C]carboxymagnesium halides with amines
under microwave-enhanced conditions, J. Label. Compd. Radiopharm. 46 (2003)
1249–1259.
[28] LU, S.Y, et al., Alternative methods for labeling the 5-HT1A receptor agonist,
1-[2_(4-fluorobenzoylamino)ethyl]-4-(7-methoxynaphthyl)piperazine (S14506),
with carbon-11 or fluorine-18, J. Label. Compd. Radiopharm. 48 (2005), 971.
157
PIKE et al.
158
SESSION 10
159
.
[18F]FLUOROETHYLATED AND [11C]METHYLATED PET
TRACERS FOR RESEARCH AND ROUTINE DIAGNOSIS
M. MITTERHAUSER, W. WADSAK
Department for Nuclear Medicine, Medical University of Vienna,
Vienna, Austria
Email: markus.mitterhauser@meduniwien.ac.at
Abstract
Development of new tracers for PET is a prerequisite for the success of this
technique as a tool for routine diagnosis. Routine PET is mainly used for oncological,
cardiological and neurological/psychiatric purposes. Two important characteristics of
the radiotracers are specificity and selectivity, requiring strong focus on research in this
field. Also necessary is a wide understanding of the mechanisms involved in the uptake
process for the improvement of the status of existing radiotracers, with consideration
given to saturation processes or whether there are enzymes or receptors involved as
targets. Radiopharmacological aspects such as metabolic stability and the behaviour of
these potential radioactive metabolites should also play a pivotal role in the design of
the new radiotracers. Additionally, the feasibility of the preparation method should be
considered for routine demands. There are few methods for the introduction of the 18F
into molecules, including electrophilic substitution, nucleophilic substitution and simple
isotopic exchange. The preparations and formulations play a pivotal role in the selection
of new radiotracers. Owing to the short half-lives of 18F (~109 min) and 11C (~20 min),
only fast and reproducible methods can be used for these radiosyntheses. Additionally,
the preparation method can significantly influence the stability of the radiotracer in
vivo. The paper addresses aspects of the preparation and application of the newly
developed radiopharmaceuticals and discusses differences and improvements, and also
drawbacks to the routine tracers. Emphasis will be put on tracers for the dopamine
transporter, for the GABA receptor, the serotonin receptor (5HT1A) and the µ-opioid
receptor. Also, new attempts for the imaging of adrenocortical pathologies ([11C]-MTO
and [18F]-FETO) will be discussed as examples for innovative oncological tracers.
1. HISTORICAL BACKGROUND
161
MITTERHAUSER and WADSAK
2. PET NUCLIDES
Positron emitters can be found throughout the entire chart of nuclides but
only few display suitable physical properties while being capable of being
produced under simple conditions. The most important PET nuclides for
clinical applications are summarized in Table 1. Fluorine-18 is the most widely
used nuclide since it combines a relatively long half-life (109.7 min) with the
possibility of production in a small, medical cyclotron.
F-18 (F-) 18
O (p,n) 18
F 109.7 min
20 18
F-18 (F2) Ne (d,α) F 109.7 min
14 11
C-11 N (p,α) C 20.4 min
16 13
N-13 O (p,α) N 10.0 min
14 15
O-15 N (d,n) O 2.0 min
64 64
Cu-64 Ni (p,n) Cu 12.7 h
86 86
Y-86 Sr (p,n) Y 14.7 h
76 76
Br-76 Se (p,n) Br 16.0 h
68 68
Ga-68 Ge/ Ga generator 67.6 min
162
SESSION 10
4. FLUORALKYLATIONS
Since many biologically active compounds contain alkylic side chains, e.g.
methyl and ethyl groups, these structural units may be targets for the affixation
of a radiolabel. In fact, many compounds have been labelled with a [11C]methyl
group for PET (vide infra). Thus, the development of [18F]fluoroalkylated
tracers was the logical consequence. A variety of different fluoroalkylating
agents have been developed so far: [18F]bromofluoromethane [5, 6], [18F]fluoro-
iodomethane [7], 2-[18F]bromofluoroethane [8, 9], 2-[18F]tosyloxyfluoroethane
[10, 11], 3-[18F]bromofluoropropane [12–14], 3-[18F]fluoroiodopropane [14, 15]
163
MITTERHAUSER and WADSAK
5. FLUOROETHYLATIONS
6.1. Methylations
164
Labelling with C-11
6.2. [18F]FE@CIT
165
MITTERHAUSER and WADSAK
166
SESSION 10
acid. After 10 min at 150°C, the product was purified using a C-18 SepPak. The
radiosynthesis evinced radiochemical yields of >90% (based on [18F]BFE), the
specific radioactivity was >416 GBq/µmol. An average 30 µA·h cyclotron
irradiation yielded more than 2.5 GBq [18F]FE@CIT (Figs 3–5).
Me Me Me
N N N O F/[18F]
COOMe a) COOH b) O
I I c) I
1 2 3
FIG. 3. A scheme for the synthesis of FE@CIT and [18F]FE@CIT. Reagents and condi-
tions: (a) 6N HCl, reflux; (b) standard synthesis: 2-fluoroethanol, DMAP, EDCI, dichloro-
methane; (c) radiosynthesis: TBAH, DMF, [18F]BFE, 150°C.
100
80
rc. yield [%]
60
40 % [18F]FE@CIT
20 % [18F]BFE
0
0 20 40 60 80 100 120 140 160 180 200
Temp. [°C]
100
rc. yield [18F]FE@CIT [%]
5mM
80
60
2mM
40
20
0 1mM
0 10 20 30
time [min]
167
MITTERHAUSER and WADSAK
5,0
Striatum
P=0.02
* *
Thalamus/Hypothalamus
4,0
P=0.03
P=0.05 P=0.002
3,0
DVR
n.s.
2,0
1,0
0,0
5min 15min 30min 60min 120min 120min
168
SESSION 10
6.3. [18F]FE@FMZ
R1 R2
FMZ Ethyl- Methyl-
FEFMZ Ethyl- 2-Fluoroethyl
FE@FMZ 2-Fluoroethyl- Methyl-
N-desmethyl FFMZ 2-Fluoroethyl- H-
Precursor H- Methyl-
169
MITTERHAUSER and WADSAK
6.4. [18F]FE@ETO
170
SESSION 10
1,80
1,40
Tissue/blood ratio
1,20
1,00
0,80
0,60
0,40
0,20
0,00
x
n
us
)
(*
te
M
lu
ra
m
Z
or
F
el
tb
FM
a
]F
C
b
al
8F
es
re
FE
Th
Ce
[1
]
8F
n
ai
[1
br
n
ai
le
br
ho
le
W
ho
W
FIG. 9. Showing tissue to blood ratios of brain regions: cerebellum, cortex, thalamus,
whole brain and rest of brain (* data taken from Ref. [55]).
0,7
Cerebellum
Cortex
Thalamus
0,6
Rest brain
Whole brain
%I.D./g
0,5
0,4
0,3
0 10 20 30 40 50 60
Time [min]
FIG. 10. Showing time–activity curves of FFMZ for various brain regions.
171
MITTERHAUSER and WADSAK
the first step, [18F]fluoride was reacted with 2-bromoethyl triflate using the
kryptofix/acetonitrile method to yield 2-bromo-[18F]fluoroethane ([18F]BFE)
(Fig. 11). In the second step, [18F]BFE was reacted with the tetrabutylam-
monium salt of (R)-1-(1-phenylethyl)-1H-imidazole-5-carboxylic acid to yield
[18F]FE@ETO, a novel inhibitor of the 11b-hydroxylase.
Male Sprague-Dawley rats were injected with 1.73–3.06 MBq of
FE@ETO into a tail vein after venodilatation in a 40°C water bath. Eighteen
rats were sacrificed by exsanguination from the abdominal aorta in deep ether
anaesthesia after 10 (n = 6), 30 (n = 6) and 60 min (n = 6) and the organs
removed, weighed and counted. For binding experiments, rat cerebellar
membranes were incubated for 90 min at 4°C in TC-50 buffer, 150mM NaCl,
and 2nM of [3H]flunitrazepam in the absence or presence of 10µM diazepam or
various concentrations of ETO, MTO and FE@ETO.
In vivo evaluation evinced very high uptake in the adrenal glands (7.52 ±
1.19% ID/g at 30 min), followed by lung (1.18 ± 0.19% ID/g, 10 min), liver (0.59
± 0.13% ID/g, 10 min) and duodenum (0.7 ± 0.29% ID/g, 60 min) (Figs 12–14).
No defluorination or fluoroethyl-ester cleavage was observed. As etomidate-
analogues have been used as anesthetics for a long time and the way of action is
explained GABA-ergic [61, 62], the authors in addition evaluated binding
parameters on GABA receptors.
Brain regions have been compared and showed highest relative uptake in
the cortex (2.34) followed by rest brain (2.13), cerebellum (1.96) and thalamus
(1.0, reference value). FE@ETO and ETO were able to increase the binding of
[3H]flunitrazepam with similar potencies and to a comparable extent (Fig. 15).
FE@ETO shows characteristics suitable for the imaging of adrenocortical
pathology with PET. Binding experiments on GABA receptors demonstrate a
comparable effect of FE@ETO and ETO (Fig. 16). Hence, FE@ETO possibly
anhydrous DMF
O O
Br 135°C-150°C, 20-30min
+
+ TBA O - N 18 O N
18 F
F
[18F]BFE N N
172
SESSION 10
9 ,0
8 ,0
7 ,0
1 0 m in 3 0 m in 6 0 m in
6 ,0
5 ,0
% ID/g
4 ,0
3 ,0
2 ,0
1 ,0
0 ,0
nd
s
d
um
lon
als
t
art
s
ng
r
x
h
um
um
s
er
ain
le
Fa
mu
ey
mu
rt e
e
oo
lee
ac
sc
Liv
yro est
gla
Lu
He
ren
br
Co
en
ell
ec
dn
om
Bl
Co
Fe
Mu
Sp
ala
T
re b
Ca
od
st
Ki
id
Ad
St
Th
Re
Du
Ce
Th
FIG. 12. The values counted in various organs at different time points after application of
1.73–3.06 MBq FE@ETO. Bars express values as per cent injected dose per gram organ
(%ID/g).
173
MITTERHAUSER and WADSAK
100
80
60
40
20
0
Adrenal Liver Heart Kidney Brain
FIG. 14. Relative binding of FE@ETO, ETO and MTO to various rat tissues; normalized
to adrenal binding (100%).
300
250
200
relative binding (%)
150
100
50
0
Thalamus Cerebellum Cortex Rest Brain
FIG. 15. Calculated ratios of brain regions: cerebellum, cortex and rest brain calculated as
per cent of thalamic uptake.
174
SESSION 10
100
80
relative binding (%)
60
40
20
FIG. 16. Organs with the highest uptake. Ratios are expressed as %ID/g organ/blood at
the time of highest uptake (lung, liver 10 min; adrenals 30 min; duodenum 60 min).
could also be used to elucidate the function, dynamics and kinetics of narcotic
drugs with PET.
6.5. [18F]FE@CFN
175
MITTERHAUSER and WADSAK
7. CONCLUSION
2-Bromoethyltriflate [18F]BFE
Br anhydrous DMF
N N
18 + N N
150°C, 20 min
2. F
O O
O O
O- O
TBA+
18
F
176
SESSION 10
177
MITTERHAUSER and WADSAK
100 a
100 b
rc. yield [%]
80
60
40
20
0
0 5 10 15
precursor concentration [mM]
100 c
rc. yield [%]
80
60
40
20
0
0 5 10 15 20
time [min]
FIG. 18. The evaluated reaction conditions for the preparation of [18F]FE@CFN.
specific synthetic demand has to be evaluated separately for each single case.
For example, the addition of NaI has been reported to increase the radio-
labelling yields [38, 80], but in the authors’ experience with fluoroethylated
esters, this effect is reversed (a significant reduction of the labelling yield was
observed) [32].
In conclusion, fluoroethylations represent an important and valuable
strategy to widen the list of available PET radiotracers without the necessity of
synthesizing compounds ‘from scratch’. A major drawback of the method is
hidden in the fact that fluoroethylated compounds do not exist in nature and
are rarely evaluated drugs. This fact demands de novo biomedical evaluations
prior to their use for human PET applications in routine diagnosis. Fluoroethyl-
ation is a method with restrictions but once established it can serve as a valuable
tool for the evaluation and development of new and innovative PET tracers.
178
SESSION 10
cerebellum
cerebral cortex
brain stem
olfactory bulb midbrain
ACKNOWLEDGEMENTS
The authors are indebted to Profs Kletter and Dudczak, Drs Ettlinger,
Lanzenberger and Mien, and all the staff at the PET centre Vienna. Parts of the
presented work were supported by two research grants from the Austrian
National Bank (Jubiläumsfonds der Österreichischen Nationalbank), project
numbers 8263 (W. Wadsak) and 11439 (M. Mitterhauser).
REFERENCES
179
MITTERHAUSER and WADSAK
180
SESSION 10
181
MITTERHAUSER and WADSAK
182
A NOVEL FINDING: ANTI-ANDROGEN FLUTAMIDE
KILLS ANDROGEN INDEPENDENT PC-3 CELLS
A radiolabelled methyl-choline incorporation into tumour
cells
F. AL-SAEEDI
Nuclear Medicine Department, Faculty of Medicine,
Kuwait University (Health Sciences Center),
Safat, Kuwait
Email: Fatimas@hsc.edu.kw
Abstract
183
AL-SAEEDI
1. INTRODUCTION
Many studies suggest that changes in the uptake of PET tracers [1–3] such
as 18F-2-fluoro-2-deoxy-D-glucose (18F-FDG) and 18F-fluorothymidine (18F-
FLT) after chemotherapy (4, 5), and radiotherapy (6, 7), compared with
pretreatment uptake can reflect the efficacy of the drug and the tumour
responses to chemotherapy treatment [8]. 18F-FDG-PET has been widely used
to investigate many types of cancer such as breast cancer and colorectal cancer
after chemotherapy [9]. However, 18F-FDG imaging was found to be limited in
the detection of localized prostate cancer because of excessive excretion of the
radionuclide into the urine, masking any lower pelvic region lesions [10]. The
evaluation of the efficacy of the treatment of men with prostate cancer is
mainly based on post-treatment levels of the prostate specific antigen. In
addition, digital rectal examination and conventional imaging techniques are
used but they are not sensitive enough to detect a local recurrence. [Methyl-
11
C]-choline PET scan provides a sensitive metabolic and non-invasive imaging,
which is not dependent on anatomical distortions. Recently, [Methyl-11C]-
choline was introduced to image prostate cancer [1], and in addition to evaluate
the post-treatment of the localized prostate cancer [11]. Flutamide is often used
as part of the initial treatment of prostate cancer. In this study, the questions of
whether flutamide treatment inhibits cell proliferation and whether
modulation of [Methyl-3H]-choline incorporation is induced or not in PC-3
cells were investigated.
2.1. Materials
The human prostate cancer cells (PC-3 cells) were obtained from the
American Type Culture Collection (ATCC).
184
SESSION 10
2.2. Methods
Cell viability was assessed using both the MTT assay and the trypan blue
exclusion methods. For MTT assay, cells were seeded in flat bottomed 96 well
tissue culture plates at a concentration of 1 × 105 cells/mL medium in a volume
of 200 µL per well and were allowed to grow to 70% confluency before adding
drugs. The plates were set up for controls and different drug concentrations,
then incubated in a humidified atmosphere with 5% CO2:95% air at 37°C.
After reaching 70% confluency, different concentrations of flutamide were
added separately into triplicate plates of PC-3 cells and were incubated for 3 d,
after which the medium was pipetted out and new medium was added in a final
volume of 170 µL per well. Then, 30 µL of MTT (5 mg/mL in phosphate
buffered saline (PBS)) was added to each well and incubation of the cells
continued. After 4 h, the medium was carefully pipetted out, leaving the
adherent cells and precipitate in the wells. A volume of 200 µL of DMSO was
then added to each well with gentle mixing for 20 min to dissolve the
precipitate and the appearance of crystals was checked under the microscope.
Then plates were read in a 96 well plate scanner (ELISA plate reader) at dual
filter wavelengths of 540 and 690 nm.
185
AL-SAEEDI
Cells were incubated in triplicate for 10 min at 37°C with 37 kBq [Methyl-
3
H]-choline/mL of DMEM medium in 25 cm2 flasks, washed 6 times with ice
cold PBS, then incubated with non-radioactive DMEM medium for 10 min.
The cells were washed once with ice cold PBS, trypsinized with 0.5 mL trypsin,
neutralized with 0.5 mL medium and centrifuged at 10 000g for 5 min at 4°C.
Activity in the media was counted using a liquid scintillation counting analyser
(LSC-1900CA; Tri-CARB; Packard). Cell pellets were washed once with PBS
to remove extracellular protein and the intracellular location of [Methyl-3H]-
choline determined by extraction with buffer and organic solvents.
186
SESSION 10
Cells harvested by trypsinization were washed twice in ice cold PBS then
resuspended in 100 µL PBS. A volume of 1 mL of ice cold 70% ethanol was
added to the cell suspension whilst vortexing and the cells left overnight at
-20°C for flow cytometry. For flow cytometry analysis, cell number was adjusted
to 1 × 106 cells/sample. A volume of 1 mL of PI/RNAse and triton x-100
staining buffer was added to the cells and the suspension incubated for 20 min,
protected from light at room temperature. Flow cytometry was performed
using a 488 nm laser on a FACSCalibur flow cytometer (Becton Dickinson) and
CELLQuest software (Becton Dickinson) equipped for fluorescence detection,
forward, 90° angle light scatter and doublet discrimination.
3. RESULTS
Figure 1 shows the treatment effect, viability and IC50 for flutamide on
PC-3 cells using MTT (%MTT, n = 12) and trypan blue (%TB, n = 3) tests
compared to controls. The IC50 for flutamide on PC-3 cells was 10nM.
Therefore, PC-3 cells were treated with 10nM flutamide for 3 d in the
subsequent experiments.
100
Viability (%)
75
50
25
0
0 10 20 30 40 50 60 70 80 90 100
Concentration (nM)
FIG. 1. The viability tests using MTT and trypan blue after 3 d exposure to flutamide of
PC-3 cells. Cell viability is expressed as the mean ± SD of the values obtained from three
independent experiments of TB (open circles) and from 12 replicates of MTT 96 well
plates (closed circles).
187
AL-SAEEDI
4. DISCUSSION
G1 S G2 G1 S G2
FIG. 2. DNA histograms of PC-3 cells alone (left) and when incubated with 10nM
flutamide for 3 d (right).
188
SESSION 10
189
AL-SAEEDI
the AR in the prostate. Inaloz et al. [24] have shown that the administration of
flutamide in rats inhibits skin growth and proliferation in parallel with a
decrease in the epidermal growth factor, a key hormone for skin development,
and is known to stimulate cell proliferation and differentiation in a wide range
of tissues. Flutamide may result in the inhibition of epidermal growth factor
action in PC-3 cells, leading to the inhibition of the cells proliferation and their
DNA synthesis, and result in the subsequent alteration to their cellular
phospholipid metabolism, especially choline incorporation after the treatment
with flutamide, and this is may involve the inhibition of phospholipase D
activity.
Another possibility could be mediated through oestrogen receptors. For
example, Kawashima et al. [25] reported that in PC-3 cells both the cell growth
and the AR activity were remarkably inhibited by tamoxifen (TAM) at 50µM
and flutamide at 5–50µM. They reported that anti-oestrogens modulate the
transactivation activity of the AR in prostate cancer cells. Although the most
prominent characteristic of prostate cancer is its androgen dependence,
oestrogen seems to be involved. In the human prostate, ERα is localized in the
stromal tissue of the prostate and associated with the progression, metastasis
and the hormone refractory phenomenon of the prostate cancer [26, 27]. It was
reported that both ERα and ERβ were expressed in PC-3 cells [28]. The
activation of ERβ by its putative ligands 5α, androstane-3β and 17β-diol was
reported to suppress the growth of the ventral prostate [29]. Activation of such
a receptor may be involved in inhibition of cell proliferation and subsequently
reduced choline incorporation in PC-3 cells treated with flutamide.
Flutamide may act through similar mechanisms to those involved in the
inhibitory effect of TAM, the widely used anti-oestrogen therapy for breast
cancer, which are unclear at present. Flutamide may act through caspase
activation. Some studies reported that anti-oestrogen, raloxifen induces
apoptosis in androgen independent prostate cancer cells through caspase
activation [30] and TAM inhibits prostate cancer by inhibiting protein kinase C
followed by the induction of p21(waf1/cip1) [31]. Kiss and Crilly [32] reported
that in an ER deficient multidrug resistant subline of MCF-7 human breast
carcinoma cells, clinically relevant concentrations of TAM inhibited the uptake
and phosphorylation of choline and, as a result, the synthesis of corresponding
phospholipids. Here, PC-3 cells treated with flutamide showed dose dependent
reduced [Methyl-3H]-choline uptake, reflecting a decrease in phospholipid
synthesis. Moreover, flutamide can induce apoptosis in PC-3 cells through the
formation of free radicals [33], which will inhibit the ability of cells to
synthesize new DNA and ultimately reduce its choline uptake. In this study
flutamide inhibited cell growth and proliferation as well as [Methyl-3H]-choline
incorporation in the AR negative PC-3 cells in vitro. Currently, the study is
190
SESSION 10
REFERENCES
[1] HARA, T., KOSAKA, N., KISHI, H., PET imaging of prostate cancer using
carbon-11-choline, J. Nucl. Med. 39(6) (1998) 990-5.
[2] AL-SAEEDI, F., WELCH, A.E., SMITH, T.A.D., [Methyl-3H]-Choline Incorpo-
ration Into MCF-7 Tumour Cells: Correlation with Proliferation, Eur. J. Nucl.
Med. Mol. Imaging (2005) (in press).
[3] AL-SAEEDI, F., WELCH, A.E., SMITH, T.A.D., Radiolabelled Methyl-Choline
Incorporation Into Tumour Cells. Relationship with Proliferation and Chemo-
therapy-Induced Modulation. 12th European Symposium on Radiopharmacy and
Radiopharmaceuticals ESRR'04. September 9-12, 2004 Gdansk/Poland.
[4] ALLAL, A.S., et al., Standardized uptake value of 2-[(18)F] fluoro-2-deoxy-D-
glucose in predicting outcome in head and neck carcinomas treated by radio-
therapy with or without chemotherapy, J. Clin. Oncol. 20(5) (2002)1398-404.
[5] LICHY, M.P., et al., Monitoring individual response to brain-tumour chemo-
therapy: proton MR spectroscopy in a patient with recurrent glioma after stereo-
tactic radiotherapy, Neuroradiology 46(2) (2004)126-9.
[6] HIGASHI, K., CLAVO, A.C., WAHL, R.L., In vitro assessment of 2-fluoro-2-
deoxy-D-glucose, L-methionine and thymidine as agents to monitor the early
response of a human adenocarcinoma cell line to radiotherapy, J. Nucl. Med. 34(5)
(1993) 773-9.
[7] KUBOTA, K., et al., Tracer feasibility for monitoring tumor radiotherapy: a
quadruple tracer study with fluorine-18-fluorodeoxyglucose or fluorine-18-fluor-
odeoxyuridine, L-[methyl-14C]methionine, [6-3H]thymidine, and gallium-67, J.
Nucl. Med. 32(11) (1991) 2118-23.
[8] HABERKORN, U., et al., Metabolic design of combination therapy: use of
enhanced fluorodeoxyglucose uptake caused by chemotherapy, J. Nucl. Med.
33(11) (1992) 1981-7.
[9] KRAK, N.C., HOEKSTRA, O.S., LAMMERTSMA, A.A., Measuring response
to chemotherapy in locally advanced breast cancer: methodological considera-
tions, Eur. J. Nucl. Med. Mol. Imaging 31 (2004) Suppl 1:S103-11.
[10] LIU, I.J., ZAFAR, M.B., LAI, Y.H., SEGALL, G.M., TERRIS, M.K., Fluorode-
oxyglucose positron emission tomography studies in diagnosis and staging of
clinically organ-confined prostate cancer, Urology 57(1) (2001)108-11.
191
AL-SAEEDI
[11] DE JONG, I.J., PRUIM, J., ELSINGA, P.H., VAALBURG, W., MENSINK, H.J.,
11C-choline positron emission tomography for the evaluation after treatment of
localized prostate cancer, Eur Urol 44(1) (2003a) 32-8; discussion 38-9.
[12] BLIGH, E.G., DYER, W.J., A rapid method of total lipid extraction and purifica-
tion, Can J Med Sci 37(8) (1959) 911-7.
[13] HOBSON, R.S., BEYNON, A.D., Preliminary quantitative microradiography
study into the distribution of bone mineralization within the basal bone of the
human edentulous mandible, Arch Oral Biol 42(7) (1997) 497-503.
[14] KOTZERKE, J., et al., Experience with carbon-11 choline positron emission
tomography in prostate carcinoma, Eur. J. Nucl. Med. 27(9) (2000) 1415-9.
[15] ZHENG, Q.H., et al., [11C]Choline as a PET biomarker for assessment of
prostate cancer tumor models, Bioorg Med Chem 12(11) (2004) 2887-93.
[16] KAIGHN, M.E., NARAYAN, K.S., OHNUKI, Y., LECHNER, J.F., JONES, L.W.,
Establishment and characterization of a human prostatic carcinoma cell line (PC-
3), Invest Urol 17(1) (1979) 16-23.
[17] WARE, J.L., PAULSON, D.F., MICKEY, G.H., WEBB, K.S., Spontaneous metas-
tasis of cells of the human prostate carcinoma cell line PC-3 in athymic nude mice,
J Urol 128(5) (1982) 1064-7.
[18] KOZLOWSKI, J.M., et al., Metastatic behavior of human tumor cell lines grown
in the nude mouse, Cancer Res 44(8) (1984) 3522-9.
[19] PIZZI, H., et al., Androgen regulation of parathyroid hormone-related peptide
production in human prostate cancer cells, Endocrinology 144(3) (2003) 858-67.
[20] CATALONA, W.J., Management of cancer of the prostate, N Engl J Med 331(15)
(1994) 996-1004.
[21] BRUFSKY, A., et al., Finasteride and flutamide as potency-sparing androgen-
ablative therapy for advanced adenocarcinoma of the prostate, Urology 49(6)
(1997) 913-20.
[22] FULHAM, M.J., et al., Mapping of brain tumor metabolites with proton MR
spectroscopic imaging: clinical relevance, Radiology 185(3) (1992) 675-86.
[23] DeGRADO, T.R., et al., Synthesis and evaluation of 18F-labeled choline as an
oncologic tracer for positron emission tomography: initial findings in prostate
cancer, Cancer Res 61(1) (2000) 110-7.
[24] INALOZ, H.S., KETANI, M.A., INALOZ, S.S., YILMAZ, F., KETANI, S., The
effects of sialoadenectomy & flutamide on skin development, Clin Exp Obstet
Gynecol 27(3-4) (2000) 231-4.
[25] KAWASHIMA, H., et al., Effect of anti-estrogens on the androgen receptor
activity and cell proliferation in prostate cancer cells, Urol Res. (2004).
[26] GRIFFITHS, K., Estrogens and prostatic disease. International Prostate Health
Council Study Group, Prostate 45(2) (2000) 87-100.
[27] BONKHOFF, H., FIXEMER, T., HUNSICKER, I., REMBERGER, K.,
Estrogen receptor expression in prostate cancer and premalignant prostatic
lesions, Am J Pathol 155(2) (1999)641-7.
192
SESSION 10
[28] LAU, K.M., LaSPINA, M., LONG, J., HO, S.M., Expression of estrogen receptor
(ER)-alpha and ER-beta in normal and malignant prostatic epithelial cells: regu-
lation by methylation and involvement in growth regulation, Cancer Res 60(12):
(2000) 3175-82.
[29] WEIHUA, Z., et al., A role for estrogen receptor beta in the regulation of growth
of the ventral prostate, Proc Natl Acad Sci U S A 98(11) (2001) 6330-5.
[30] KIM, I.Y., et al., Raloxifene, a mixed estrogen agonist/antagonist, induces
apoptosis in androgen-independent human prostate cancer cell lines, Cancer Res
62(18) (2002) 5365-9.
[31] ROHLFF, C., et al., Prostate cancer cell growth inhibition by tamoxifen is associ-
ated with inhibition of protein kinase C and induction of p21(waf1/cip1), Prostate
37(1) (1998) 51-9.
[32] KISS, Z., CRILLY, K.S., Tamoxifen inhibits uptake and metabolism of
ethanolamine and choline in multidrug-resistant, but not in drug-sensitive, MCF-
7 human breast carcinoma cells, FEBS Lett 360(2) (1995) 165-8.
[33] NUNEZ-VERGARA, L.J., FARIAS, D., BOLLO, S., SQUELLA, J.A., An elec-
trochemical evidence of free radicals formation from flutamide and its reactivity
with endo/xenobiotics of pharmacological relevance, Bioelectrochemistry 53(1)
(2001) 103-10.
193
.
A SEMI-AUTOMATED [13N]NH3 PRODUCTION
MODULE: DESIGN, QUALITY CONTROL AND
OPTIMIZATION
Abstract
The compound [13N] NH3 was prepared using a home-made, cost effective
prototype module meeting pharmaceutical criteria. The 16O(p,α)13N nuclear reaction was
chosen as the best method of 13N production. High purity natural water was bombarded
by 18 MeV protons with a current intensity of 8 μA. A production yield of 13.5 mCi·μA-1·h-1
was obtained. Chemical modification of [13N]nitrogen oxides into [13N]NH3 was accom-
plished using in-house made De Varda’s catalyst. Chemical, radiochemical and radionu-
clide purity controls performed on the final product showed high radiopharmaceutical
purity in the form of [13N]NH3 produced by a prototype production module. [13N]NH3
was formulated in normal saline media under appropriate pH and sterile conditions.
Microbial and LAL tests were performed on the final product in order to assure its
safety for administration to experimental subjects.
1. INTRODUCTION
195
JALILIAN et al.
13
N + H-OH Æ 13NH3
13
NH3 + HOH Æ 13NH4++ OH–
13
NH4+ ….Æ 13NO3– + 13
NO2–
Using anion retardation resins, the anions can be removed from the target
solution. Therefore, by the addition of various radical scavengers such as
ethanol or acetic acid, the [13N]NH3 compound will be the major product. The
use of reducing agents such as De Varda’s catalyst or Ti salts [8] may be the
alternative.
2. EXPERIMENTAL
196
SESSION 10
In this research, 16O(p,α)13N was selected as the best nuclear reaction for
the production of 13N, using natural water as the target material, owing to the
small amount of produced 18O resulting in 18F, that could be easily separated by
physical methods and also for cost effectiveness. Other metal impurities such as
51
Cr, 52Mn, 55Co, 56Co, 57Co,58Co and 57Ni radioisotopes were also produced as a
result of proton irradiation of the windows and target body, but they could be
easily removed in the radiochemical or physical separation process. The
optimum proton beam energy was calculated and types of possible impurities
197
JALILIAN et al.
were predicted using previous experimental data given in the literature which
showed that the maximum production yield can be achieved with the lowest
level of radioactive impurities at 18 MeV proton energy.
198
SESSION 10
199
JALILIAN et al.
14000 12000
13000
11000
12000
10000
11000
9000
10000
9000 8000
8000 7000
A.U.C
A.U.C.
7000 6000
6000
5000
5000
4000
4000
3000 3000
2000 2000
1000 1000
0
0
1 2 3 4 5 6 7 8 9 10
1 2 3 4 5 6 7 8 9 10
Chromatogram Slices
Chromatogrm Slices
In order to ensure the presence of 13N in the final solution and the
absence of other PET radionuclides, activity measurements were performed on
radiopharmaceutical samples (3.2 ± 0.1 mCi) and the activities were plotted
versus time (Fig. 3).
3.5
2.5
Activity (mCi)
1.5
0.5
0
0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30
Time (min)
200
SESSION 10
80
16 15
O(p,pn) O
70
50
40
30
20
M.G.Albouy et al 1962
M.Sajjad et al - 1985
10
S.W.Kitwanga et al - 1989
0
15 25 35 45 55 65 75 85 95 105 115 125 135 145
Proton Energy (MeV)
The experimental data given in the literature for the 16O(p,α)13N nuclear
reaction were compared in order to select the optimum proton beam energy
(Fig. 4) [9]. It is clearly obvious that all the results with the exception of one
report are in good agreement.
The best proton energy for this reaction is 18 MeV. At this energy, the
16
O(p,pn)15O nuclear reaction which also takes place has been reported by
many reseach groups [10–12] (Fig. 5). The probability of 15O production is
200
16 13
O(p,α) N M.G.Albouy et al - 1962
H.A.Hill et al - 1961
160
Furukawa et al - 1960
Whitehead et al - 1985
140
Cross-Section (mb)
S.W.Kitwanga et al - 1989
120 W.Gruhle et al - 1977
S.Takacs et al - 2003
100
P.Guazzoni et al - 1971
80
60
40
20
0
5 10 15 20 25 30 35 40 45
Proton Energy (MeV)
201
JALILIAN et al.
higher at energies above 18 MeV. The SRIM nuclear code was used for target
thickness determination [13].
[13N]NH3 was prepared by 18 MeV proton bombardment of the natH2O
target. The target was bombarded with a current intensity of 8 μA for 20 min.
The chemical separation process was based on a distillation method. The
resulting activity of [13N]NH3 was 36 mCi at the end of bombardment and the
production yield was 13.5 mCi·μA-1·h-1 (Fig. 6).
The production yield obtained in this study was comparable to other
published data. The yield was calculated in the hot cell, at 10 m distance from
the target room using a long PE hose that imposed a percentage of activity loss
through the line. The ion exchange chromatography method proposed in some
previous publications was rather time consuming. Radiochemical separation
was performed by a one step reduction of nitroxy anions in a sealed disposable
chamber containing in-house made De Varda’s catalyst.
The reaction took place as soon as the target aqueous content was added to
the catalyst chamber equipped with a small stirrer rod and in less than 40–60 s the
maximum formation of NH3 took place with a yield of more than 95%. Quality
control of the product was performed in two steps. Radionuclide control
FIG. 6. [13N]NH3 production module details: (1) aluminium window, (2) target outlet, (3)
proton beam, (4) cooling gas, (5) target inlet, (6) irradiated liquid, (7) silver body target,
(8) Havar window, (9) 6-wedge valve, (10) module inlet valve, (11) catalytic reduction unit
outlet, (12) catalytic reduction unit inlet, (13) De Varda’s catalyst, (14) gas buffer chamber,
(15) 0.22 μm microbial filter, (16) dose calibrator, (17) final sterile solution, (18) over
pressure outlet, (19) excess ammonia chemical trap.
202
SESSION 10
1000
10 S.Takacs et al - 2003
M.Sajjad et al - 1986
1 N.N.Krasnov et al - 1969
M.Sajjad et al - 1985
This Work
0.1
0.01
0.001
6 9 12 15 18 21 24 27 30 33
Proton Energy (MeV)
FIG. 7. Comparison of the literature data with the production yield in the present work.
13
showed the presence of 511 keV gamma energy, originating from N, and
purity was higher than 99%.
203
JALILIAN et al.
As a result of the use of a high purity natural water sample, the presence
of O nuclide in natural oxygen (0.038%) could lead to the formation of 18F via
18
the 18O(p,n)18F nuclear reaction. By selection of the best proton energy, this
possiblity was minimized, but the possibilty of forming 18F ionic forms in the
target still remained. It was shown that if the De Varda alloy pretreatment were
not performed properly, the presence of impurities in the starting materials,
such as organic traces, etc., could produce amounts of volatile 18F-fluorinated
wastes that could be distilled from the reaction vessel and lead to the contami-
nation of the final product. By carefully taking into account the probabilities in
a normal run, the half-life determination of the final sample could be an
important criterion for the presence of long half-life PET radionuclides such as
18
F that could not be identified by gamma spectroscopy. A half-life determi-
nation diagram is illustrated in Fig. 3.
The Pyrogen test was performed using a commercial LAL kit (sensitivity
0.125 EU/mL, Charles River Endosafe Co., United States of America).
Microbial/fungal tests showed suitable pharmaceutical sterility.
4. CONCLUSION
The method used in this research for the production and chemical
separation of [13N]NH3 was quite simple and cost effective, while previous
studies given in the literature have reported a production yield similar to the
author’s work (Fig. 7). Total labelling and formulation of [13N]NH3 took about
2 min, with a yield of 98%. A significant specific activity (≈1200 Ci/mmol) was
formed via production of [13N]nitroxy anions. No unlabelled and/or labelled
by-products were observed upon RTLC analysis of the final preparations by
chemical determination tests. The radiopharmaceutical was usable in aqueous
solution for at least 25 min and no significant amounts of other radioactive
species were detected by RTLC. Trace amounts of other PET radionuclides
204
SESSION 10
REFERENCES
[1] MASUDA, D., et al., Improvement of regional myocardial and coronary blood
flow reserve in a patient treated with enhanced external counterpulsation: evalu-
ation by nitrogen-13 ammonia PET. Jpn Circ J 1999 May; 63(5): 407-411.
[2] PHELPS, M.E., HOFFMAN, E.J., RAYBOUND, C., Factors with affect cerebral
uptake and retention of N-13 NH3. Stroke 1977; 8: 694-702.
[3] KITSIOU, A.N., et al., 13N-ammonia myocardial blood flow and uptake: relation
to functional outcome of asynergic regions after revascularization, J Am Coll
Cardiol 1999 Mar; 33(3): 678-686.
[4] SAWADA, S., et al., Interobserver and interstudy variability of myocardial blood
flow and flow-reserve measurements with nitrogen 13 ammonia-labeled positron
emission tomography. J Nucl Cardiol 1995 Sep-Oct; 2(5): 413-422.
[5] JALILIAN, A.R., et al., Production Iranian Journal of Nulear Medicine, 12, 13-24,
1998.
[6] JALILIAN, A.R., J. Pharmacy & Pharmaceutical Sciences, 3(1) January-April,
2000. 114-124.
[7] JALILIAN, A.R., et al., Iranian Journal of Nulear Medicine, 21, 49-62, 2004.
[8] AKUTSU, Y., et al., Jpn Circ J 1997 Aug; 61(8): 665-672.
[9] SAJJAD, M., et al., Radiochimica Acta 1986; 39: 165-168.
[10] ALBOUY, M.G., et al., Spallation of oxygen by protons with 20 to 150 MeV, J
Phys Lett 1962; 2:306.
[11] KITAWANGA, S.W., LeLEUX, P., LIPNIK, P., VANHORENBEECK, J.,
Production of N-13 radioactive nuclei from 13C(p,n) or 16O(p,α) reactions, J Phys
Rev/C 1989 ; 40: 35.
[12] SAJJAD, M., LAMBRECHT, R.M., WOLF, A.P., Cyclotron isotopes and radiop-
harmaceuticals, Investigation of some excitation functions for the preparation of
15O, 13N, 11C, J Radiochimica Acta 1985; 38: 57.
[13] ZIEGLER, J., The code of SRIM- the Stopping and Range of Ions in Matter,
Version 2000.XX, (2000).
[14] GATLEY, S.J., et al., Int J Rad Appl Instrum [A], 1991; 42(9): 793-796.
205
.
RADIOIODINE RADIOPHARMACEUTICALS
(Session 11)
Chairpersons
H.S. BALTER
Uruguay
G.-J. BEYER
Switzerland
.
PRODUCTION OF RADIOIODINES WITH MEDICAL
PET CYCLOTRONS
Abstract
The authors discuss the possibility of producing radioiodine with mass numbers
120, 123 and 124 via proton reactions on enriched TeO2 targets with medical PET cyclo-
trons under three aspects: yield and impurity considerations, technical feasibility and
radiopharmaceutical chemistry. The thick target yield for the XXXTeO2(p,n)XXXI process
is of the order of 2.5 × 109 atoms and relatively independent of the mass number. Conse-
quently, the (p,n) process could be a suitable alternative to the well-known 124Xe(p,x)
method — large scale production technology for 123I. Concerns regarding radionuclide
impurities do not play a role any longer, because of availability of the target material
with an enrichment close to 100% without increased costs. The standard medical PET
cyclotrons and a standardized separation technique can be used for the production of all
three isotopes: 120I, 123I and 124I in high purity via the (p,n) reaction. A suitable target
consists of a platinum disc carrying about 200–300 mg of TeO2. A dedicated COmpact
Solid Target Irradiation System (COSTIS) has been developed, which is now available
commercially. Irradiation conditions are: 13–15 MeV protons, 20 µA/cm2 beam intensity.
Thus, 10 GBq or 1.5 GBq batches of 123I or 124I, respectively, are practically available
using a PET cyclotron. The radioiodine is separated from irradiated TeO2 targets using
a thermochromatographic process. The TeO2 targets cycled in this way can be reused
immediately for the next production run without further treatment. A corresponding
good manufacturing practice conform separation module has now been developed
(TERIMO = TEllurium based RadioIodine production MOdule) and is available
commercially. The losses of target material for one cycle is negligible (<0.2 mg/cycle).
Local in-house production of radioiodine has the advantage of obtaining an iodinated
radiopharmaceutical compound. Preliminary examples along this line will be presented.
The technology described is identical for all three mentioned iodine isotopes with the
mass numbers 120, 123 and 124.
209
ČOMOR et al.
1. INTRODUCTION
210
SESSION 11
description of a GMP compliant technology for the production of 120gI, 123I and
124
I, based on a COmpact Solid Target Irradiation System (COSTIS) and on a
TEllurium oxide based RadioIodine separation MOdule (TERIMO).
211
ČOMOR et al.
FIG. 2. Stopping of protons in the target disc used for radioiodine production. The
thickness of the TeO2 layer is 0.6 mm; after passing this layer the protons are stopped in the
platinum backing.
212
SESSION 11
Mass number 120 122 123 124 125 126 128 130
Natural abundance 0.096 2.60 0.908 4.816 7.14 18.95 31.69 33.80
Target material composition 0.01 2.3 67.3 11.7 3.9 5.7 5.0 4.1
Radionuclide composition 100 2.3 0.04 0.26 2.1
the enriched 123Te is not the most important quality parameter of a target
material; it is much more important to have a very small 124Te/123Te ratio [23].
Nowadays, the manufacturer of enriched isotopes can provide a 123Te target
material with a 124Te/123Te ratio of 6 × 10-4 [24]. With such a high purity target
material and compact PET cyclotrons, it is possible to produce, via the (p,n)
process, 123I having all the quality parameters (radionuclidic purity as well as
radionuclide concentration) comparable to the high purity 123I product
obtained in the well-known large scale 124Xe(p,x)123I production process. The
(p,n) process, however, cannot compete with respect to the productivity.
The chemical form of the target material is another important factor for
the design of the radioiodine production technology. In general, metallic
targets are preferred due to their better heat conduction and mechanical
stability. However, in the case of tellurium, targets in the form of TeO2 are the
most suited chemical form for two reasons: first, the vapour pressure of
tellurium oxide is lower than that of metallic tellurium at the same temper-
ature, thus less expensive target material is lost during irradiation and thermal
processing; second, tellurium oxide targets might be reused immediately after
thermochromatographic separation of the produced radioiodines, without
troublesome wet chemical processing of the irradiated targets.
Even though compact medical cyclotrons can deliver rather high currents
of proton beams (50–100 µA are common), the large scale production of radio-
iodines with them is not feasible as the beam power acceptance of the tellurium
oxide targets is limited. Practical experiences have shown that a p-beam density
213
ČOMOR et al.
of 20 µA·cm-2 seems to be about the limit. If one distributes the p-beam over a
larger area, aiming to increase the productivity, one creates the problem of
inconvenience in the following separation process. Nonetheless, batches of the
order of 3.7 GBq 124I have already been obtained on a routine basis using the
technology described in this paper. The investment needed for such a small
scale production is reasonably low, i.e. the usage of sophisticated target stations
and an automated rabbit target transport system is not really needed and not
justified. However, the automation of the post-irradiation target processing is
justified, as automation can provide reproducibility, repeatability and reliability
required by current GMP for the production of radiopharmaceuticals.
3.1. Targetry
a b
1 2 3 4 5 6 7 8
FIG. 3. (a) Simplified cross-sectional view of COSTIS: (1) quick connection flange; (2)
water cooled collimator with graphite insert; (3) insulating alumina rings; (4) helium
cooling cavity; (5) helium inlet; (6) platinum target disc with TeO2 in its central groove; (7)
water jet; (8) cooling water inlet; (9) cooling water outlet; (10) helium outlet; (11) window
foil. (b) Loading of the target coin into COSTIS.
214
SESSION 11
COSTIS
a b
FIG. 4. COSTIS installed on a GE PETrace (a) and IBA Cyclone 18/9 (b) cyclotrons
(courtesy of IBA Molecular Imaging).
common compact medical cyclotrons. The beam passes through a water cooled
graphite collimator, which forms a circular beam 12 mm in diameter. The next
element is a window foil separating the helium cooling loop from the vacuum.
The helium stream in this loop cools the window foil as well as the front face of
the target disc. Finally, a water jet cools the back face of the target disc.
The target is a 2 mm thick platinum disc, 24 mm in diameter with a
circular groove, 12 mm in diameter, in its centre. This groove is filled with the
target material, for instance with isotopically enriched TeO2, which is melted
into the groove. Platinum is selected as a metal with a sufficiently high thermal
conductivity, which is also chemically inert and not heavily activated by the
beam.
The target disc is loaded manually before the irradiation. It is locked in
position by two pneumatic actuators during the irradiation. By reversing the
action of these actuators, the irradiated target disc is released into a shielded
transport container conveniently placed below COSTIS before the irradiation.
COSTIS has been designed as a compromise between high power
metallic Te targets, which require troublesome post-irradiation radiochemical
processing, and compact low power TeO2 targets, which allow for easy
separation of the produced radioiodine by thermal chromatography. After dry
distillation of iodine from the melted oxide and cooling down of the target disc,
it can be immediately reused for the next production cycle without any further
preparation. This makes the production process simple and cost effective, even
though the yields are limited by the beam power that such an oxide target may
withstand, which is typically of the order of 350 W. Since January 2005, COSTIS
is commercially available from IBA S.A., Louvain-la-Neuve, Belgium.
215
ČOMOR et al.
p2
ln
t 1/ 2 = 216 h 2
p D
where t1/2 is the release half-life, D is the diffusion coefficient and h is the
thickness of the TeO2 layer. However, if convection inside the liquid phase is the
time determining step, one can use the following simple equation for describing
the relation between fractional release (F) and the release time (t) [16]:
F ( t ) = e - m 1t
where the empirical factor µ1 describes the desorption process from the surface.
The radioiodine release kinetics from TeO2 targets have been extensively
studied [16] and it became evident that the release rate is proportional to the
target thickness to diameter ratio, provided that circular targets are employed.
From the dependence of the release half-time on this ratio one can easily
determine that the release half-time for targets designed to be irradiated on
COSTIS is 26.3 s (see Fig. 5); thus 97% of the produced radioiodine will be
released within just 2 min. This is of utmost importance, as the vapour pressure
of molten TeO2 is not negligible, thus the faster the radioiodine release is, the
shorter the time the target has to be kept in a molten state and the lower the
losses of expensive enriched target material. The loss of target material during
the thermochromatographic process has to be taken into account as a potential
source of contamination of the final product as well.
The apparatus for thermochromatographic separation of radioiodine
from the irradiated TeO2 is shown schematically in Fig. 6. It has a quartz target
holder for positioning of the target discs, a fast electric heater, a thermocouple
for temperature measurements, an alumina trap for adsorbing tellurium oxide
vapours and a quartz bulb containing the trapping solution. The iodine vapours
216
SESSION 11
100000
1000
100
10
0.01 0.1 1 10
TeO2 target thickness/diameter ratio
are carried through the apparatus via sterile filtered air. The quartz apparatus is
shown in Fig. 7.
The quartz oven is integrated into a GMP compliant module for carrying
out the whole procedure of radioiodine separation. The scheme of the module
is presented in Fig. 8. The operation of the module starts with loading the target
coin onto the quartz target holder and closing the quartz apparatus. The rest of
the process is carried out fully automatically, controlled by an industrial PLC
based control system and monitored on a notebook computer.
°C
heater thermocouple
air in
217
ČOMOR et al.
heater
air in
target holder
V6
F1 F2 Quartz furnace F5
V11 V10 V7
Air in Target
V9
B6 Waste V12
C1
T F3 F4
B1 B2 B3 B4 B5 V8
R1
N1 N2 P1
D1
V1 V2 V3 V4 V5
B7 Product Air out
FIG. 8. Technological scheme of TERIMO: (F1–F4) sterile filters; (F5) iodine trap; (B1–
B6) process reservoirs; (B7) sterile vial for the product; (N1, N2) sterile needles; (V1–V11)
solenoid operated valves; (V12) air flow regulator; (P1) vacuum pump; (R1) trapping
vessel; (C1) programmable temperature controller; and (D1) radioactivity detector.
218
SESSION 11
trap the iodine that is still adsorbed at different surfaces inside the furnace and
on the alumina filter that retains TeO2 traces. The trapping solution containing
the radioiodine in the form of iodide is then transferred to the sterile product vial
passing through a 0.2 μm sterile filter. Finally, the quartz bulb is washed twice
with the content of the reservoirs B4 and B5 in order to prepare it for the next run.
The operation of all of the elements of the module are logged into a file
that can be visualized on the screen of the computer (see Fig. 9) and it can be
printed out as well and attached to the production protocol.
Instead of an alkaline iodine trapping solution, one may load the quartz
bulb with a solution containing certain precursors. This way, one may directly
produce certain radiopharmaceuticals during the release of the radioiodine
from the target. This is possible owing to the fact that the radioiodine carried by
the air from the hot target to the quartz bulb is in the form of a highly reactive
iodine radical (I*) with clear electrophylic properties. The possibility of
producing o-iodo-hippuric acid and m-iodo-benzylguanidine directly during
the thermochromatographic process has already been reported [16].
FIG. 9. Synoptic view of the graphical user interface running on a notebook computer
while the PLC controls the operation of the module.
219
ČOMOR et al.
4. CONCLUSION
ACKNOWLEDGEMENTS
220
SESSION 11
REFERENCES
221
ČOMOR et al.
[17] KUDELIN, B.K., GROMOVA, E.A., GAVRILINA, L.V., SOLIN, L.M., Purifi-
cation of recovered tellurium dioxide for re-use in iodine radioisotope produc-
tion, Appl. Radiat. Isot. 54 (2001) 383.
[18] ČOMOR, J.J., et al., “COSTIS: Compact Solid Target Irradiation System”,
Proceedings of the Ninth International Workshop on Targetry and Target Chem-
istry, Turku, Finland, May 23-25 (2002) 46.
[19] ČOMOR, J.J., STEVANOVIĆ, Ž., RAJČEVIĆ, M., KOŠUTIĆ, Đ., “Modeling of
thermal properties of a TeO2 target for radioiodine production”, Nucl. Instrum.
Meth. A 521 (2004) 161.
[20] GLASER, M., et al., Improved targetry and production of iodine-124 for PET
studies, Radiochim. Acta 92 (2004) 951.
[21] YLIMAKI, R.J., KISELEV, M.Y., ČOMOR, J.J., BEYER, G.-J., “Development of
Target Delivery and Recovery System for Commercial Production of High Purity
Iodine-124”, Proceedings of the Tenth International Workshop on Targetry and
Target Chemistry, Madison, Wisconsin, USA, August 13-15 (2004) 40.
[22] INTERNATIONAL ATOMIC ENERGY AGENCY, Charged Particle Cross-
section Database for Medical Radioisotope Production: Diagnostic Radioiso-
topes and Monitor Reactions, IAEA-TECDOC-1211, IAEA, Vienna (2001).
[23] TIKHOMIROV, A.V., “Enriched stable isotopes for 123I production: Pathways
and prospects”, J. Radioanal. Nucl. Chem. 257 (2003) 157.
[24] KUDELIN, B.K., SOLIN, L.M., JAKOVLEV, V.A., “Experience in iodine-123
production via p,n reaction. Synthesis and Applications of Isotopically Labeled
Compounds”, Vol. 7, Proc. 7th Intern. Symp., Dresden, Germany 18-22 June 2000,
PLEISS, U., VOGES, R. (Eds), J.Wiley & Sons Ltd., p. 37.
222
NEW DEVELOPMENT IN RADIOIODINATED
RADIOPHARMACEUTICALS FOR SPECT AND
RADIONUCLIDE THERAPY: [123I]/[131I] LABELLED
L- AND D-PHENYLALANINE ANALOGUES
M. BAUWENS, J.J.R. MERTENS, T. LAHOUTTE, K. KERSEMANS,
C. GALLEZ, A. BOSSUYT
ICMID/BEFY, Vrije Universiteit Brussel,
Brussels, Belgium
Email: jjmerten@vub.ac.be
Abstract
223
BAUWENS et al.
1. INTRODUCTION
224
SESSION 11
excitatory amino acids. Initially, the amino acid transporters were classified on
the basis of their functional and pharmacological characteristics. They differ in
substrate selectivity and Na+ dependency of the transport, and also in tissue
expression and subcellular localization according to their specific role.
Furthermore, the physiological role type can also vary according to the cell or
tissue where it is expressed.
The L transporter is a major nutrient transport system responsible for
Na+ independent transport of large neutral amino acids including synthetic
amino acids by an obligatory exchange mechanism coupled to an anti-port
system [6]. The heterodimeric LAT transport system contains 2 subunits, a
heavy chain 4F2/CD98 and a light chain named LAT1 or LAT2.
LAT1 expression was scarcely detected in non-tumour areas [7–9] but
highly expressed (up-regulated) in proliferating tissues, in particular malignant
tumours, as it plays a critical role in cell growth and proliferation [9]. A
remarkable characteristic of system L and the hLAT-1 amino acid uptake is its
broad substrate selectivity, which enables the transport system to accept amino
acid related compounds, such as D-amino acids and cancer drugs like
Melphalan [10]. LAT2 on the other hand has a high level of expression in the
small intestine, kidney, placenta and brain and in the epithelia and blood–tissue
barriers [11]. It transports all of the isomers of neutral alpha amino acids by
facilitated diffusion; however it does not transport D-amino acids. Previously, it
was supposed that the amino acid tracers had to be incorporated into the
tumour cell proteins [21]. More recently, Langen et al. [22] and Lahoutte et al.
[24] showed that it is the increased L mediated transport of amino acid
analogues into tumour cells and not necessarily the incorporation that is
needed for efficient tumour imaging and follow-up. Langen et al. [25],
moreover, have demonstrated that the expression of L transporters and related
reversible uptake depend on the proliferation rate of human glioma cells. The
authors’ group recently developed 2-I-L-tyrosine [31], which shows a very high
tumour selectivity, better than [18F]-FDG which is taken up considerably in the
brain and inflammatory tissue, and is not retained in the kidney as shown by
Tamemasa, Goto et al. and Takeda et al. [32–34]. They showed uptake of [14C]
labelled D-Leucine, D-Alanine and D-Tryptophane in mice tumour and human
tumour derived tumour cells induced in nude mice, without being able to
explain this phenomenon at that time. They suggested that D-Leucine was
transported by the same transporter as L-Leucine, but that the isomers were
supposed to bind to different parts of the transporter. This transporter was later
defined as the L transporter. In 1985, Meyer et al. [1] showed that both the L
and D forms of the [11C]-methionine could accumulate in human brain
tumours. Since then all the development of, and studies with, radioactively
225
BAUWENS et al.
labelled amino acids for tumour diagnosis with PET and SPECT were focused
on the L-enantiomeric form.
The knowledge of the molecular biology of the LAT transporter system
drove the authors to explore the D-analogues of tyrosine and phenylalanine,
having also noticed that [14C]-D-phenylalanine and D-phenylalanine were
taken up in R1M cells by the LAT1 transport system [35]. This paper describes
the evaluation of two new tracers L- and D-[123I]-2-I-phenylalanine in R1M
tumour bearing rats by scintigraphic imaging.
2. EXPERIMENTAL
2.1.2. [99mTc]-Pyrophosphate
226
SESSION 11
All in vitro experiments were carried out in 6-well plates (VWR), using at
least three wells for each data point. Cells were counted by means of a Bürker
counting chamber. Influx and efflux were studied both in a Na+ containing
buffer (HEPES+: pH7.4; 100mM NaCl (VWR), 2mM KCl (Sigma), 1mM
MgCl2 (VWR), 1mM CaCl2 (VWR), 10mM Hepes (Sigma), 5mM Tris (VWR),
1 g/L glucose (VWR) and 1 g/L bovine serum albumin (Sigma)), a Na+ free
buffer (HEPES-: pH7.4; 100mM choline-Cl (Sigma), 2mM KCl, 1mM MgCl2,
1mM CaCl2, 10mM Hepes, 5mM Tris, 1 g/L glucose and 1 g/L bovine serum
227
BAUWENS et al.
228
SESSION 11
Water and food was ad libidum during the experimental period. For the
tumour model, male Wag/Rij rats (n = 6) (Bioservices, Netherlands) were
injected subcutaneously in the right flank (armpit region) with 15 × 106 R1M
rhabdomyosarcoma cells.
Imaging experiments with [123I]-2-Iodo-D-phenylalanine, [123I]-2-Iodo-L-
phenylalanine and [99mTc]-PP were performed 6 h p.i. of the R1M cells.
During all imaging experiments, the animals were anaesthetized intra-
peritoneal with 350 µL (21 mg) of a solution containing 60 mg pentobarbital
per mL (Nembutal, 60 mg/mL, Ceva Santé Animale, Belgium). For the biodis-
tribution experiments by dissection, the animals were killed without sedation
by cervical dislocation and the organs of interest were dissected.
All tracers were injected intravenously into the penis vein. The study
protocol was approved by the ethical committee for animal studies at the
authors’ institution. Guidelines of the National Institute of Health principles of
laboratory animal care (NIH publication 86-23, revised 1985) were followed.
229
BAUWENS et al.
3. RESULTS
230
SESSION 11
compound was stored at 4°C. HPLC analysis during the long duration of the
experiments revealed the quality of the preparation to be stable for at least a
month. After that period, a small amount of free radioiodide was observed
(2–3%). This was quantitatively removed by filtering the solution through a
sterile 0.22 µm Ag membrane filter.
The in vitro evaluation of 2-I-L-Phe has been described in detail in Ref. [27].
FIG. 1. [125]-2-I-D-Phe and [125]-2-I-L-Phe uptake time kinetics in R1M cells in MEM
buffer: mean ± SD (n = 9).
231
BAUWENS et al.
3,50E+09
L-Phe
3,00E+09
0.1 mM 2-I-L-Phe as inhibitor
1/mass uptake L-Phe (1/mol)
2,50E+09
0.1 mM 2-I-D-Phe as inhibitor
2,00E+09
1,50E+09
1,00E+09
5,00E+08
0,00E+00
The PCR coupled gel electrophoresis of the R1M cell line is depicted in
Fig. 3 and clearly shows the presence of potentially functional LAT1 and LAT2
transport systems proven by the presence of the bands for hLAT1, hLAT2 and
light chain 4F2hc. The actin band is used for positive control.
FIG. 3. Gel electrophoresis of LAT1 light chain (LAT1), LAT2 light chain (LAT2),
CD98 heavy chain (CD98hc) and actine (Actine).
232
SESSION 11
HPLC analysis of the blood plasma showed that within the first 2 h p.i. of
123
[ I]-2-I-D-Phe, neither metabolites nor free radioiodide could be significantly
detected. For [123I]-2-I-L-Phe in the same conditions, about 4% free
radioiodide and no metabolites were found.
3.3.2. DPI
233
BAUWENS et al.
FIG. 4. Composite image (20–30 min) of a R1M rhabdomyosarcoma tumour bearing rat
p.i. of 18.5 MBq [123I]-2-I-D-Phe (left) and [123I]-2-I-L-Phe (right). Image: gray scale
gamma 1.0, the scale is set to the maximum in the tumour.
Figure 6 shows that the injection of 200 µL of a 20mM L-Phe solution (2.2
mg L-Phe per kg at 60 min) displaces about 25% of [123I]-2-I-D-phenylalanine
activity from the tumour. A re-accumulation occurs after displacement as
shown by the positive slope up from 65 min.
[123I]-2-Iodo-D- [123I]-2-Iodo-L-
DUR value
phenylalanine phenylalanine
234
SESSION 11
4,0
3,5
[123I]-2-I-L-Phe
3,0
[123I]-2-I-D-
2,5
Phe
DUR
2,0
1,5
1,0
0,5
0,0
0 5 10 15 20 25 30 35 40 45 50
time (min)
3.3.2.2. Clearance
3,5
2,5
2
DUR
1,5
0,5
0
0 10 20 30 40 50 60 70 80 90
tim e (m in)
235
BAUWENS et al.
8
D - Bladder D - Kidney D - Heart
5
DUR
0
0 5 10 15 20 25 30 35 40 45
time (min)
bladder as a function of time is much higher for the D-isomer than for the L-
isomer (the slope of the curve at 5–10 min is 0.27 DUR values/min for the D-
isomer and 0.096 DUR values/min for the L-isomer), resulting in a twofold
amount of activity in the bladder for the D analogue compared with 2-I-L-Phe
at the end of imaging.
4. DISCUSSION
236
SESSION 11
FIG. 8. Composite images of a R1M rhabdomyosarcoma tumour bearing rat p.i. of 18.5
MBq [123I]-2-I-D-Phe (left) and [123I]-2-I-L-Phe (right) at 48 h p.i. Image: gray scale
gamma 1.0, the scale is set to the maximum in the tumour.
237
BAUWENS et al.
5. CONCLUSION
238
SESSION 11
ACKNOWLEDGEMENTS
The authors thank the FWO Vlaanderen and GOA-VUB for their
financial support.
REFERENCES
[1] MEYER, G.J., SCHOBER, O., HUNDESHAGEN, H., “Uptake of 11C-L- and
D-methionine in brain tumours Eur. J. Nucl Med (1985);10(7-8):373-6.
[2] OHKAME, H., MASUDA, H., ISHII, Y., KANAI, Y., Expression of L-type
amino acid transporter 1 (LAT1) and 4F2 heavy chain in liver tumour lesions of
rat models, Journal of Surgical oncology, (2001), Vol. 78, p. 265-272.
[3] MEIER, C., RISTIC, Z., KLAUSER, S., VERREY, F., Activation of system L
heterodimeric amino acid exchangers by intracellular substrates, The EMBO
Journal, Vol. 21 Nr. 4, (2002), p. 580-589.
[4] LAHOUTTE, T., et al., SPECT and PET amino acid tracer influx via system L
(h4F2hc-hLAT1) and its transstimulation, J Nucl Med. (2004), Vol 45 N° 9,
p.1591-1596.
[5] YANAGIDA, O., KANAI, Y., Human L-Type amino acid transport system 1
(LAT1) : characterisation of function and expression in tumour cell lines, Biochim
Biophys Acta, Vol. 1514, (2001), p. 291-302.
[6] SEGAWA, H., et al., Identification and functional characterisation of a Na+-
independent neutral L-amino acid transporter with broad substrate selectivity,
The Journal of Biological Chemistry, Vol. 274, (1999), p. 19745-19751.
[7] KANAI, Y., et al., “Expression cloning and characterisation of a transporters for
large neutral amino acids activated by heavy chain 4F2 antigen (CD98)” J. Biol.
Chem. (1998) 273,23629-23632.
[8] CAMPBELL, W.A., THOMPSON, N.L., “ Overexpression of LAT1/CD98 light
chain is sufficient to increase system L-amino acid transport activity in mouse
hepatocytes but not fibroblasts J. Biol. Chem (2001) 276,16877-16884.
[9] KIM DO, K., et al., “ Characterization of the L amino acid transporter in T24
human bladder carcinoma cells Biochem Biophys Acta (2002) Sep 20; 1565(1):
112.
[10] YANAGIDA, O., KANAI, Y., Human L-Type amino acid transport system 1
(LAT1) :characterisation of function and expression in tumour cell lines, Biochim
Biophys Acta, Vol. 1514, (2001), p. 291-302.
[11] RAJAN, D.P., et al., Cloning and functional characterisation of a Na(+)-inde-
pendent, broad-specific neutral amino acid transporter from mammalian intes-
tine, Biochim BioPhys Acta 2000, Jan 15, 1463(1):6-14.
[12] BERGSTROM, M., et al., The normal pipituary examined with positron emission
tomography and (methyl-11C)-L-Methionine and (methyl-11C)-D-Methionine,
Neuroradiol. (1987), 29, 221-225.
239
BAUWENS et al.
[13] COENEN, H.H., KLING, P., STÖCKLIN, G., “Cerebral metabolism of L-2-18F-
fluorotyrosine, a new PET tracer for protein synthesis” J. Nucl. Med., (1989),
30:1376.
[14] BIERSACK, H.J., et al., “Imaging of brain tumours with L-3-123I-iodo-alpha-
methyltyrosine and SPECT” J. Nucl. Med. (1989), 30:110.
[15] JAGER, P.L., DE VRIES, E.G., PIERS, D.A., TIMMER-BOSSCHA, H.,
“Uptake mechanism of L-3-125I-iodo-alpha-methyl-tyrosine and SPECT” Nucl.
Med. Comm. (2001), 22(1):87.
[16] HEISS, P., et al., Investigation of transport mechanism and uptake kinetics of O-
2- [18F]fluoroethyl-L-tyrosine in vitro and in vivo” J. Nucl Med (1999), 40:1367.
[17] VEKEMAN, M., JOOS, C., MERTENS, J., “L-[2-radioiodo]Tyrosine, a new
potential protein synthesis and tumour tracer for SPECT: radiosynthesis and
biodistribution in rodent”. Eur. J. Nucl. Med., 26(9) (1999), 971.
[18] MERTENS, J., et al., “Uptake study of L-2-125I-o-tyrosine, a new tracer in human
colon carcinoma WiDr cells”. Eur. J. Nucl. Med. (2000)27:1063.
[19] MERTENS, J., et al., “In Vitro and in Vivo Evaluation of L-2-radioiodo-tyrosine
as a new potential tumour tracer for SPECT” J. Labelled Cmp Radiopharm
(2001).
[20] LAHOUTTE, T., et al., “Biodistribution of iodinated amino acids: Selection of
the optimal analog for oncologic imaging outside the brain”. J. Nucl Med (2002).
[21] RAU, F.C., et al., O-(2-[(18)F]fluoroethyl)-L-Tyrosine (FET): a tracer for differ-
entiation of tumour from inflammation in murine lymph nodes, Eur J Nucl Med,
(2002), Vol. 29, p. 1039-1046.
[22] LANGEN, K.J., PAULEIT, D., COENEN, H.H., “3-123I-Iodod-alpha-methyl-L-
tyrosine:uptake mechanisms and clinical applications. Nucl Med Biol 29(2002)
625-631.
[23] LAHOUTTE, T., et al., In vitro characterisation of the influx of 3-[125I]-iodo-L-
Tyrosine and 2-[125I]-iodo-L-Tyrosine into U266 human myeloma cells : Evidence
for system T transport, Nucl Med Biol , Vol 28, Issue 2, (2001), p. 129-134.
[24] LAHOUTTE, T., et al., Increase tumour uptake of 3-[123I]-Iodo-L-alpha-methyl-
tyrosine after preloading with amino acids, an in vivo animal imaging study, J Nucl
Med, Vol 43(9), (2002), p. 1201-1206.
[25] LANGEN, K.J., et al., 3-[123I]Iodo-alpha-methyltyrosine and [methyl-11C]-
methionine uptake in cerebral gliomas: a comparative study using SPECT and
PET, J Nucl Med, (1997), Vol. 38, p. 517-522.
[26] LAHOUTTE, T., et al., Comparative biodistribution of iodinated amino acids in
rats: selection of the optimal analog for oncologic imaging outside the brain, Nucl
Med. (2003), Vol 44 N° 9, 1489-1494.
[27] MERTENS, J., et al., Synthesis, radiosynthesis and in vitro characterisation of
[125I]-2-iodo-L-phenylalanine in a R1M rhabdomyosarcoma cell model as a new
potential tumour tracer for SPECT, Nucl Med Biol, Vol 31, (2004), p. 739-746.
[28] MERTENS, J., et al., “In Vitro and in Vivo Evaluation of L-2-radioiodo-tyrosine
as a new potential tumour tracer for SPECT” J. Labelled Cmp Radiopharm
(2001).
240
SESSION 11
241
.
COMPARISON OF 131I-TYR3-OCTREOTATE AND 131I-
DOTA-TYR3-OCTREOTATE: THE EFFECT OF DOTA ON
PHARMACOKINETICS AND STABILITY
Abstract
The authors compared the biodistribution, and in vivo and in vitro stabilities of
131
I-Tyr3-octreotate and 131I-DOTA-Tyr3-octreotate. The peptides were radioiodinated
by the chloramine T method and high radiochemical yields were obtained (greater than
97%). Both labelled compounds showed high stability when incubated in human plasma
at 37°C. The 131I-Tyr3-octreotate showed significant hepatic uptake and biliary excretion.
The biodistribution of 131I-DOTA-Tyr3-octreotate, however, can be compared with the
distribution of radiometal labelled octreotide analogues.
1. INTRODUCTION
243
DE ARAÚJO et al.
positron emission tomography (18F) and peptide receptor therapy (131I, 125I and
211
At). Consequently, 123I labelled Tyr3-octreotide was the first compound to be
used for the imaging of somatostatine receptor positive tumours [6].
However, the experience gained with radioiodinated sst ligands showed
that the diagnostic and therapeutic usefulness of these ligands was limited by
their unfavourable biokinetics, in vivo deiodination and resulting dosimetry.
Owing to fast hepatic uptake and biliary clearance, most of the tracers showed
high abdominal background acitivity and fast blood clearance, leading to low
tumour uptake. Additionally, they exhibited low tumour retention, which was
often attributed to fast intracellular degradation of the tracers and subsequent
extracellularization [7].
In this paper, the authors prepared radioiodinated octreotates, 131I-Tyr3-
octreotate and 131I-DOTA-Tyr3-octreotate, with high radiochemical yield.
Although the DOTA chelating group was not necessary to the radioiodination
procedure, the authors evaluated the influence of the chelating group on
biodistribution, particularly on hepatic uptake, biliary excretion and renal
clearance. Tumour uptake was evaluated in nude mice bearing AR42J tumour.
2.1. Reagents
2.2. Radiolabelling
244
SESSION 11
3.1. Radioiodination
245
DE ARAÚJO et al.
produced only one radiochemical species with a retention time of 22.73 min
(Fig.1). When using a molar peptide to radionuclide ratio of 0.54 (37 MBq
131
I/μg peptide), a second radiochemical species can be observed in the
HPLC profile (Fig. 2), with a retention time of 24.9 min, probably related to
the di-iodinated form of the peptide.
246
SESSION 11
In vitro studies show clearly the high level of stability of the iodinated
ligands even after 24 h of incubation in human plasma (Table 1).
247
TABLE 2. BIODISTRIBUTION OF 131I-Tyr3-OCTREOTATE AND 131I-DOTA-Tyr3-OCTREOTATE IN NORMAL
248
SWISS MICE
131 131
I-Tyr3-octreotate I-DOTA-Tyr3-octreotate
Tissue Dose/g (%) Dose/g (%)
1h 4h 24 h 1h 4h 24 h
Total blood 3.17 ± 0.32 0.81 ± 0.12 0.103 ± 0.006 2.56 ± 0.27 1.19 ± 0.13 0.020 ± 0.004
Liver 2.28 ± 0.78 0.42 ± 0.06 0.161 ± 0.034 0.70 ± 0.05 0.36 ± 0.05 0.088 ± 0.018
Spleen 0.76 ± 0.25 0.37 ± 0.11 0.114 ± 0.044 0.57 ± 0.06 0.27 ± 0.04 0.057 ± 0.018
Stomach 3.51 ± 1.67 1.94 ± 1.02 0.093 ± 0.032 3.32 ± 0.45 2.02 ± 0.59 0.19 ± 0.05
Int. (small) 12.30 ± 4.36 1.33 ± 0.96 0.045 ± 0.015 1.48 ± 0.13 0.57 ± 0.12 0.41 ± 0.19
Int. (large) 0.96 ± 0.58 14.85 ± 0.83 0.103 ± 0.032 0.44 ± 0.08 2.18 ± 0.25 1.90 ± 0.82
Kidney 12.54 ± 0.51 9.77 ± 3.46 3.752 ± 1.183 12.18 ± 0.86 9.86 ± 1.00 1.60 ± 0.22
Muscle 0.47 ± 0.24 0.13 ± 0.06 0.022 ± 0.002 0.26 ± 0.02 0.13 ± 0.03 0.03 ± 0.01
DE ARAÚJO et al.
Brain 0.08 ± 0.01 0.024 ± 0.003 0.007 ± 0.001 0.08 ± 0.03 0.04 ± 0.010 0.006 ± 0.003
Heart 0.66 ± 0.11 0.20 ± 0.08 0.049 ± 0.020 0.50 ± 0.10 0.20 ± 0.05 0.019 ± 0.003
Lung 1.59 ± 0.40 0.45 ± 0.14 0.013 ± 0.021 0.80 ± 0.42 0.49 ± 0.22 0.052 ± 0.005
Thyroid * 0.49 ± 0.07 1.04 ± 0.20 0.878 ± 0.304 0.55 ± 0.10 1.23 ± 0.17 0.29 ± 0.06
Adrenals* 0.020 ± 0.007 0.007 ± 0.004 0.002 ± 0.0002 0.012 ± 0.003 0.008 ± 0.001 0.0014 ± 0.0005
Pancreas 0.87 ± 0.17 0.23 ± 0.05 0.023 ± 0.008 1.11 ± 0.52 0.79 ± 0.12 0.030 ± 0.010
* % dose organ; values are mean ± SD (n = 6).
SESSION 11
Tracer uptake in the pancreas, tumour and adrenal were similar for both
compounds (p = 0.01) after 1 h p.i. (Table 3). Tumour to blood ratios at 1 h p.i.
were similar for both compounds but tumour to liver and tumour to intestine
ratios were superior to 131I-DOTA-Tyr3-octreotate (Table 4).
Both labelled peptides presented low uptake in thyroid, which suggests
low in vivo dehalogenation of the compounds.
The distribution pattern of 131I-Tyr3-octreotate was similar to that
reported for 123I-Tyr3-octreotide in rats [9], with relatively high activity levels in
liver and intestine. The biodistribution of 131I-DOTA-Tyr3-octreotate, however,
can be compared with the distribution of radiometal labelled octreotide
analogues in mice [10, 11].
Total blood 2.12 ± 0.49 0.085 ± 0.035 2.93 ± 0.32 0.124 ± 0.006
Liver 2.07 ± 0.73 0.17 ± 0.03 1.34 ± 0.09 0.149 ± 0.009
Int. (small) 8.75 ± 1.87 0.061 ± 0.001 2.78 ± 0.60 0.067 ± 0.022
Muscle 0.37 ± 0.12 0.024 ± 0.009 0.42 ± 0.21 0.039 ± 0.013
Thyroid * 0.28 ± 0.13 0.516 ± 0.031 0.54 ± 0.17 1.19 ± 0.31
Adrenals* 0.021 ± 0.010 0.002 ± 0.001 0.018 ± 0.004 0.003 ± 0.001
Pancreas 0.78 ± 0.05 0.038 ± 0.001 1.15 ± 0.29 0.047 ± 0.013
Tumour 1.10 ± 0.45 0.18 ± 0.08 1.73 ± 0.01 0.13 ± 0.01
* % dose organ; values are mean ± SD (n = 3).
249
DE ARAÚJO et al.
ACKNOWLEDGEMENTS
REFERENCES
250
SESSION 11
[8] BAKKER, W.H., et al., Iodine-131 labelled octreotide:not an option for somato-
statin receptor therapy, Eur. J. Nucl. Med., 23 (1996) 775-781.
[9] BAKKER, W.H., et al., Receptor scintigraphy with a radioiodinated somatostatin
analogue: Radiolabeling, purification, biologic activity, and in vivo application in
animals, J. Nucl. Med., 31 (1990) 1501-1509.
[10] AKIZAWA, H., et al., Renal metabolism of 111In-DTPA-D-Phe1-octreotide in
vivo, Bioconjugate Chem., 9 (1998) 662-670.
[11] LEWIS, J.S., et al., In vitro and in vivo evaluation of 64Cu-TETA-Tyr3-octreotate.
A new somatostatin analog with improved target tissue uptake, Nucl. Med. Biol.,
26 (1999) 267-273.
251
.
IODINE LABELLED DIETHYLSTILBESTROL (DES)
OF HIGH SPECIFIC ACTIVITY: A POTENTIAL
RADIOPHARMACEUTICAL FOR THERAPY OF
ESTROGEN RECEPTOR POSITIVE TUMOURS AND
THEIR METASTASES?
T. FISCHER, H. SCHICHA, K. SCHOMÄCKER
Department of Nuclear Medicine,
University of Cologne,
Germany
Email: thomas.fischer@uk-koeln.de
Abstract
253
FISCHER et al.
1. INTRODUCTION
254
SESSION 11
well as Dulbecco’s Medium without Phenol Red. The cells’ ER status was
evaluated with the ER-ICA test from ABBOTT.
For investigation of cell viability, the cells were incubated with a
tetrazolium salt (cell proliferation reagent WST-1, Roche Diagnostics), which
was cleaved by succinat tetrazolium reductase, an enzyme of the respiratory
chain in the membrane of mitochondria. The reaction product was a coloured
formazan, which could be measured by optical methods. For the experiments,
100 µL Dulbecco’s Modified Eagle Medium containing 1 × 104 MCF-7-cells was
transferred to each well of a microtitre plate (MTP) with a flat bottom. One
row was kept empty. The cells were incubated in the MTP for 24 h at 37°C and
under the above mentioned atmospheric conditions. Then to each well, 100 µL
*
I-DES or *I-iodide of different concentrations between 0.1 MBq/mL and 5
MBq/mL was added. One row of eight wells was only filled with 100 µL of
medium for control. The row that was kept without cells was now filled with
200 µL medium in each well. All set-ups were made fourfold to eightfold. The
cells were now incubated for 18 h under the mentioned conditions and then
mixed with 20 µL WST-1-reagent. After 2 h incubation at 37°C, the extinction
of the samples was determined with the ELISA reader at 450 nm (reference
filter: 620 nm).
For determination of apoptosis or necrosis, the Cell Death Detection
ELISA from Roche Diagnostics was used. The amount of apoptotic cells in
comparison to a control group can be measured by detection of DNA
fragments by special antibodies included in the kit. According to whether
detection of those fragments occurs before or after cell lysis, it will be possible
to distinguish between necrosis and apoptosis. Measured DNA fragments in
the supernatant before cell lysis were due to apoptosis, detected DNA
fragments after lysis indicated necrosis. In the experiment, 100 µL DMEM
containing 1 × 104 MCF-7 cells was transferred to each well of a flat bottomed
MTP. The cells were incubated in the MTP for 24 h at 37°C and under the
above mentioned atmospheric conditions. Samples, that should be tested in the
presence of vitamin C, were mixed with 20 µL of ascorbic acid solution (0.01
mg/mL or 0.1 mg/mL giving 0.001 mg/mL or 0.01 mg/mL in the well). Then, to
each well (in the presence or absence of vitamin C) 100 µL *I-DES or *I-iodide
in concentrations of between 0.1 and 5 MBq/mL was added. Eight wells were
filled with 100 µL of medium instead of radioactive material. All other samples
were carried out fourfold to eightfold.
The MTPs were incubated for 18 h under the usual conditions and then
centrifuged at 200g. A 20 µL sample of each liquid was transferred for
measurement of the amount of necrosis to another streptavidine coated MTP
from the Cell Death Detection ELISAPLUS kit. Now, the residual liquid in the
first MTP was carefully removed and 200 µL of lysis buffer placed instead in
255
FISCHER et al.
every well. After 30 min incubation at room temperature, the plate was
centrifuged again at 200g and then 20 µL of the liquid was transferred to a
second streptavidine coated MTP for determination of apoptosis. For
background measurement, some wells were filled with 20 µL of incubation
buffer. In addition, some positive controls were also carried out.
Animal experiments were carried out with male tumour bearing DBA/2
mice (Charles River). Three groups of animals (n = 5 in each group) were
injected with: (a) 1.5 MBq 123I-DES, (b) 7 µmol DES-2-P + 2 MBq 123I-DES, (c)
7 µmol Estradiol + 2 MBq 123I-DES. Three hours after IV injection, the animals
were sacrificed and organs, tissues and blood were measured in a well counter.
3. RESULTS
120
I-123-DES
I-123
100
reductase activity in % of control group
80
60
40
20
0
0 0,5 1 1,5 2 2,5 3 3,5 4 4,5 5
activity concentration [MBq/ml]
FIG. 1. Relative metabolic succinat tetrazolium reductase activity in MCF-7 cells after 18 h
incubation with 123I-DES or 123I- in per cent enzyme activity in control group not exposed
to radioactivity (n = 8 per point, SD = ±15–20%).
256
SESSION 11
120
I-125-DES
I-125
reductase activity in % of control group 100
80
60
40
20
0
0 0,5 1 1,5 2 2,5 3 3,5 4 4,5 5
activity concentration [MBq/ml]
FIG. 2. Relative metabolic succinat tetrazolium reductase activity in MCF-7 cells after
18 h incubation with 125I-DES or 125I- in percent enzyme activity in control group not
exposed to radioactivity (n = 8 per point, SD = ±15 – 20%).
120
I-131-DES
I-131
100
reductase activity in % of control group
80
60
40
20
0
0 0,5 1 1,5 2 2,5 3 3,5 4 4,5 5
activity concentration [MBq/ml]
FIG. 3. Relative metabolic succinat tetrazolium reductase activity in MCF-7 cells after 18 h
incubation with 131I-DES or 131I- in per cent enzyme activity in control group not exposed to
radioactivity (n = 8 per point, SD = ±15–20%).
257
FISCHER et al.
40
35
30
enrichment factor
25
20
15
10
0
0 0,1 0,2 0,3 0,4 0,5 0,6 0,7 0,8 0,9 1
activity concentration [MBq/ml]
I-123-DES I-123-Iodid
I-123-DES+0,001 mg/ml Vitamin C I-123-DES+0,01 mg/ml Vitamin C
I-123-Iodid I-123-DES+0,001 mg/ml Vitamin C
I-123-DES+0,01 mg/ml Vitamin C
FIG. 4. Apoptosis accumulation factors of MCF-7 cells incubated with different radio-
activity concentrations of 123I-DES or 123I-iodide in comparison to cells of an untreated
control group (n = 4–8 per point, incubation time: 18 h, SD = ±15%).
258
SESSION 11
12
10
enrichment factor 8
0
0 0,1 0,2 0,3 0,4 0,5 0,6 0,7 0,8 0,9 1
activity concentration [MBq/ml]
I-125-DES I-125-Iodid
I-125-DES+0,001 mg/ml Vitamin C I-125-DES+0,01 mg/ml Vitamin C
I-125-Iodid I-125-DES+0,001 mg/ml Vitamin C
I-125-DES+0,01 mg/ml Vitamin C
FIG. 5. Apoptosis accumulation factors of MCF-7 cells incubated with different radio-
activity concentrations of 125I-DES or 125I-iodide in comparison to cells of an untreated
control group (n = 4–8 per point, incubation time: 18 h, SD = ±15%).
4
enrichment factor
0
0 0,1 0,2 0,3 0,4 0,5 0,6 0,7 0,8 0,9 1
activity concentration [Bq/ml]
I-131-DES I-131-Iodid
I-131-DES+0,001 mg/ml Vitamin C I-131-DES+0,01 mg/ml Vitamin C
I-131-Iodid I-131-DES+0,001 mg/ml Vitamin C
I-131-DES+0,01 mg/ml Vitamin C
FIG. 6. Apoptosis accumulation factors of MCF-7 cells incubated with different radio-
activity concentrations of 131I-DES or 131I-iodide in comparison to cells of an untreated
control group (n = 4–8 per point, incubation time: 18 h, SD = ±15%).
259
FISCHER et al.
120
Blood
100 Liver
Spleen
80 Kidney
Muscle
60 Femur
Thyroid
40 Prostate
Intestines
20
Tumor
Urine
0
I-123-DES I-123-DES + I-123-DES + E
HON.
FIG. 7. Biodistribution (in %ID/g) of 123I-DES in tumour bearing male mice with and
without co-application of HONVAN™ (DES-2P) or estradiol.
4. CONCLUSIONS
REFERENCES
260
SESSION 11
261
FISCHER et al.
[27] TUBIS, M., et al., The distribution of 131I labeled diethylstilbestrol diphosphate in
animals and men with observations on its potential use as a scanning agent of the
prostate, Nucl. Med. 5 (1967) 184.
[28] MENDE, T., et al., Experimentelle Untersuchungen über die Verteilung von
intravenös injiziertem jodmarkiertem Cytonal (Diäthylstilböstroldiphosphat), Z.
Urol. u. Nephrol. 71 (1978) 529.
[29] MENDE, T., et al., Verteilung von Jod-markiertem Diäthylstilböstroldiphosphat
nach intravenöser Injektion, Gynäk. Rdsch. 19 (1979) 30.
[30] MELLER-REHBEIN, B., et al., 123I-Diethystilbestrolphophate-a potential tracer
for receptorimaging and therapy ? Eur. J. Nucl. Med. 21 (1994) 878 (abstract).
[31] SCHOMÄCKER, K., et al., “Investigations into biokinetics of I-123 Diethyl-
stilbestrol phosphate in tumour-bearing mice”, Radioactive Isotopes in Clinical
Medicine and Research (Bergmann H, Sinzinger H, eds). Birkhäuser Verlag,
Basel (1997) 333-342.
[32] SCHOMÄCKER, K., et al., Biokinetics of I-123-Diethylstilbestrol (DES) in
mamma-carcinoma bearing mice. J. Nucl. Med. 40 (1999) 100P (abstract).
[33] FISCHER, T., et al., Labelling, purification, and receptor affinity of radioactive
iododiethylstilbestrol (*I-DES) with high specific activity and first structure
analysis with natI-DES, J. Labelled Compd. Radiopharm. 47 (2004) 669.
262
18
F RADIOPHARMACEUTICALS
AND AUTOMATION OF SYNTHESIS
(Session 12)
Chairpersons
V.W. PIKE
United States of America
D. SOLOVIEV
Switzerland
.
18
F BASED RADIOPHARMACEUTICALS AND
AUTOMATION OF SYNTHESIS
New 18F radiopharmaceuticals
Abstract
Fluorine-18 is one of the most commonly used positron emitting isotopes for
clinical and research needs with a physical half-life of 110 min. PET isotopes deposit
higher radiation absorbed dose than nuclear medicine isotopes. Because of their rela-
tively short half-life, larger quantities of these isotopes are used at the start of synthesis.
Therefore, increased shielding and remote automated synthesis are essential for their
safe handling. Unlike other radiopharmaceuticals, it is not practical to produce PET
radiopharmaceuticals at a central location for subsequent distribution to clinical and
research facilities around the country. This limitation compels various academic and
research facilities to manufacture their own PET radiopharmaceuticals for in-house use.
For multiple reasons, 18F fluorodeoxyglucose ([18F]FDG) is one of the most commonly
used radiopharmaceuticals. The synthesis of [18F]FDG has been optimized and auto-
mated, thus allowing independent laboratories to produce this radiopharmaceutical safely.
Nonetheless, these laboratories should acquire resources and expertise to fulfil ever
increasing regulatory requirements for the safe production and usage of PET radio-
pharmaceuticals. In addition to [18F]FDG, a wide array of new and novel radiotracers is
being developed to explore various biological processes. This paper emphasizes the fact
that it is possible to accomplish research and fulfil clinical needs within an academic
setting with modest resources. A careful assessment of the need for due diligence in
radiation safety issues is very important for the longevity of any PET research endeavour.
265
GARG and GARG
266
SESSION 12
applications. One of the approaches pursued was to design newer PET imaging
molecules using templates for exisiting drugs. To that end, the authors
developed 18F labelled p-fluorobenzylguanidine [1, 2], 18F labelled monoclonal
antibodies and their fragments [3, 4] and peptides [5], and 18F labelled
androgens [6] and various other neuroimaging agents The authors derived
these newer molecules from templates that have been tested and used in
clinical or preclinical studies using other imaging modalities. The other benefit
of this approach was that the biological properties of the parent analogues were
known. Therefore, successful translation of these molecules for PET imaging
was straightforward. Some of the molecules that were produced and evaluated
over the last few years are briefly described.
Meta-iodobenzylguanidine (MIBG; Fig. 1) was first developed as a
norepinephrine (NE) mimic to detect and treat neuroendocrine tumours.
Successful use of MIBG in the detection of neural crest tumours and cardiac
sympathetic innervations led the authors to develop the 18F labelled analogue,
i.e. 18F p-fluorobenzylguanidine ([18F]PFBG, Fig. 1) for PET imaging. The
structures of PFBG, MIBG and NE are shown in Fig. 1 for comparison and to
highlight the similarity in their structures.
The synthesis of [18F]PFBG and their initial in vitro and in vivo results
have already been reported [2, 7, 8]. In small animals, [18F]PFBG shows higher
specificity for the target tissues compared to that with MIBG [1, 8].
Encouraged by these studies, the authors explored the potential of [18F]PFBG
in detecting myocardial infarct in a dog model. As shown in Fig. 2, [18F]PFBG
images acquired 16 d post-infarction lack 18F accumulation in the area of
infarction despite reperfusion of the myocardium from newly formed collateral
HO H2N
N
HO HN
NH2
OH
HN
norepinephrine (NE) guanethidine
18
F
H
N NH2 H
I N NH2
NH
NH
meta-iodobenzylguanidine para-fluorobenzylguanidine
(MIBG) (PFBG)
267
GARG and GARG
FIG. 2. PET images using [18F]PFBG (innervation tracer) and 13N ammonia (perfusion
tracer) in dog heart. Left panel shows well perfused heart prior to surgery and a distinct
tracer uptake deficit 2 d post-surgery scans. The right panel shows continued [18F]PFBG
uptake deficit 16 d post-surgery.
blood vessels [9]. As shown in the left hand graphic of Fig. 2, [18F]PFBG and
13
N ammonia both showed an area of marked decrease in accumulating both
radiotracers 2 d post-surgery compared with control (pre-surgery scans). The
[18F]PFBG accumulation deficit persisted even 16 d post-surgery as shown for
two dogs in Fig. 2, right hand panel. These observations suggest that blood
vessel ligation not only leads to perfusion deficit, but may have caused damage
to the myocardium that was not detectable from blood perfusion scans, but
which was quite evident on [18F]PFBG scans.
A graphical representation of these results is shown in Fig. 3. The graph
presents radioactivity uptake in the area of infarction using 13N ammonia for
myocardial perfusion and using [18F]PFBG to assess innervation. A sharp
decrease in both the tracers’ uptake was noticed in the area of infarction when
images were acquired 2 d post-surgery: [18F]PFBG 59% of control values; 13N
ammonia 61% of control values. Although perfusion deficit resolved 16 d post-
surgery as seen from 13N images, [18F]PFBG deficit remained unchanged even
23 d post-surgery (Fig. 3). These observations suggest that the [18F]PFBG
accumulates in tissues that were damaged during infarction and the damage
existed three weeks after surgery despite recovery of blood perfusion [9, 10].
[18F]PFBG also shows promise for its applications in oncology. When
tested on dogs with spontaneous pheochromocytoma, [18F]PFBG showed a
rapid and high uptake at tumour sites. PFBG accumulation peaked within
minutes of injection [11] and remained constant for the duration of the study.
268
SESSION 12
A coronal view of the PET scan from one of the dogs with pheochromocytoma
acquired after injecting with [18F]PFBG is shown in Fig. 4. The [18F]PFBG
localized in the right adrenal gland mass within the first few minutes and
remained constant. A rapid clearance of radioactivity from the blood pool and
a persistent retention of tracer within the area of the adrenal mass were
observed from time activity curves generated from region of interest analysis.
PET images of dogs with spontaneous pheochromocytomas utilizing
[18F]PFBG showed an SUV of 20–45 [11].
The authors further evaluated [18F]PFBG utility in a diabetic rat heart
model. Diabetes was induced by injecting streptozotocine (55 mg/kg i.p.) in
male Sprague Dawley rats. One week later, blood glucose levels were
FIG. 4. [18F]PFBG accumulation in the area of adrenal mass of a dog with suspected
pheochromocytoma at 20–30 min post-injection of [18F]PFBG.
269
GARG and GARG
30 MIN
3 60 MIN
% Injected dose/g
*
2
*
0
n=5 NORMAL DIABETIC
p <0.05
FIG. 5. [18F]PFBG uptake in diabetic and normal rat hearts at 30 and 60 min p.i.
monitored and rats with glucose levels of >200 mg/dL were used for the study.
A separate group of rats with no diabetes was used as control. As shown in
Fig. 5, a higher [18F]PFBG accumulation was observed in diabetic rat hearts at
30 min followed by rapid and significant washout of activity from the heart by
60 min. In comparison, [18F]PFBG uptake and washout was less dramatic in
normal rat hearts. These observations indicate a reduced retention of
[18F]PFBG in the vesicles, perhaps due to reduced NE transporter acitivy in
diabetic rat heart. These observations are similar to those reported earlier
using MIBG in the same model [12].
One of the probable causes of failed or ineffective radiation therapy is the
presence of hypoxia in tumours. These hypoxic areas are radioresistant and
also respond poorly to chemotherapy and other therapeutic interventions.
2-nitroimidazoles have been shown to possess excellent radiosensitization
properties. These compounds are taken up by the tumours and under hypoxic
conditions they are metabolized via a 2 electron reduction pathway. The
reduced species remains trapped within the hypoxic areas. If developed, 2-
nitroimidazole compounds could help in the assessment of the tumour hypoxia
fraction. Therefore, misonidazole, a first generation radiation sensitizer was
one of the first 2-nitroimidazoles to be labelled with 18F for its use as a hypoxia
imaging agent [13, 14]. Although the high lipophilicity of this molecule causes
low target tissue to blood ratios (<1.6), imaging experiments showed promise
[15]. Encouraged from these studies, newer 18F labelled 2-nitroimidazoles are
being developed to obtain a hypoxia imaging agent with better imaging charac-
terisitics and pharmacokinetics. Some of the recently developed compounds
include 18F FAZA [16], 18F EF [17, 18] and 18F PK 110 [19]. Unfortunately, most
of these compounds continue to possess high lipophilicity, leading to slow
clearance from the blood pool and confounding image analysis.
270
SESSION 12
FIG. 6. MicroPET image of a mouse bearing HT-29 human colorectal tumour xenograft.
A 30 min frame image acquired 90–120 min after injecting with 18F pimonidazole.
271
GARG and GARG
uptake and distribution pattern of this tracer in mice and rats. As with most
steroidal compounds, including [18F]FDHT, the clearance of [18F]FMDHT
occurred via the GI track in mice and rats.
The authors further assessed the potential of [18F]FMDHT as a prostate
imaging agent using microPET. The imaging characteristics of [18F]FMDHT
were assessed using microPET imaging of an androgen dependent LnCAP
tumour xenograft in mice. As shown in Fig. 7, the tumour was clearly visible on
the microPET image. Additional studies were carried out using [18F]FMDHT
and comparing its imaging characteristics with other currently available PET
imaging agents. Figure 8 shows a head-on comparison of [18F]FMDHT images
with [18F]choline and [18F]FDG in the same mice bearing the LnCAP tumour
xenograft. As evident from these images, a better visualization of tumour was
FIG. 8. MicroPET image of a mouse bearing the LnCAP tumour xenograft. Among the
three ligands used, more intense uptake of radioactivity was evident with [18F]FMDHT
compared with [18F]choline or [18F]FDG.
272
SESSION 12
273
GARG and GARG
STRIATUM CEREBELLUM
18
FIG. 9. PET image of a babboon brain after injecting F FPCBT. An intense uptake in
striatal area and minimal to negligible uptake in cerebellum are evident.
A rapid uptake of this tracer in the brain with highest accumulation in the
striatum was observed. Despite excellent accumulation in the striatum, a
modest striatum to cerebellum ratio (St:Cb = 3.3) observed for this ligand
reduced the enthusiasm to pursue it. A large collection of 18F labelled
radiotracers exist for neuroreceptor imaging. Recently developed tracers
include 18F ACF and 18F AFM for the SERT [28], 18F reboxitine analogue to
image NET [29] and 18F Fallypride for D2 dopamine receptor imaging [30].
While 18F compounds and molecular imaging probes are being developed
at a fierce pace, their handling and usage in radiochemical synthesis requires
particular care. In general, a positron emitting isotope imparts 2–10 fold higher
radiation doses to personnel than those received from the isotopes commonly
used for nuclear medicine imaging. For example, a 1 mCi unshielded gamma
emitting nuclear medicine isotope produces an exposure of 0.2–2.2 R/h at 1 cm
from the surface (depending on gamma energies) while exposure to a 1 mCi
PET isotope at 1 cm would be ~5.8 R/h, signifying a need for more careful and
cautious handling of PET isotopes. Because of a short physical half-life, larger
quantities of these isotopes are routinely used for radiochemical synthesis, thus
further increasing the risk of higher exposure. Therefore, it is important to use
extra caution and to develop safe handling practices in the laboratories. An
automated or remote handling procedure for the delivery of the radioisotopes
and for the synthesis of PET radiopharmaceuticals is essential to minimize
personnel exposure. Although several manufacturers are developing
automated synthesis boxes, their application is mainly limited to carrying out
synthesis of ligands that are fully evaluated and closer to routine use. It is
important to highlight that a number of PET tracers with somewhat
complicated chemistries are still synthesized manually with limited or no
274
SESSION 12
automation. Once evaluated and the synthesis optimized, it is likely that the
synthesis of those molecules can be adapted to the existing modules.
There are several synthesis modules that are now commercially available
for [18F]FDG production. One can now produce multiple batches of [18F]FDG
with one set-up routine in the beginning. Although multiple radiotracers
utilizing similar chemistries can be produced using an [18F]FDG box, it would
be difficult to adapt new ligand synthesis to these modules without the help of
experienced radiochemistry personnel. Therefore, there is a growing need for
more flexible automation sytems.
Because of the different chemistries involved, it is difficult to develop an
automated synthesis module for each individual ligand. Therefore, a large
number of research groups design their own semi-automated synthesis modules
that are either operated by remote electric valves or by programmable logic
controllers. The complicated aspect of this option is that these modules are
custom tailored for individual needs and are often prone to failure. Since these
modules are highly individualized, it is difficult for others to adapt and use
them with any reproducibility and reliability. One of the low cost automated
synthesis boxes commercially available is shown in Fig. 10. Although all the
synthesis functions are automated and streamlined, they are customized for the
synthesis of a particular radiotracer. Several manufacturers have commercial
modules that are now available to perform simple to complex radiochemical
syntheses. Most of these modules are computer controlled and adapt well to a
FIG. 10. An automated synthesis module for producing 18F fluorobromo methane from
18
F fluoride.
275
GARG and GARG
11
FIG. 11. A fully automated synthesis module housed in a hot cell for producing C
labelled radiopharmaceuticals.
hot cell, cave, or even mini-cells. One of the examples is shown in Fig. 11. This
particular unit is used for the synthesis of 11C labelled molecules. A large
number of neuroreceptor ligands and other 11C radiopharmaceuticals use 11C
methyl iodide as a common synthone. Therefore, a number of molecules can be
synthesized using this module. There are several advantages of these
commercial modules; the user does not need to learn the mechanics behind the
module and the procedures are mostly standardized by the manufacturers. The
three major categories of modules that are available in the market are: a 11C
methyl iodide production and methylation module; 18F nucleophilic fluori-
nation modules and 18F electrophilic fluorination modules. Although not all
synthesis can be accomplished using one of the available modules, they are
optimized for the production of most commonly used radiopharmaceuticals. It
is important to remember that some of the fully automated modules are quite
expensive and could cost well above US $100 000. Nonetheless, it does provide
an option to establish reliable and regulatory compliant production of radio-
pharmaceuticals. Using automated synthesis modules is by far the most
effective, safe and practical approach to manufacture radiopharmaceuticals.
The production of PET radiopharmaceuticals is becoming increasingly
valuable and indispensable. Unlike a decade ago, at present, one has multiple
choices for the safe and effective handling of PET isotopes. It is imperative that
radiopharmaceutical laboratory design should consider addressing require-
ments of their local regulatory agencies and personnel safety. All PET research
radiopharmaceutical laboratories should require a properly shielded
276
SESSION 12
FIG. 12. Fume hood converted to lead lined cave for remote handling of radioisotopes.
The reagents are added from outside using syringes and slider valves (located on upper
right hand corner of lead wall) and push buttons (on right side sash) to the reaction vessel
located behind the lead wall.
277
GARG and GARG
FIG. 13. A typical hot cell fitted with robotic arms (manipulators). The front door is
closed for radiochemistry operations. The front panel provides easy access to various
valves or other optional procedures.
shielded at the top and the bottom ends which significantly reduces the
radiation exposure in and around the radiochemistry laboratory. The hot cells
provide easy access to inside work space for preparatory work before the
synthesis and for easy cleanup after use. During the synthesis, all doors and side
panels are closed to minimize radiation exposure to laboratory personnel.
These hot cells are ideal to house synthesis boxes and automated procedures.
Depending on the option purchased, the final product can be delivered either
to a lead lined side drawer or accessed from a side panel without opening the
hot cell doors. It is advisable to wait for the radioactivity to decay before
opening the hot cell doors for cleanup or for setting up for reuse. There are
several manufacturers that sell hot cells internationally. Since this is a large
investment, it is advisable to match the options that a particular vendor
supplies and the site requirements for safe and effective operation before
finalizing a hot cell vendor. It should not be assumed that all hot cell vendors
will provide the same options, designs and accessories.
A large number of laboratories acquire robotic arms (manipulators) to
help with remote handling of radioactivity within the hot cell (Fig. 14). If semi-
automated synthesis or manual interventions are anticipated during the radio-
synthesis, it is strongly advised to have manipulators installed in the hot cells
(Figs 14 and 15). As shown in Figs 14 and 15, the operator works with the
manipulator arms from outside the hot cell. The arms aid in moving and
transfering radioactivity from one place to another and activating or
proceeding with the next process as needed, similar to that performed during
manual processing. Once the skill to operate these manipulators is mastered,
278
SESSION 12
FIG. 14. Operation of robotic arms (manipulators) from outside for safe and effective
radiochemical handling.
FIG. 15. Inside view of the hot cell while operator is working with the manipulators from
outside the hot cell.
they are helpful in accomplishing manual synthesis steps remotely and safely.
In addition, this option facilitates safe handling of multicurie quantities of
radioactivity while maintaining safety. These manipulators provide convenient
work access within the hot cell area while the doors are closed (Fig. 15). There
are several vendors manufacturing manipulators that work in hot cells and
most of them ship these items internationally. Most hot cell vendors can retrofit
these manipulators if not purchased during the hot cell acquisition or installation.
279
GARG and GARG
REFERENCES
[1] GARG, P.K., GARG, S., ZALUTSKY, M.R., Fluorine-18 labelled analogs of
meta-iodobenzyl guanidine. J. Labelled Compd. and Radiopharm., 32: 544-546,
1993.
[2] GARG, P.K., GARG, S., ZALUTSKY, M.R., Synthesis and preliminary evalua-
tion of para- and meta-[18F]fluorobenzylguanidine. Nuc. Med. Biol., 21: 97-103,
1994.
[3] GARG, P.K., GARG, S., BIGNER, D.D., ZALUTSKY, M.R., Selective localiza-
tion of an 18F-labeled Mel-14 monoclonal antibody F(ab')2 fragment in a subcuta-
neous human glioma xenograft model. Cancer Res., 52: 5054-5064, 1992.
[4] GARG, P.K., GARG, S., ZALUTSKY, M.R., Fluorine-18 labeling of monoclonal
antibodies and fragments with preservation of immunoreactivity. Bioconj. Chem.,
2: 44-49, 1991.
[5] GARG, S., GARG, P.K., ZALUTSKY, M.R., Acylation reagents for the radiohal-
ogenation of peptides. J. Labelled Compd. and Radiopharm., 32: 212-213, 1993.
280
SESSION 12
[6] GARG, P.K., LABAREE, D.C., HOYTE, R.M., HOCHBERG, R.B., [7α-
18
F]Fluoro-17α-methyl-5α-dihydrotestosterone: a ligand for androgen receptor-
mediated imaging of prostate cancer. Nucl. Med. Biol., 28: 85-90, 2001.
[7] GARG, P.K., NG, C.K., SOUFER, R., Synthesis and biodistribution of meta-
[18F]fluorobenzylguanidine in the presence and absence of desimipramine (DMI,
an uptake 1 blocker) using rats. J. Labelled Compd. and Radiopharm., 40, 1997.
[8] GARG, P.K., GARG, S., WELSH, P., ZALUTSKY, M.R., Fluorine-18 labeled
analog of metaiodobenzylguanidine. J. Labelled Compd. and Radiopharm., 22:
544-546, 1999.
[9] BERRY, C.R., et al., Para-[18F]fluorobenzylguanidine kinetics in a canine
coronary artery occlusion model. J Nucl Cardiol, 3: 119-129, 1996.
[10] BERRY, C.R., GARG, P.K., ZALUTSKY, M.R., COLEMAN, R.E.,
DeGRADO, T.R., Uptake and retention kinetics of para-fluorine-18-fluoroben-
zylguanidine in isolated rat heart. J Nucl Med, 37: 2011-2016, 1996.
[11] BERRY, C.R., et al., Imaging of pheochromocytoma in 2 dogs using p-[18F] fluor-
obenzylguanidine. Vet Radiol Ultrasound, 43: 183-186, 2002.
[12] KIYONO, Y., KAJIYAMA, S., FUJIWARA, H., KANEGAWA, N., SAJI, H.,
Influence of the polyol pathway on norepinephrine transporter reduction in
diabetic cardiac sympathetic nerves: implications for heterogeneous accumulation
of MIBG. Eur J Nucl Med Mol Imaging, 32: 438-442, 2005.
[13] VALK, P.E., MATHIS, C.A., PRADOS, M.D., GILBERT, J.C., BUDINGER, T.F.,
Hypoxia in human gliomas: demonstration by PET with fluorine-18-fluoromisoni-
dazole. J Nucl Med, 33: 2133-2137, 1992.
[14] MARTIN, G.V., et al., Noninvasive detection of hypoxic myocardium using
fluorine-18-fluoromisonidazole and positron emission tomography. J. Nucl. Med.,
33: 2202-2208, 1992.
[15] RAJENDRAN, J.G., et al., Hypoxia and glucose metabolism in malignant tumors:
evaluation by [18F]fluoromisonidazole and [18F]fluorodeoxyglucose positron
emission tomography imaging. Clin Cancer Res, 10: 2245-2252, 2004.
[16] SORGER, D., et al., [18F]Fluoroazomycinarabinofuranoside (18FAZA) and
[18F]Fluoromisonidazole (18FMISO): a comparative study of their selective
uptake in hypoxic cells and PET imaging in experimental rat tumors. Nucl Med
Biol, 30: 317-326, 2003.
[17] MAHY, P., et al., Preclinical validation of the hypoxia tracer 2-(2-nitroimidazol-1-
yl)- N-(3,3,3-[(18)F]trifluoropropyl)acetamide, [(18)F]EF3. Eur J Nucl Med Mol
Imaging, 31: 1263-1272, 2004.
[18] JOSSE, O., LABAR, D., GEORGES, B., GREGOIRE, V., MARCHAND-
BRYNAERT, J., Synthesis of [18F]-labeled EF3 [2-(2-nitroimidazol-1-yl)-N-
(3,3,3-trifluoropropyl)-acetamide], a marker for PET detection of hypoxia.
Bioorg Med Chem, 9: 665-675, 2001.
[19] GARG, P.K., DeGRAFF, W., GARG, S., ZALUTSKY, M.R., MITCHELL, J.B.,
4-Fluorobenzyl amine and phenylalanine methyl ester conjugates of 2-nitroimida-
zole: Synthesis and evaluation as hypoxic cell radiosensitizers. Int. J. Radiat.
Oncol. Biol. Phys., 23: 593-596, 1992.
281
GARG and GARG
282
RAPID METHOD FOR RADIOFLUORINATION OF
PYRIDINE DERIVATIVES: PROSTHETIC GROUPS
FOR LABELLING BIOACTIVE MOLECULES
Abstract
1. INTRODUCTION
283
AL JAMMAZ et al.
molecules [4, 5]. Furthermore, the short half-life of 18F imposes an additional
time constraint on the application of such a method. The efficiency and
simplicity of the nucleophilic fluoride substitution has been accepted and
applied for the production of several radiopharmaceuticals from precursor
substrates bearing a good leaving group [6, 7]. As part of their on-going
research effort to develop prosthetic precursors for the radiohalogenation of
bioactive molecules such as peptides, the authors have synthesized N-succin-
imidyl activated esters of the 2-[18F]fluoro-4-pyridine and 6-[18F]fluoro-3-
pyridine carboxylates. These intermediates were then used to label a potent
chemotactic peptide [N-Formyl-Nle-Leu-Phe-Nle-Tyr-Lys] and a biodistri-
bution study in normal CBA/J mice was investigated.
2. EXPERIMENTAL
The chemicals used in the study were all analytical reagent grade
purchased from Aldrich (St. Louis, United States of America) and were used
without further purification unless stated. Acetonitrile was kept over molecular
sieves. Sep-Pak cartridges were purchased from Waters-Millipore (USA). Thin
layer chromatographic (TLC) sheets were purchased from Gelman Sciences
Inc. (Ann Arbor, USA). High performance liquid chromatography (HPLC)
analysis was carried out on Econosil C-18 reversed phase columns (semi-
preparative, 250 mm × 10 mm or analytical, 250 mm × 4.6 mm). The solvent
system used for the latter was non-linear gradient (eluant A, water with 0.1
TFA; eluant B, acetonitrile/water, 3/1 v/v with 0.1% TFA; gradient, 0–90% B,
90–0% B and 90–10% B over 10 min each at a flow rate of 1.5 mL/min).
A Jasco (Tokyo, Japan) chromatographic system equipped with a variable
wavelength ultraviolet monitor and in tandem with a Canberra flow through
radioactivity detector was used. Ultraviolet absorption was monitored at
254 nm. Chromatograms were acquired and analysed using BORWIN
software. Melting points were determined on a Thomas Hoover Unimelt
capillary melting point apparatus. Mass spectroscopy was run on a Quattra
electrospray mass spectrometer (ES-MS).
Compounds (2a,b), (3a,b), (4a,b), (5a,b), (6a), (7a) and (8a) in Figs 1 and
2 were all synthesized utilizing the methods reported by Stöcklin et al. [8] and
Amartey et al. [9, 10]. Compound (5b), (250 mg, 1.25 mmol) was dissolved in
dry dichloromethane (3 mL) and was purged with nitrogen for 5 min. Methyl-
trifluoromethane sulphonate (150 uL, 1.25 mmol) was added through a rubber
284
SESSION 12
Compound (8a), (50 mg, 0.35 mmol) was dissolved in absolute ethanol
(2 mL) followed by the addition of concentrated sulphuric acid (100 μL). The
mixture was heated in a sealed vial overnight at 60°C. The reaction mixture was
carefully basified using ammonium hydroxide while cold. The product was then
extracted with ether (2 × 10 mL) and dried over anhydrous sodium sulphate.
Evaporation of the ether yielded an oily greenish material, which was solidified
by cooling. The same procedure was used to produce ethyl 6-fluoropyridine-3-
carboxylate (9b) starting from 6-fluoropyridine-3-carboxylic acid (8b).
285
AL JAMMAZ et al.
2.7. Radiosynthesis
Aqueous [18F]-fluoride was produced by the 18O (p,n) 18F reaction. The
fluoride activity (5–20 mCi, 185–740 MBq) was trapped in Kryptofix 2.2.2 (5 mg)
and potassium carbonate (1 mg) in acetonitrile/water solution (950 μL/50 μL), then
dried by azeotropic distillation with aliquots of acetonitrile. The solid residue was
resolubilized in 0.2 mL of CH3CN containing the required amount of precursor
ethyl-2-(N,N,N-trimethylammonium)-4-pyridine carboxylate.triflate (6a) or
ethyl-6-(N,N,N-trimethylammonium)-3-pyridine carboxylate.triflate (6b). The
reaction mixtures were heated in a capped 2 mL reaction vial at 95°C and
fractions were taken for chromatographic analysis at 2, 5, 8 and 10 min. The
286
SESSION 12
287
AL JAMMAZ et al.
The synthesis of the precursor triflates (6a and 6b) needed for the
radiofluorination reactions is outlined in Fig. 1, whereas the scheme shown in
Fig. 2 was used to synthesize the reference compounds (8a,b, 9a,b, 10a,b and
HBr/Br2 KMnO4
NaNO2
N NH2 N Br N Br
HN(CH3)2
CF3SO3CH3 Ethanol
H2SO4
+ CH3 CH3 CH3
N N N N N N
H3C
CH3 CH3 CH3
(6) (5) (4)
N NH2 N F N F
O
O
O N O
Peptide-NH O COOCH2CH3
Peptide / TEA
N F N F N F
288
SESSION 12
289
AL JAMMAZ et al.
3.2. Radiochemistry
TSTU/TEA
O
O
O N O
Peptide-NH O
Peptide / TEA
18 18
N F N F
(15) (14)
290
SESSION 12
temperature. Because of the ionic character of TSTU, the authors have been
able to purify [18F]SFPa,b esters (14a,b) by adsorption on Sep-Pak cyano
cartridge, followed by elution with hexane/ethyl acetate (8/2, v/v) and replacing the
laborious HPLC purification. The radiochemical yields ranged between 60 and
70% (decay corrected) with a preparation time of about 60 min. Radiochemical
purities of both [18F]SFPa,b esters (14a,b) were always greater than 98% as
determined by HPLC and confirmed by TLC-SG using ethyl acetate:methanol
(8:2) as a mobile phase. The retention times and Rf values for all intermediates
and the final product are shown in Table 1. The dry residues of [18F]SFPa,b
(14a,b) were reacted with the chemotactic peptide in DMF and acetonitrile in
the presence of TEA for 15 min at 95°C. The difference between the
unlabelled peptide and the conjugates permitted the isolation of the peptide-
[18F]SFPa,b conjugates (15a,b) by passing through a Sep-Pak C-18 cartridge and
eluting with absolute ethanol (1 mL). Hence, the Sep-Pak purification
technique is amenable to automation and holds considerable promise as a rapid
and simple method for the radiofluorination of bioactive molecules with high
specific activity, a prerequisite for studying low capacity and saturable sites.
The overall radiochemical yields ranged between 40 and 50% (decay
corrected) with a preparation time of about 80 min. Radiochemical purities of
both peptide-[18F]SFPa,b conjugates (15a,b) were greater than 98% as
determined by HPLC and confirmed by TLC-SG.
291
AL JAMMAZ et al.
Retention time
Compound Rf values
(min)
4. CONCLUSION
292
SESSION 12
The values are average of %dose/g ± standard deviation for n = 4. Italicized results
reported by Vaidyanathan and Zalutsky [12].
high yields (>90%) and in a relatively short ‘synthetic’ time (2 min). These
acids were subsequently converted to their N-succinimidyl activated esters
followed by conjugation with chemotactic peptide as a model through their
lysine moieties. Sep-Pak purifications make this technique amenable to
automation and hold considerable promise as a rapid and simple method for
the radiofluorination of bioactive molecules. Tissue distribution in normal
CBA/J mice showed that the peptide-[18F]SFPa was excreted predominantly
(as expected) through the renal system.
293
AL JAMMAZ et al.
ACKNOWLEDGEMENTS
REFERENCES
[1] WILKINSON, J., Recent advances in the selective formation of C-F bond, Chem.
Rev. 92 (1992) 505.
[2] BUTLER, R., The diazotization of heterocyclic primery amines, Chem. Rev. 75
(1975) 241.
[3] CAMPBELL, A., CHAN, E., CHOOI, S., DEADLY, L., SHANKS, R., The
synthesis of some substituted methyl pyridinecarboxylates, Aust. J. Chem. 2
(1971) 377.
[4] DOLL, F., et al., Synthesis and nicotinic acetylcholine receptor in-vivo binding, J
Med. Chem. 42 (1999) 2251.
[5] HORTI, A., RAVERT, H., LONDON, E., DANNALS, R., Synthesis of radi-
otracer for studying nicotinic acetylcholine receptors (+/-)-exo-2-(2-[18F]fluor-5-
pyridyl)-7-azabicyclic[2.2.0] heptane, J. Labelled compd. Radiopharm. 38 (1996)
356.
[6] BERRIDGE, M., TEWSON, T., Chemistry of fluorine-18 radiopharmaceuticals,
J. Appl. Radiat. Iso., 37, 8, 685.
[7] HAKA, M., KILBOURN, M., WATKINS, D., TOORONGIAN, S., Aryltrimeth-
ylammonium trifluoromethanesulfonates as precursors to aryl [18F]fluorides:
improved synthesis of [18F]GRB-13119, J. Label. Compds. Radiopharm. 27 (1989)
833.
[8] WESTER, H., HAMACHER, K., STÖCKLIN, G., A comparative study of n.c.a.
fluorine-18 labeling of proteins via acylation and photochemical conjugation,
Nucl. Med. Biol. 23 (1996) 365.
[9] AL JAMMAZ, I., AL OTAIBI, B., AMARTEY, J., Synthesis of 2-[18F]-fluoroi-
sonicotinic acid hydrazide: Potential radiotracers for tuberculosis, J. of Labeled
Compounds and Radiopharm. 44 (2001) 451.
[10] AMARTEY, J., AL JAMMAZ, I., AL OTAIBI, B., Synthesis of 2-[18F]-fluoroi-
sonicotinic acid hydrazide and initial biological evaluation, J. of Nucl. Med. Biol.
29/8 (2002) 801.
[11] OLAH, G., et al., Synthetic methods and reactions. 63. Pyridinium poly (hydrogen
fluoride) (30% pyridine-70% hydrogen fluoride): a convenient reagent for
organic fluorination reactions, J. Org. Chem. 44 (1979) 3872.
[12] VAIDYANATHAN, G., ZALUTSKY, M., Fluorine-18 labeled chemotactic
peptides: A potential approach for the PET imaging of bacterial infection, J Nucl.
Med. Biol. 22, 6 (1995) 759.
294
1-[18F]FLUOROETHYLENEGLYCOL-2-
NITROIMIDAZOLES: A NOVEL CLASS OF
POTENTIAL HYPOXIA PET MARKERS
*Chemistry Department,
Faculty of Science,
Hashemite University,
Zarka, Jordan
Email: jalil@hu.edu.jo
** Radiopharmazie,
PET-Zentrum,
Universitätsklinikum Tübingen
Tübingen, Germany
295
ABDEL-JALIL et al.
(a) (b)
N N OTs N N X
N NH O O
n n
NO2 (1) NO2 NO2
X=F X = 18F
2a n = 1 3a n = 1 4a n = 1
b n=2 b n=2 b n=2
c n=3 c n=3 c n=3
a) TsO-(CH2CH2O)n = 2, 3, 4-Ts, Et3N, DMF, rt, 4 d; b) (i) CsF, [bmim][BF4], CH3CN, 100 °C, 1h or
(ii) n-Bu4NF, THF, rt, 5d; 18F-radiolabeling: (iii) K18F/Kryptofix 2.2.2., CH3CN, 80 °C, 10 min.
REFERENCES
[1] LIM, J.L., BERRIDGE, M.S., Appl. Radiat. Isot. 44 (1993) 1085-1091.
[2] TOLVANEN, T., et al., J. Nucl. Med. 43 (2002) 1674-1680.
[3] KAEMAERAEINEN, E.-L., KYLLOENEN, T., NIHTILAE, O., BJOERK, H.,
SOLIN, O., J. Label. Compd. Radiopharm. 47 (2004) 37-45.
[4] REISCHL, G., et al., J. Nucl. Med. 43 (2002) 364.
[5] MACHULLA, H.-J., Imaging of Hypoxia, Kluwer Acad. Publishers, Netherlands,
1999.
[6] MATHIAS, C.J., et al., Life Sci. 41 (1987) 199-206.
296
RADIOSYNTHESIS AND IN VIVO EVALUATION IN
MELANOMA-BEARING MICE OF O-(2-
[18F]FLUOROETHYL)-L-TYROSINE AS A TUMOUR
TRACER
MINGWEI WANG, DUANZHI YIN, YONGXIAN WANG
Radiopharmaceutical Centre,
Shanghai Institute of Applied Physics, CAS,
Shanghai, China
Email: wmwnuclear@163.com
Abstract
1. INTRODUCTION
297
WANG et al.
diagnosis [6, 7]. During the search for new PET tracers, radiolabelled amino
acids were widely studied and their powerful clinical potential as tumour PET
tracers in neuro-oncology and for peripheral tumours such as lymphoma
demonstrated [8]. More attention has been paid to the positron emitter 18F
labelled amino acids owing to its advantages over 11C for PET and 123I for
SPECT [9], of which the most typical example is O-(2-[18F]fluoroethyl)-L-
tyrosine ([18F]FET), an analogue of tyrosine developed recently [3, 10]. It was
synthesized by a nucleophilic displacement reaction with much higher radio-
chemical yield than L-[18F]FT [11, 12] and [18F]FMT [13, 14], which were both
derived from an electrophilic substitution reaction. Originally, [18F]FET was
prepared by a two-step reaction consisting of fluorination of 1,2-
bis(tosyloxy)ethane and fluoroethylation of unprotected L-tyrosine, then using
HPLC [10] or simple solid phase extraction [15] to separate the final product.
However, the existing two-step synthesis of [18F]FET appears cumbersome
because of the two separate purifications involved in the process. Later, one-
step syntheses of [18F]FET via direct nucleophilic radiofluorination of corre-
sponding precursors were developed by Hamacher and Coenen [16] and
Wanga et al. [17].
In this paper, the authors present a novel synthesis method of [18F]FET that
they have developed from easily available precursor to improve the radiochemical
yield and to simplify the purification procedure. The feasibility of [18F]FET as a
PET tracer for brain and probably peripheral tumour imaging was evaluated in
B16 melanoma-bearing mice via biodistribution and autoradiography.
2.1. General
298
SESSION 12
Alltech Associates, Inc., USA, respectively. GF254 silica gel plate based on glass
came from the Huiyou Silica Development Co. Ltd, Yantai, Shangdong, China.
1
H NMR assays were performed on an AVANCE 500 NMR spectrometer
(BRUKER). Mass spectra were recorded on a MicroMass GCT CA 055 mass
spectrometer (Agilent LCMSD-SL, Agilent Technologies, Palo Alto, USA).
Autoradiography was performed on a Packaged Cyclone Storage Phosphor
System with storage phosphor scanner and screen (Perkin Elmer, USA).
Radioactivity was measured with either a radioactivity counter (FJ-391A2,
Beijing Nuclear Instruments, Beijing, China) or radioimmune gamma counter
(SN-697, Rihuan Photoelectric Instruments Co. Ltd, Shanghai, China). Thin
layer chromatography (TLC) was performed using an imaging scanner
(AR-2000, Bioscan, USA) or visualized by UV lamp (Anting Electronic Instru-
ments, Shanghai, China). HPLC was carried out on a Dionex summit system
equipped with a P680 HPLC pump (Dionex, USA), PDA-100 photodiode
array detector, flow count detector (Bioscan, USA), analytical Vydac C18
column (10 μm, 2.5 mm × 250 mm, Shimadzu Corporation, Japan).
No-carrier-added aqueous [18F]fluoride ion was supplied by Amersham
Kexing Pharmaceuticals Co. Ltd.
299
WANG et al.
After cooling to room temperature with a cold nitrogen stream, the dry
residue was redissolved in dichloromethane (1.5 mL). The solution was passed
through a Sep-Pak Silica Plus cartridge preconditioned with diethyl ether and
then eluted with diethyl ether (3 mL). The eluate was dried again with a stream
of N2 gas. Trifluoroacetic acid (0.5 mL) was added to the dry residue and
maintained at room temperature for 5 min. Afterwards, the solvent was
evaporated under a continuous N2 flow. To the residue was added 1 mol/L
aqueous NaOH solution (0.5 mL), followed by heating for 10 min at 80°C to
conduct hydrolysis. The reaction mixture was neutralized with a 1 mol/L
solution of HCl (0.5 mL). After adding a certain volume of phosphate buffer
saline (PBS, pH7.4), the solution was passed through a sterile 0.22 µm
membrane filter to afford the isotonic [18F]FET injection solution.
COOMe COOH
a), b)
TsO 18
NHBoc F NH2
O O
N-BOC-(O-TsE)-L-Tyr-OMe [18F]FET
300
SESSION 12
The tumour-bearing mice were injected with 2.90–3.70 MBq (80–100 μCi)
of [ F]FET in 100 μL of PBS through the tail vein. Three mice were sacrificed
18
at each time point by extirpation of eyeball at 5, 10, 30, 60, 90 and 120 min post-
injection. After, the dissection was carried out and the tissue samples of
interest, including tumour, brain, liver, heart, lung, spleen, kidney, stomach,
small intestine, pancreas, muscle, bone, skin and blood, were collected. All
samples were weighed and the radioactivity was measured using a SN-697 type
radioimmune gamma counter, applying a decay correction. Counts were
compared with those of standards and the results were expressed as a
percentage of injected radioactivity dose per gram of tissue (%ID/g).
301
WANG et al.
Before the above animal experiments, three normal mice were injected in
the tail vein with [18F]FET for the investigation of activity–time correlation of
blood. 5 μL of blood was collected by cutting the tail at 1, 3, 5, 10, 15, 30, 45, 60,
90, 120, 150 and 180 min post-injection followed by measurement of the radio-
activity of blood at each time point. The activity–time correlation of blood was
expressed as a function of time with radioactivity per μL of blood and analysed
with DAS 1.0 drug analysis software.
302
SESSION 12
Uptake (%ID/g)
5 min 10 min 30 min 60 min 90 min 120 min 150 min 180 min
Tumour 12.7 ± 6.6 19.4 ± 5.6 16.3 ± 5.7 14.6 ± 0.7 12.9 ± 2.0 8.2 ± 3.8 2.4 ± 0.1 2.0 ± 0.1
Skin 5.0 ± 3.6 7.5 ± 1.8 4.8 ± 0.2 3.4 ± 1.4 2.5 ± 1.2 1.9 ± 0.0 1.0 ± 0.1 1.3 ± 0.2
Brain 2.4 ± 1.3 4.3 ± 0.7 3.9 ± 0.6 4.0 ± 0.0 3.5 ± 0.3 3.4 ± 1.8 2.5 ± 0.3 2.0 ± 0.1
Liver 11.3 ± 4.4 14.2 ± 1.5 7.5 ± 0.5 4.8 ± 0.4 4.2 ± 0.3 2.9 ± 0.6 1.8 ± 0.7 1.5 ± 0.7
Heart 12.5 ± 4.5 14.3 ± 2.6 8.1 ± 0.6 5.3 ± 0.4 4.7 ± 0.5 3.0 ± 0.8 2.4 ± 0.4 1.8 ± 0.5
Lung 10.9 ± 3.9 14.4 ± 2.6 8.6 ± 0.6 5.6 ± 0.6 4.7 ± 0.6 3.3 ± 1.0 2.7 ± 0.2 1.8 ± 0.7
Spleen 9.3 ± 5.1 15.0 ± 1.7 8.4 ± 0.8 6.0 ± 0.5 5.8 ± 0.6 3.5 ± 0.9 3.4 ± 0.9 2.0 ± 0.7
Kidney 15.4 ± 6.5 14.9 ± 1.8 9.3 ± 0.1 6.6 ± 0.6 5.7 ± 0.9 3.7 ± 0.7 3.3 ± 0.7 2.1 ± 0.2
Intestine 9.7 ± 4.3 12.8 ± 1.6 7.3 ± 0.8 5.0 ± 1.0 5.0 ± 0.6 5.0 ± 0.3 2.5 ± 0.6 2.0 ± 0.2
Pancreas 27.5 ± 3.7 59.8 ± 9.1 41.1 ± 8.1 32.9 ± 8.6 26.4 ± 9.2 21.1 ± 7.0 15.8 ± 7.8 11.1 ± 1.5
Muscle 9.5 ± 4.9 12.2 ± 2.4 8.1 ± 0.6 5.1 ± 0.5 4.6 ± 0.5 3.0 ± 0.6 2.8 ± 0.4 1.9 ± 0.0
Bone 10.0 ± 4.1 17.7 ± 1.7 14.6 ± 3.3 11.9 ± 1.1 11.3 ± 1.0 10.9 ± 3.8 9.6 ± 0.8 8.2 ± 1.2
Stomach 10.3 ± 5.8 10.7 ± 1.3 6.0 ± 1.2 5.9 ± 0.8 5.8 ± 1.8 4.4 ± 1.3 3.3 ± 0.5 2.5 ± 0.2
Blood 12.0 ± 3.8 12.6 ± 3.8 8.0 ± 0.6 5.2 ± 0.2 5.0 ± 1.4 2.8 ±0.6 2.7 ± 0.3 2.0 ± 0.3
19.4 ± 5.6, 4.3 ± 0.7 and 59.8 ± 9.1 at 10 min post-injection to 2.0 ± 0.1, 2.0 ± 0.1
and 11.1 ± 1.5 at 180 min post-injection in the tumour, brain and pancreas,
respectively. [18F]FET showed a certain volume of bone uptake and retention
of [18F]FET in the pancreas was higher than that in the other organs as
previously reported [10, 17].
The ratios of T/B, T/M, T/Bd and T/S of [18F]FET were 4.13 ± 0.38, 2.01 ±
0.20, 2.03 ± 0.12 and 3.40 ± 0.14 at 30 min post-injection and 3.63 ± 0.44, 2.85 ±
0.32, 2.80 ± 0.76 and 4.26 ± 0.63 at 60 min post-injection. The higher T/B ratios
indicated the usefulness of [18F]FET as a PET tracer for brain tumour imaging.
Moreover, [18F]FET was also shown to be a promising PET tracer for
peripheral tumour imaging with great potential due to the relatively high ratios
of T/M, T/Bd and T/S.
3.3. QARG
303
WANG et al.
FIG. 2. QARG via non-sectioned whole body autoradiography in the B16 melanoma-
bearing mice after intravenous injection of [18F]FET. The arrow indicates the location of
the tumour.
304
SESSION 12
3000
2500
2000
cpm/uL
1500
1000
500
0
0 20 40 60 80 100 120 140 160 180 200
time (min)
phase (T1/2α = 3.7 min) and long clearance phase (T1/2β = 448.0 min). Combined
with the above results of biodistribution and QARGs, this suggested that
[18F]FET might be a rapid PET imaging agent for tumour diagnosis.
4. CONCLUSION
ACKNOWLEDGEMENTS
305
WANG et al.
REFERENCES
[1] HUSTINX, R., BENARD, F., ALAVI, A., Whole-body FDG-PET imaging in the
management of patients with cancer, Semin Nucl Med 32 (2002) 35-46.
[2] GAMBHIR, S.S., et al., A tabulated summary of the FDG PET literature, J Nucl
Med (2001) 42(Suppl):1S-93S.
[3] HEISS, P., et al., Investigation of transport mechanism and uptake kineticsof O-
(2-[18F]fluoroethyl)-L-tyrosine in vitro and in vivo, J Nucl Med 40 (1999) 1367-
1373.
[4] KUBOTA, R., et al., Microautoradiographic study for the differentiation of intra-
tumoral macrophages, granulation tissues and cancer cells by the dynamics of
fluorine-18-fluorodeoxyglucose uptake, J Nucl Med 35 (1994) 104-112.
[5] SHREVE, P.D., ANZAI, Y., WAHL, R.L., Pitfalls in oncologic diagnosis with
FDG PET imaging: physiologic and benign variants, Radiographics 19 (1999) 61-
77.
[6] BROWN, R.S., et al., Glucose transporters and FDG uptake in untreated primary
human non-small cell lung cancer, J Nucl Med 40 (1999) 556-565.
[7] MAROM, E.M., et al., Correlation of FDG-PET imaging with Glut-1 and Glut-3
expression in early-stage non-small cell lung cancer, Lung Cancer 33 (2001) 99-
107.
[8] JAGER, P.L., et al., Radiolabeled amino acids: basic aspects and clinical applica-
tions in oncology, J Nucl Med 42 (2001) 432- 45.
[9] LAVERMAN, P., BOERMAN, O.C., CORSTENS, F.H.M., OYEN, W.J.G.,
Fluorinated amino acids for tumour imaging with positron emission tomography,
Eur J Nucl Med 29 (2002) 681-690.
[10] WESTER, H.J., Synthesis and radiopharmacology of O-(2-[18F]fluoroethyl)-L-
tyrosine for tumor imaging, J Nucl Med 40 (1999) 205-212.
[11] COENEN, H.H., KLING, P., STÖCKLIN, G., Cerebral metabolism of L-[2-
18
F]fluorotyrosine, a new PET tracer of protein synthesis, J Nucl Med 30 (1989)
1367–1372.
[12] WIENHARD, K., et al., Increased amino acid transport into brain tumors
measured by PET of L-(2-18F)fluorotyrosine, J Nucl Med 32 (1991) 1338-1346.
[13] INOUE, T., et al., Biodistribution studies on L-3-[fluorine-18]fluoro-alpha-
methyl tyrosine: a potential tumor-detecting agent, J Nucl Med 39 (1998) 663-667.
[14] INOUE, T., et al., 18F alpha-methyl tyrosine PET studies in patients with brain
tumors, J Nucl Med 40 (1999) 399-405.
[15] TANG, G.H., WANG, M.F., TANG, X.L., LUO, L., GAN, M.Q., Fully automated
synthesis of O-(2-[18F]fluoroethyl)-L-tyrosine, Journal of Nuclear and Radio-
chemistry 25 (2003) 44-48 (in Chinese).
[16] HAMACHER, K., COENEN, H.H., Efficient routine production of the 18F-
labelled amino acid O-(2-[18F]fluoroethyl)-L-tyrosine, Appl Radiat Isot 57 (2002)
853-856.
306
SESSION 12
[17] WANGA, H.E., et al., Evaluation of F-18-labeled amino acid derivatives and
[18F]FDG as PET probes in a brain tumor-bearing animal model, Nucl Med Biol
32 (2005) 367-375.
[18] BLOCK, D., COENEN, H.H., STÖCKLIN, G., Nucleophilic 18F-Fluorination of
1,n-disubstituted alkanes as fluoroalkylated agents, J Label Compd Radiopharm
24 (1987) 1029–1042.
[19] BLOCK, D., COENEN, H.H., STÖCKLIN, G., 18F-Fluorination of H-acidic
compounds, J Label Compd Radiopharm 25 (1988) 200-216.
[20] PAULEIT, D., et al., PET with O-(2-18F-Fluoroethyl)-L-Tyrosine in Peripheral
Tumors: First Clinical Results, J Nucl Med 46 (2005) 411-416.
[21] BERGSTRÖM, M., et al., Autoradiography with Positron Emitting Isotopes in
Positron Emission Tomography Tracer Discovery, Mol Imag Biol 5 (2003) 390–
396.
[22] BAUER, A., et al., Evaluation of 18F-CPFPX, a Novel Adenosine A1 Receptor
Ligand: In Vitro Autoradiography and High-Resolution Small Animal PET, J
Nucl Med 44 (2003) 1682-1689.
307
.
CYCLOTRON BASED RADIONUCLIDES
AND GENERATORS
(Session 13)
Chairpersons
M.M. VORA
Saudi Arabia
M. HAJI-SAEID
IAEA
.
PRODUCTION OF RADIONUCLIDES WITH A
CYCLOTRON
D.J. SCHLYER
Department of Chemistry,
Brookhaven National Laboratory,
Upton, New York,
United States of America
Email: schlyer@bnl.gov
Abstract
1. INTRODUCTION
Nuclear medicine offers one of the safest ways to diagnose and/or treat a
number of serious, life threatening diseases including cancer. It does so without
adverse effects on normal organs and without the debilitating side effects of
some of the more common treatments and extended hospital stays. Each day,
thousands of patients with cancer, heart disease and other illnesses receive a
radioisotope injection either for diagnosis or for treatment. Radioisotopes and
radiopharmaceuticals, which are at the heart of nuclear medicine, are used in
the United States of America alone in almost 40 000 procedures every day, and
in more than 100 million laboratory tests each year [1–4]. Radioisotopes for
these uses are produced either by neutron bombardment of a target material in
nuclear reactors or from charged particle bombardment in particle accelerators.
311
SCHLYER
2. NUCLEAR REACTIONS
MA
U= T + Sa
MA + Ma a
312
SESSION 13
2.2. Q values
There is a minimum energy below which a nuclear reaction will not occur
except by tunnelling effects. When the compound nucleus decomposes, the
kinetic energy of all the products may be either greater or less than the total
kinetic energy of all the reactants. If the energy of the products is greater, then
the reaction is said to be exoergic. If the kinetic energy of the products is less
than the reactants, then the reaction is endoergic. The magnitude of this
difference is called the Q value. If the reaction is exoergic, Q values are
positive.
Q= Â D(react) - Â D(prod)
ELASTIC
a + A SCATTERING
INELASTIC
a + A SCATTERING
a A Aa
b + NUCLEAR
B
REACTION 1
b + c + D NUCLEAR
REACTION 2
313
SCHLYER
Q value Threshold
(Z p e)(Z T e) Z pZ T e 2 0.90Z p Z T
Vcoul = = ª MeV
d Rp + RT p + AT )
(A 1/3 1/3
314
SESSION 13
Since the compound nucleus must carry off some kinetic energy, the
Coulomb barrier measured in the laboratory will be corrected by the amount:
ÈA ˘
lab
Vcoul = Í CN ˙ Vcoul
Î AT ˚
where ACN is the mass of the compound nucleus and AT is the mass of the
target.
Thus, the energy required to induce a nuclear reaction increases as the Z
of the target material increases and as the Z of the incident particle increases.
For many low Z materials it is possible to use a low energy accelerator, but for
high Z materials, it is necessary to increase the particle energy [8].
dn = I0NAdsσAB
315
SCHLYER
FAC¡
NA =
AA
C = concentration in weight
ℑ = Avogadro’s number
316
SESSION 13
The rate of production is of course affected by the fact that the resulting
nuclide is radioactive and is thus undergoing radioactive decay. For short lived
nuclides, the competition between formation and decay will come to
equilibrium after sufficiently long bombardment times. This point is called
saturation, meaning that no matter how much longer the irradiation occurs the
production rate is equal to the rate of decay and no more product will be
formed.
The rate of formation in this case is given by:
R = Nλ/(1-e-λt)
l = decay constant
È Al N i ˘
s i = 2.678 x 10 -10 Í - lt ˙
Î Ir x(1 - e ) ˚
317
SCHLYER
It should be noted that the time of irradiation and the decay constant are
both given in the seconds. This is due to the fact that the beam current is
defined in microamperes (μA) which is 6.2 × 1012 charges/second.
318
SESSION 13
FIG. 3. Cross-sections for the production of 123I and 124I from 124Te using the 124Te(p,n)124I
and 124Te(p,2n)123I nuclear reactions.
319
SCHLYER
4. CYCLOTRON TARGETS
The goal of a cyclotron target is to get the target material into the beam,
keep it there during the irradiation and then remove the product radionuclide
from the target material efficiently and quickly. The specific design of the
cyclotron target is what allows one to achieve this goal. Unless care is taken in
the design and fabrication of the target, the production of the radioisotope can
be far from optimal and may even be impossible. Although the underlying
nuclear phenomena are very well known, target behaviour in the form of yield,
maximum beam current and obtainable specific activity varies within and
across different users. Much of this variation can be attributed to less than
optimal matching of the targets and their operating conditions to the actual
cyclotrons used. There is a close interplay between beam parameters (size,
intensity, emittance, orientation and energy) and the performance of a given
target. This variation is seen even within the range of commercial target/
cyclotron combinations. A great deal of useful information about targets and
target chemistry can be found in the proceedings of the International
Workshop on Targets and Target Chemistry. These proceedings are available
on-line by courtesy of TRIUMF at http://www.triumf.ca/wttc/proceedings.html
This is a very valuable resource for all who are concerned with the production
of radioisotopes.
320
SESSION 13
Targets can be made for solids, liquids and gases. There are some
advantages and disadvantages to each of these states of matter. The gases are
usually easy to get in and out of the target and the separation of the radioactive
product from the target material is often simple when compared to solids or
liquids. However, gas targets suffer from such effects as beam density reduction
and in-target chemistry. Liquid targets often boil and solid targets may vaporize
and the radioisotopes may be difficult to separate from the target material after
irradiation. Many of these problems are a direct result of the heating of the
target by the beam.
- dE 4p z2 e4 ¡Z 2m0 V2
= ln
dx m0 V2 A I
321
SCHLYER
ℑ = Avogadro’s number
E 9
V = 1.384 10 cm/s
m
The other useful approximations for the adjusted ionization potential are:
I = 13 Z (eV) if Z ≤ 13
3
He S (E) = 4Sp(E/3)
4
He S(E) = 4Sp(E/4)
322
SESSION 13
beam. There are three modes of heat transfer which are active in targets —
conduction, convection and radiation. Radiation is only a significant mode of
heat loss at high temperatures (>500ºC). Gases and liquids can transfer heat via
convection and conduction. Heat transfer in solids is somewhat simpler than in
other media since the heat usually flows through the target matrix mainly by
conduction.
Once the heat has been transferred to the cooled surface of the target, it
will usually be removed by a fluid such as water flowing against the back of the
target. Most of the problems arise at the interfaces, where there are discontinu-
ities in the heat transfer, such as where the target material meets the backing
material or where the backing material meets the cooling water. The more
efficient the transfer design at these interfaces, the better the heat transfer will
be and the less likely one is to have problems with loss of target material or
damage to the target during the irradiation.
In cyclotron targetry, it is usual to have both free convection and forced
convection. In a gas or liquid target during irradiation, the heating of the fluid
inside the target body will cause free convection currents to be set up which will
aid in removing heat from the fluid and decrease the effects of density reduction.
It is also usual to have some forced convective flow of gas over the front entrance
window to the target. For many reasons, this gas is often helium. Since this gas
has very low viscosity, it is very efficient in cooling the front window.
There are several factors which contribute to the maximum beam
currents which may be run with a solid target. Just as is the case with gas and
liquid targets, the beam density is a determining factor. If there are ‘hot’ spots
in the beam, the highest current at which the target may be run will be much
lower than if the beam has a uniform profile.
As a simple example of heat transfer in a solid target, use can be made of
a thallium target which can be considered as a three layer system facing the
accelerator vacuum on one side and the coolant fluid on the other as shown in
Fig. 4 [15].
q Tl = q ’’’
Tl aS
323
SCHLYER
Cu-coolant
Layers Interface
1 2 3
Tl Cu-b Cu-c
VACUUM
Coolant Fluid
cc
30 MeV protons
(Turbulent flow)
aa bb
I( A)
a b c
FIG. 4. The three layer system showing the target material on the far left with the coolant
on the far right.
Layer 2: The Cu–b layer has a physical thickness of b cm. In this layer protons
emerging from the Tl layer are stopped completely. The heat production rate
qCu and the heat production rate per unit volume q’’’Cu in this layer are linked
by:
q Cu = q ’’’
Cu bS
Layer 3: The Cu–c layer has a physical dimension of c cm and merely serves as
a mechanical support for the Tl layer. The total heat (qT) produced in the layers
is transferred to the coolant fluid through this layer by conduction.
qT = qTl + qCu
T = temperature
324
SESSION 13
T
Cu-coolant
T Tl Cu-b Cu-c
m Interface
T
i
T
b
T
c
a b c
T
w x
The chemistry occurring inside a cyclotron target is the basis for the
chemical products which are produced during irradiation. The reaction of the
highly excited nucleogenic atom with the surroundings during the de-excitation is
the determining factor by which radiolabelled molecules will be formed. Many of
the chemical reactions occurring were first studied using hot atom chemistry.
However, the conditions inside the production target are quite different from a
typical hot atom experiment. In the case of a hot atom experiment, the beam
current is usually less than 1 μA and the gas is at a pressure of much less than
100 kPa. In a normal production gas target, the beam current may be 20 or 30 μA
(or higher with the newer targets) and the pressure up to 7 × 106 Pa. However, in
many cases the results from hot atom experiments have been very successful in
explaining the product distributions from production gas targets. The state of the
matter inside a cyclotron target of course depends on the state of the matter
325
SCHLYER
FIG. 6. View inside a gas target (left) during irradiation where the highly excited and
ionized gas molecules emit light and heat. Water target (right) shows light emission from
the highly excited molecules (with perhaps a small contribution from Cherenkov radia-
tion) as the beam slows in the water. The effects of boiling can also be observed in the
picture in the second band of light near the top of the target as the beam passes through the
water vapour and is stopped in the water behind the bubble. Gas target picture courtesy of
S.-J. Heselius.
being bombarded. In a gas target, the gas is highly ionized and ion–molecule, as
well as highly endothermic, reactions are occurring. A view inside the target
during irradiation is shown in Fig. 6 [16–18].
Nearly any chemical species can be formed in this ionic soup. When other
gases are present, either as contaminants or as additives, the situation becomes
very complex. In most cases the final product distribution will be determined
by the thermodynamics of the situation since there is more than enough energy
to overcome the kinetic activation barriers which would place constraints on
the product distribution at lower temperatures.
The separation of the radioisotope from the target material in gas and
liquid targets is usually a simple matter of trapping the desired compound. In
the case of a liquid target, this can be a resin or a solid support such as the
trapping of fluoride ion on a resin column after irradiation of 18O water [19]. In
the gas target, the desired radioisotope can be reacted with some other
compound while still in the gas stream, as is the case with carbon dioxide
produced in a nitrogen gas target where the [11C]CO2 can be transformed into
[11C]methanol in lithium aluminium hydride [20, 21].
326
SESSION 13
6. CONCLUSION
327
SCHLYER
ACKNOWLEDGEMENTS
REFERENCES
[1] ADELSTEIN, S.J., MANNING, F.J., Isotopes for Medicine and the Life Sciences,
National Academy Press, Washington, D.C., (1995).
[2] UNITED STATES DEPARTMENT OF ENERGY, Health and Environmental
Research Advisory Committee, Subcommittee on Nuclear Medicine. Review of
the Office of Health and Environmental Research Program: Nuclear Medicine.
Washington, D.C., U.S. Department of Energy, August (1989).
[3] SRIVASTAVA, S.C., Is there life after technetium: What is the potential of devel-
oping new broad-based radionuclides. Semin. Nucl. Med. 26, 119-131 (1996).
[4] HOLMES, R.A., "National Biomedical Tracer Facility: Planning and Feasibility
Study", U.S. Department of Energy, Society of Nuclear Medicine, New York, New
York (1991).
[5] HELUS, F., COLOMBETTI, L.G., (ed) (1983) Radionuclide Production, CRC
Press Inc. Boca Raton, Florida.
[6] SCHLYER, D.J., (1987) Production of Short-lived Radiopharmaceuticals for
PET. Nuclear Instruments and Methods in Physics Research B24/25 925-927.
[7] GANDARIAS-CRUZ, D., OKAMOTO, K., (1988) Status on the Compilation of
Nuclear Data for Medical Radioisotopes Produced by Accelerators, IAEA
Report INDC(NDS)-209/GZ.
[8] DECONNINCK, G., (1978) Introduction to Radioanalytical Physics, Nuclear
Methods Monographs No.1 Elsevier Scientific Publishing Co. Amsterdam.
328
SESSION 13
329
SCHLYER
[23] VAN DEN BOSSCHE, B., FLORIDOR, G., DECONINCK, J., VAN DEN
WINKEL, P., HUBIN, A., Steady-state and pulsed current multi-ion simulations
for a thallium electrodeposition process, Journal of Electroanalytical Chemistry
531 (1) p.61-70 (2002).
[24] WAHL, R., editor, Principles and Practice of Positron Emission Tomography,
Lippincott Williams & Wilkens, Philadelphia, 2002.
[25] WELCH, M.J., REDVANLY, C.S., editors. Handbook of Radiopharmaceuticals.
Sussex: John Wiley & Sons, 2003.
330
PERSPECTIVES FOR THE LARGE SCALE
PRODUCTION OF RADIOLANTHANIDES
WITH MEDICAL POTENTIAL
**CERN ISOLDE
Geneva, Switzerland
Email: gerd.beyer@hcuge.ch
Abstract
331
BEYER et al.
1. INTRODUCTION
(i) Owing to the chemical similarity of the rare earth elements we are
practically allowed to handle, the radionuclides of this group serve as
‘homologues’ in a standard protocol for the therapy.
(ii) The chemical similarity of the lanthanides provides a unique possibility to
study relationships between physicochemical molecule parameters and
the biological response without changing the basic tracer molecule. A large
number of suitable radionuclides may be used simultaneously in a most
efficient way for this kind of R&D work.
(iii) The radionuclides of the rare earth elements provide an almost universal
variety of half-life and form of radiation such as single photon emitters
for single photon emission tomography (SPECT), beta emitters for
therapy with a large range of beta energies, positron emitters for positron
emission tomography (PET), an alpha emitter (149Tb), as well as several
interesting Auger electron emitters.
With regard to the first aspect, it has already been shown that 90Y and
177
Lu are used with the same peptide conjugate without changes in the
protocol. As regards the second aspect, mixtures (cocktails) of several
radiolanthanides allow a form of ‘fine tuning’ in tracer development, especially
for therapy, as reported, for example, in Ref. [2]. Finally, the third aspect opens
the door to individual in vivo dosimetry using PET based on the homologues
positron emitter available in the group.
332
SESSION 13
Diagnosis Therapy
R&D
SPECT PET b– a e
87g 44 88 47 149 165
Y Sc Y Sc* Tb Er
147 85 173m 90 225
Eu Y Ce Y Ac/
progeny
147 86 139 142
Gd Y Ce Pm
149 134 141 143
Gd Ce/La Ce Pm
155 138 143 149
Tb Nd/Pr Pm Pm*
157 140 144 153
Dy Nd/Pr Ce/Pr Sm*
167 142 144 156
Tm Sm/Pm Pm Eu
169 152 145 159
Yb Tb Sm Gd
145 161
Eu Tb
146 166
Gd/Eu Ho*
147 169
Nd Er
148 177
Gd Lu*
149
Eu
152
Eu
153
Gd
159
Dy
168
Tm
170
Tm
171
Lu
172
Hf/Lu
173
Lu
174
Lu
Note: The generator–parent nuclides listed under the PET isotopes are also pure Auger
electron emitters if one neglects the positrons of the short lived progeny nuclides.
The b– nuclides for therapy labelled with an asterisk (*) show gamma transitions
suitable for SPECT. The nuclides highlighted in bold are those that are already
routinely used in the nuclear medical practice.
333
BEYER et al.
therapy or for certain R&D work. The following criteria were used in selecting
the isotopes:
(a) Nuclear medical imaging using gamma cameras or SPECT requires from
the radionuclide proper single photon radiation in the energy range of
about 100–300 keV (the ideal would be 140 keV, the photon energy of
99m
Tc, the most widely used radionuclide in classical nuclear medical
imaging with SPECT). The half-life should be between several hours and
about a month.
(b) PET requires the positron decay mode, with positron branching, as high
as possible and with a gamma contribution as low as possible.
(c) For therapeutic application it is desirable to have three types of radio-
nuclide. For treatment of manifested solid tumour nodes, beta emitting
radionuclides are useful, without gamma radiation, that could contribute
to uncontrolled whole body dose of the patient. However, an
accompanied gamma radiation suitable for SPECT imaging (100–300
keV) is an advantage. The half-life of the beta emitting isotopes should
preferably be between two days and about two weeks. Alpha emitting
nuclides are useful for treatment of single cancer cells in circulation
(targeted alpha therapy). Fortunately, there is one suitable isotope (149Tb)
in this class of elements. Owing to their chemical similarity, 225Ac has also
been included into the discussion. Finally, Auger electron emitters are
demanded for targeting the DNA inside a cancer cell directly. In
principle, all radionuclides that decay via EC mode are Auger electron
emitters. The criteria for the nuclides of this group are the half-life (<1
month), available purity of the isotope (specific activity) and the absence
of accompanying gamma radiation.
(d) Radionuclides for research distinguish themselves from others by having a
suitable half-life of between a week and a year; they must also have very
suitable characteristic gamma lines that allow them to be detected easily in
a mixture of several isotopes and beta radiation should be absent for
easier handling. Essentially, all the SPECT isotopes are suitable for R&D
as well.
334
SESSION 13
As can be seen from Table 1, there are only a few radionuclides of the
rare earth elements that are commercially available today. A review on the
general production of radiolanthanides has recently been given by Roesch [1].
There are, in principle, three main production routes: (i) radioactive decay
(90Y), (ii) neutron induced reactions performed in reactors, leading to the
neutron rich nuclides, and (iii) charged particle induced reactions, leading
mainly to the neutron deficient nuclides.
The 90Y may be seen as an exception, because this isotope is generated in
the decay from the long lived fission product 90Sr, that is, one of the more
dangerous fission products. A high degree of technical effort and a correspond-
ingly high level of investment was needed in order to set up a GMP conform
production technology that is absolutely safe from the point of view of radio-
protection and that meets the high requirements of radionuclidic purity.
The other nuclides highlighted in Table 1, 153Sm, 166Ho and 177Lu, are
generally produced in reactors via the (n,γ) process, leading to preparations
that are primarily not carrier free. According to Culter et al. [4], the contents of
required nuclides in the irradiated target are 1.4%, 0.43% and 18% for 153Sm,
166
Ho and 177Lu, respectively (conditions: MURR Reactor, n flux density = 3 ×
1014 cm-2·sec-1, 155 h irradiation time, using highly enriched target materials). In
a few cases one can nevertheless obtain non-carrier added (n.c.a.) preparations
in the (n,γ) process, if the required product is generated from the radioactive
decay of the primary (n,γ) product. Examples are 149Pm, 151Pm, 161Tb, 166Ho
(after double neutron capture) and 177Lu [5]. The high specific activity 177Lu is
produced from irradiated Yb targets according to Lebedev et al. [6]. The
double neutron capture process in combination with the beta decay provides
some possibilities for making a few radiolanthanides in n.c.a. quality. Examples
are 166Ho or 156Eu. The latter isotope can be made in reasonable quantities
from 154Sm according to:
154
Sm (n,γ) 155Sm → 155Eu (4.96 a) (n,γ) 156Eu. ( σ1 = 5.5 b) ( σ2 = 4040 b)
335
BEYER et al.
336
SESSION 13
for their production. Apart from the cost, this has important technical conse-
quences. The reactions have many exit channels so that the resulting product
mixture is very complex both chemically and isotopically. In order to make
continuously pure samples of interesting short lived exotic species, a number of
particular fast procedures for their purification has been developed especially
for the rare earth elements.
These new isotope production techniques are centred on an electromag-
netic mass separation that, in conjunction with a chemically selective release
from the target and/or the ion source, allows production of mono-isotopic ion
beams with high efficiency. By collection of the continuous ion beam of long
lived nuclei, the strength of the high purity radioisotopes obtained for medical
research has been thoroughly demonstrated and reviewed, in particular with
respect to the biomedical research done with carrier free lanthanides, mainly
performed within the experimental programme at ISOLDE [8–12]. In this
paper, emphasis is made of the virtues of the ISOL production and separation
methods that have been developed in conjunction with electromagnetic mass
separation and the possibility for their general use in the large scale production
of almost any existing radionuclide of interest.
These procedures are now ripe for application in the commercial radio-
isotope production process where they could rapidly be introduced as an
additional purification step in the existing production lines for making carrier
free variants of currently used medical isotopes. For production of more
337
BEYER et al.
uncommon isotopes of similar high quality, the present ISOL facilities only
allow supplying the samples needed for biomedical research. In future, they
may become widely available as by-products of a number of planned new large
basic physics research facilities on-line to GeV proton accelerators in the MW
class, such as EURISOL [13, 14]. Cooperation with one of these constitutes an
opportunity for their large scale industrial production. This may be done either
in an off-line mode, independent of the physics programme where the radio-
nuclides are harvested from the spent targets, or ultimately from dedicated on-
line target stations optimized for the longer lived medical radioisotopes.
The onset of the spallation reaction at proton or light ions at energies
>100 MeV opens up a vast range of neutron deficient nuclei while the almost
full range of neutron rich nuclei may be produced by the same particles or by
fast neutron induced fission of 232Th or 238U. For years this has been exploited in
the RIB facilities for production and study of the most short lived species [15].
As shown in Fig. 2 these scientifically interesting nuclei located at the extremes
of the production curve are accompanied by a huge amount of longer lived
nuclei found at the top of the yield curve. It can be seen that production rates of
up to 1013 atoms/s may be reached in the future facilities. They result from the
tens of millibarn formation cross-sections in conjunction with the use of very
thick targets (moles/cm2).
These production methods have only rarely been used for supplying
nuclear medicine research and not at all for commercial production. The
reason is not only the difficult access to the energetic particle beams and their
1.E+14
1.E+13
Production rate [atoms/s]
1.E+12
Gd
1.E+11
Dy
1.E+10
Tb
1.E+09
1.E+08
135 140 145 150 155 160 165 170
Mass
FIG. 2. Production rates at EURISOL for some rare earth isotopes which could be
retrieved from the 30 cm long Hg converter target irradiated with a 5 mA, 1 GeV proton
beam.
338
SESSION 13
Integrated
Acceleration target and ion
to 60 keV source unit
Electromagnetic
mass separation
339
BEYER et al.
FIG. 4. Elements for which radioisotopes can be produced in carrier free form using the
ISOL technique [15].
available for the elements shown in Fig. 4 by combining the release of the
products from refractory compounds kept at high temperature with an ion
source of a mass separator [15].
A typical combined target and ion source unit [17] developed for each
element or group thereof is shown in Fig. 5. The mass separation assures a
purification from any other produced isotope of the same element of >103, i.e
FIG. 5. A typical ISOLDE target and ion source unit as used for production of the rare
earth elements.
340
SESSION 13
341
BEYER et al.
58 59 60 61 62 63 64 65 66 67 68 69 70 71
Ce Pr Nd Pm Sm Eu Gd Tb Dy Ho Er Tm Yb Lu
90 91 92 93 94 95 96 97 98 99 100 101 102 103
Th Pa U Np Pu Am Cm Bk Cf Es Fm Md No Lr
FIG. 7. Elements for which the RILIS ionization scheme has been tested at ISOLDE or
other RILIS facilities using copper vapour pump lasers.
342
SESSION 13
343
BEYER et al.
FIG. 8. Schematic layout of a high power target module. A jet of mercury is injected via
an annular nozzle that allows coaxial injection of the multi-MW, 1 GeV proton beam.
Neutrons from the (p,xnyp) reaction in the Hg then interact with the surrounding hot UCx
fission target matrix, from which the radioactive atoms produced emerge for ionization
and subsequent acceleration.
A number of long lived isotopes were included that are suitable for use in
generators, thus making short lived isotopes available on-site in a hospital
without running facilities for direct production. Examples are 188W, 82Sr, 68Ge
and 44Ti, but this list is not exhaustive. Finally, the authors have included some
radioisotopes of the light elements needed for various applications in industry
and research. These radionuclides are usually difficult to obtain (32Si, 26Al and
28
Mg), but could be produced with relative ease in connection with the
EURISOL facility.
In addition, the possibility of access to samples collected from the
radioactive beam spectrum, spent targets, as well as material activated in the
spent beam absorbers, would give free access to an unlimited variety of both
conventional and new radioisotopes.
344
SESSION 13
345
BEYER et al.
REFERENCES
346
SESSION 13
[14] BEYER, G.J., KOESTER, U., RAVN, H.L., Medical Isotope Program at a Multi-
MW proton Driver, Proc. Workshop on Physics with a Multi-MW Proton Source,
May 25-27, 2004, CERN (Switzerland).
[15] RAVN, H.L., ALLARDYCE, B.W., On-line mass separators, in Treatise on Hevy
Ion Science, Vol.8, D.A.Bromley (ed.), Plenum Press, New York 1988.
[16] BEYER, G.J., NOVGORODOV, A.F., ROESCH, F., RAVN, H., Spallation
Produced Radioisotopes for Nuclear Medical Applicatioon, in Data Requirement
for Medical Radio-Isotope production, Tokyo (Japan) April 20-24, IAEA Vienna,
(1987), Isotopenpraxis 25 (1) (1989) 2-10.
[17] KÖSTER, U., ISOLDE target and ion source chemistry, Radiochimica Acta 89
(2001) 749-756.
[18] KÖSTER, U., Resonance ionization laser ion sources, Nucl. Phys. A701 (2002)
441c-451c.
[19] BEYER, G.J., HERRMANN, E., TYRROFF, H., Gewinnung tragerfreier Radio-
nuklide der Lanthaniden (Production of carrier-free radionuclides of the lantha-
nides), Isotopenpraxis 13 (6) (1977) 193-203.
[20] NOVGORODOV, N.A., et al., Isolation of lanthanide and hafnium radioisotopes
from a massive Ta-target irradiated with 1 GeV protons, Mid-Term Evaluation
Workshop on LanthanideChemistry for Diagnosis and Therapy, F.Roesch (ed.),
COST Chemistry Action D 18, Heidelberg (Germany), 22-25 July 2002, p.29.
[21] BEYER, G.J., ROESCH, F., RAVN, H.L., A high purity 82Sr/82Rb generator,
CERN-Report, CERN-EP/90-91 (1990)
[22] YUSHKEVICH, Y.V., et al., A high efficient ion source with surface ionization
for the determination of micro-traces of radioactive Sr isotopes, Report JINR
Dubna P13-94-213 (1994), Annual Report, Inst.Nuclear Chemistry, University
Mainz 1993/1994.
[23] BEYER, G.J., RAVN, H.L., A new type of 81Rb/81mKr generator made by ion
implantation, Appl. Radiat. Isot. 35 (1984) 1075.
[24] The EURISOL Report, A feasibility study for a European Isotope-Separation-
On-Line Radioactive Ion Beam Facility, http://www.ganil.fr/eurisol/
Final_Report.html
[25] RAVN, H.L., Advanced target concepts for production of radioactive ions and
neutrino beams, Nucl. Instr. and Meth. B204 (2003) 197-204.
347
.
PRODUCTION OF 123I-MIBG AT IPEN-CNEN/SP
349
DE BARBOZA et al.
M 98.89 97.68
SD 0.26 0.35
Batch (n = 37) 1h 24 h
M 98.81 97.72
SD 0.30 0.58
350
SESSION 13
REFERENCES
351
.
A NEW 82Sr–82Rb GENERATOR
A. BILEWICZ, B. BARTOŚ
Institute of Nuclear Chemistry and Technology,
Warsaw
Email: abilewicz@ichtj.wew.pl
R. MISIAK, B. PETELENZ
Institute of Nuclear Physics,
Cracow
Poland
353
BILEWICZ et al.
The 82Sr isotope was produced in the AIC-144 cyclotron located in the
Institute of Nuclear Physics, Cracow. In the pilot experiment, a target of 0.133 g
RbCl of natural isotopic abundance (72.17% 85Rb, 27.83% 87Rb) was irradiated
for 4 h with the internal proton beam of 48 MeV and 0.5 μA. At this energy,
proton activation of the natural rubidium target leads to direct or indirect
formation of 82,83,85Sr and 82,83,84,86Rb isotopes. The radionuclides detected by
gamma spectrometry in the irradiated target are presented in Table 1. After the
8 d waiting period, which is enough to allow decay of 83Sr, the RbCl target was
dissolved in 0.5M HNO3 solution. Next, the solution was passed through the
cryptomelane MnO2 column bed. The inactive rubidium (target material) and
83,84,86
Rb were quantitatively adsorbed on the cryptomelane MnO2. The
effluent from the column was made alkaline with 1M NaOH to pH6–8.
Afterwards, the strontium radionuclides from the neutralized solution were
loaded on top of the SnO2(aq) bed. The inorganic ion exchanger (tin oxide)
was prepared by acidification of sodium stannate solution according to the
procedure described in Ref. [6]. The 82Rb formed from decay of 82Sr was eluted
from the column by 0.9% NaCl (physiological saline). The elution was
performed every 10 min. The radionuclide purity of the effluent was measured
by gamma spectroscopy after the decay of 82Rb. Additionally, the decay curves
of the effluent fractions were also measured. The 82Sr and 85Sr breakthroughs
measured by gamma spectroscopy were lower than the established limits. After
passing 1 L of 0.9% NaCl through the column, no significant breakthrough was
observed either by gamma spectrometry or by analysis of the decay curves. The
half-life of the eluted 82Rb determined from the decay curve measured for
more than 6 expected half-lives, is identical with the value reported in the
literature.
354
SESSION 13
REFERENCES
355
.
CYCLOTRON PRODUCTION OF 103Pd VIA PROTON
INDUCED REACTIONS ON A 103Rh TARGET
M. HAJI-SAEID
International Atomic Energy Agency,
Vienna
Abstract
1. INTRODUCTION
357
SADEGHI et al.
by the American Brachytherapy Society [1], which also reports on the large
number of clinical studies and dosimetry problems [2–12]. Essentially, two
radionuclides, namely 125I and 103Pd, are used for this technique. As early as
1958, 103Pd was proposed by Harper et al. [13] for interstitial implantation. It
was not until 1987 that encapsulated 103Pd sources became commercially
available in the USA, where a company now operates more than 10 dedicated
accelerators to produce this nuclide [14]. Recently, a manufacturer in Europe
also brought its patented type of 103Pd seed implants to the world market.
The accelerator production method for 103Pd used nowadays is based on
the irradiation of rhodium metal with rather low energy protons via the
reaction 103Rh (p,n)103Pd, followed by a default chemical separation of the
radionuclide from the expensive target material. Harper et al. [15] and
Lagunas-Solar et al. [16] described some early procedures.
An alternative production and purification route for 103Pd employing
silver targets has been proposed by Fassbender et al. [17].
Irradiated rhodium metal targets (plated layers, foils and wires) have
been frequently dissolved by sodium bisulphate fusion (time consuming,
complex medium), by gold tetrachloroaurate oxidation (very expensive, time
consuming) and by alternating current electrodissolution in hydrochloric acid.
Up to now the latter method has been recommended for the solubility of foils
(not applicable for rhodium powder, wires or fragments) [18, 19]. A new, high
current density electrodissolution technique resulting in quantitative solubility
of the target material in acid has been developed.
Since rhodium is a precious metal, it is, therefore, essential that it be
recovered from the processed solution of the radiochemical separation and
reused for preparation of the electrodeposition bath. The electrodeposition of
Rh using Rhodex baths gives quite acceptable quality for irradiation purposes.
However, after electrodissolution and radiochemical separation, rhodium is
present as chloride complexes in about 6M HCl solutions and not suitable for
direct use in the Rhodex bath. The investigations were, therefore, conducted to
evaluate the cycle of recovery/electrodissolution/electrodeposition for routine
production of 103Pd.
2. EXPERIMENTAL
The production of 103Pd is mainly achieved via the nuclear reaction 103Rh
(p, n)103Pd, which is well suited to low energy cyclotrons [20]. The production of
this radionuclide in the Islamic Republic of Iran is highly important, therefore
the Cyclone-30 cyclotron (IBA, Belgium) at NRCAM was employed. This
work was also partially supported by the IAEA. The solid targetry system in
358
SESSION 13
359
SADEGHI et al.
2.2. Electrodissolution
360
SESSION 13
mounted target and the Rh layer was not dissolved. The resulting Cu(NO3)2/
HNO3 solution was removed by filtration through a glass/graphite filter
combination whereby the Rh fragments are collected on the filter and the
vessel walls. Rhodium fragments were washed with water and removed. A
mixture of 12M hydrochloric acid and chlorine gas was introduced into the
vessel. Electrochemical dissolution of the Rh was done by applying a high AC
density (2.4 A/cm2) between the electrographite filter and a perforated circular
upper graphite electrode mounted at an appropriate distance from the carbon
filter.
361
SADEGHI et al.
362
SESSION 13
4. CONCLUSIONS
FIG. 2. Gamma ray spectrum of an irradiated 103Rh target after electrodissolution, other
peaks include the Pb detector shielding fluorescence X rays.
363
SADEGHI et al.
103
FIG. 3. HPGe spectrum of radiochemically separated Pd. No other peaks have been
detected in the γ spectrum.
bath is recommended for low current beam irradiation (up to 200 µA) and the
latter for current beam irradiation of higher than 200 µA.
The high current density results in a high production rate of the
electroactive chlorine species that rapidly dissolves the rhodium present at the
working electrode. The dissolution rate depends on parameters such as current,
concentration of HCl and weight of rhodium. In general, the magnitude of these
parameters is time dependent. The optimum conditions of the electrodissolution
were as follows: 12N HCl solution, AC density higher than 2.4 A/cm2,
temperature 85°C and bubbling chlorine gas, in which case the dissolution rate
was greater than 99%.
Recovery of 103Pd from rhodium solution was achieved by ion exchange
column with Dowex1X8 resin and NH3+NH4Cl (1:1) as eluant; the obtained
103
Pd purity was more than 99%.
REFERENCES
[1] NAG, S., BEYER, D., FRIEDLAND, J., GRIMM, P., NATH, R., Int. J. Radiat.
Oncol. Biol. Phys. 44 (1999) 789.
[2] MERRICK, G.S., BUTLER, M.W., DORSEY, A.T., LIEF, J.H., Int. J. Radiat.
Oncol. Biol. Phys. 44 (1999) 1111.
364
SESSION 13
[3] PRETE, J.J., et al., Int. J. Radiat. Oncol. Biol. Phys. 40 (1998) 1001.
[4] LING, C.C., Int. J. Radiat. Oncol. Biol. Phys. 23 (1992) 81.
[5] MESSING, E., et al., Int. J. Radiat. Oncol. Biol. Phys. 44 (1999) 801.
[6] BLASKO, J.C., WALLNER, K., GRIMM, P.D., J. Urol. 154 (1995) 1096.
[7] BLASKO, J.C., et al., Urol. Clin. North Am. 23 (1996) 633.
[8] PORTER, A.T., BLASKO, J.C., GRIMM, P.D., REDDY, S.M., RAGDE, H.,
Californian Cancer J. Clin. 45 (1995) 165.
[9] DATTOLI, M., WALLNER, K., SORACE, R., J. Brachyther. Int. 13 (1997) 347.
[10] RAGDE, H., et al., Semin. Surg. Oncol. 13 (1997) 438.
[11] STOCK, R.G., STONE, N.N., TARBERT, A., Int. J. Radiat. Oncol. Biol. Phys. 41
(1998) 101.
[12] WHITTINGTON, R., et al., Int. J. Radiat. Oncol. Biol. Phys. 44 (1999) 1107.
[13] HARPER, P., LATHROP, K., BALDWIN, L., Ann. Surg. 148 (1958) 606.
[14] PORAZZO, M.S., et al., Int. J. Radiat. Oncol. Biol. Phys. 23 (1992) 1033.
[15] HARPER, P.V., LATHROP, K., NEED, J.L., ORNL-LR-DWG 51564 (1961) 124.
[16] LAGUNAS-SOLAR, M.C., AVILA, M.J., JOHNSON, P.C., Appl. Radiat. Isot.
38 (1987) 151.
[17] FASSBENDER, M., NORTIER, F.M., SCHROEDER, I.W., VAN DER WALT,
T.N., Radiochimica Acta 87 (1999) 87.
[18] BOX, W.D., In ORNL-3802 UC-23-Isotopes-Industrial Technology TID-450.39th,
(1964), p.29.
[19] LAGUNAS-SOLAR, M.C., AVILA, M.J., JOHNSON, P.C., Int. J. Appl. Radiat.
Isot. 38(2) (1987) 151.
[20] HERMANNE, A., SONCK, M., FENYVESI, A., DARABAN, L., Nucl. Inst. &
Meth. In Phys. Res. B, 170, (2000) 281.
[21] ZHANG, C., WANG, Y., ZHANG, Y., ZHANG, X., Applied Radiation &
Isotopes, 55, 441 (2001).
365
.
THE STATUS AND POTENTIAL OF NEW
RADIONUCLIDE GENERATORS PROVIDING
POSITRON EMITTERS TO SYNTHESIZE NEW
TARGETING VECTORS FOR PET
Abstract
367
ROESCH et al.
1. INTRODUCTION
368
SESSION 13
Parent Progeny
Generator system
β+branch Eβ+
T½ T½ Application
(%) (MeV)
82 82
Sr Rb 25.6 d 1.27 min 95.0 1.41 Perfusion
140 140
Nd Pr 3.37 d 3.39 min 51.0 0.544 Perfusion
118 118
Te Sb 6.00 d 3.6 min 74.0 0.882 Perfusion
122 122
Xe I 20.1 h 3.6 min 77.0 1.09 (Labelling)
128 128
Ba Cs 2.43 d 3.62 min 69.0 0.869 Perfusion
134 134
Ce La 3.16 d 6.4 min 63.0 0.756 Perfusion
62 62
Zn Cu 9.26 h 9.74 min 97.0 1.28 Labelling, perfusion
52 52m
Fe Mn 8.28 d 21.1 min 97.0 1.13 Perfusion
68 68
Ge Ga 270.8 d 1.135 h 89.0 0.74 Labelling, perfusion
110 110m
Sn In 4.1 h 1.15 h 62.0 0.623 Labelling
44 44
Ti Sc 47.3 a 3.927 h 94.0 0.597 Labelling
72 72
Se As 8.4 d 1.083 d 88.0 1.02 Labelling
2. AIM
369
ROESCH et al.
FIG. 1. Scheme of the 68Ge/68Ga radionuclide generator system and problems related to
the direct use of the generator eluate for medical directions.
370
SESSION 13
68
3.1. Ge/68Ga radionuclide generator
371
ROESCH et al.
Analytical reagent grade chemicals and Milli-Q water (18.2 MΩ⋅cm) were
used. A Bio-Rad AG 50W-X8 cation exchanger minus 400 mesh was preferred
to prepare a microchromatography column. DOTATOC was kindly provided
by Novartis Pharma AG.
For labelling reactions, 11 mL glass vials, Mallinckrodt and reaction
vessels PP, Brand were used. A heating block with a 1.5 cm lead thickness for
radiation adsorption was built using 24 V PTC heating elements. For processing
of the labelling product, C-18 cartridges, Phenomenex Strata-X Tubes, 30 mg
were used. Sterile filtration was done on a 0.22 µm MILLEX®GV membrane
filter. For quality control, TLC (aluminium sheets, silica gel 60, eluent 0.1M
Na3citrate) and HPLC (Machery Nagel column, Nucleosil 5 C18-AB, 250 mm ×
4 mm; eluent, 20% AcCN, 80% TFA 0.01% in H2O, 1 mL/min) were used.
4. EXPERIMENTS
372
SESSION 13
4.2. Labelling
In the case of the new 30 mCi 68Ge/68Ga generator, about 600–750 MBq
68
of Ga was obtained with 7 mL of 0.1M HCl. In addition, in order to obtain
about 1400 MBq, two 30 mCi generators were combined and eluted with 12 mL
of 0.1M HCl in a cascade scheme. These fractions were processed and applied
for labelling. After preconcentration and purification of the initial generator
eluates on the microchromatography column, 68Ga(III) was eluted with the
400 µL 98% acetone–0.05M HCl solution (2 × 10-5 mol HCl). This fraction was
used directly for labelling. For labelling with 68Ga(III), DOTA-octreotides
(DOTATOC, DOTANOC) and desferrioxamine-B-succinyl-octreotide
(DFOOC) were used. All labelling reactions were carried out at temperatures
of ~98°C. Kinetics of the syntheses were recorded up to 15 min by taking
aliquots of 1 µL at 1, 2, 5, 10, 15 min.
Two approaches were studied, namely 68Ga labelling with or without
additional buffer solutions. The processed activity was added to 4.5 mL pure
H2O in a standard glass reagent vial (11 mL, Mallinckrodt) containing 7–14 nmol
DOTATOC. To achieve higher specific activity 0.5–0.7 mL 1 molal HEPES,
pH4.0–4.1 (according to Ref. [6]) was added to 2 mL reaction vessels (PP,
Brand equipped with a vent) containing 2–5 nmol DOTATOC.
Quality control was performed using TLC and HPLC. For HPLC, an
aliquot was dissolved in TFA 0.1% in H2O and used for analysis. Acetone
content was studied by a gas chromatography HP 6890 series GC system.
373
ROESCH et al.
tubing connected the reagent vials in a heating block. The column could be
eluted using a standard single use syringe (3) and was connected to the waste
vial (2). Syntheses were performed in pure water as described above using
14 nmol DOTATOC. After purification of the product on the C-18 cartridge, the
ethanol eluate containing the pure 68Ga-DOTATOC was dissolved in 5–10 mL
0.9% saline solution and sterilized by filtration through a 0.22 µm membrane
filter. Routine quality control was performed rapidly by TLC.
5. RESULTS
68
5.1. Ge/68Ga radionuclide generator performance
( Ga yield, 68Ge breakthrough)
68
374
SESSION 13
5.3. Volume
5.4. Time
With application of the scheme described above (Fig. 2), processing of the
eluate requires only 4 min.
5.5. Labelling
The preconcentrated and purified 68Ga(III) eluted from the resin with
400 µL of the 97.6% acetone–0.05M HCl mixture was used for labelling
reactions. For labelling in pure water, up to 700 MBq 68Ga(III) was added to
4–4.5 mL (preheated) H2O in standard reagent vials containing 7–14 nmol
DOTATOC. The amount of HCl contained in the final eluate solution (400 µL
98% acetone–0.05M HCl = 2 × 10-5 mol H+) provided an overall pH of 2.30
(±0.05). No incorporation was achieved at higher or lower pH. At ~98°C, the
radiolabelling yield was over 95% within 10 min. Variable radiochemical yield
375
ROESCH et al.
and increased adsorption on the glass surface were detected if less than 14 nmol
of peptide was used. Incorporation also dropped with decreasing reaction
temperature.
5.7. Purification
376
SESSION 13
FIG. 4. Purification and pre- and post-purification quality control of 68Ga radiopharma-
ceuticals associated to the post-processing and synthesis scheme.
final product after processing on C-18 contained not more than 0.15 µg of
acetone.
6. DISCUSSION
377
ROESCH et al.
reaction was carried out in pure water using 14 nmol of DOTATOC and
resulted in high incorporation and high overall yield. Up to 400 MBq of
DOTATOC with specific activity up to 40 MBq/nmol could be obtained after
processing on C-18 with a new 30 mCi generator.
Within 20 min, an injectable radiopharmaceutical such as 68Ga-
DOTATOC can be prepared with specific activities of up to 450 MBq/nmol.
While standard specific activities of 40 MBq/nmol are quite acceptable for
clinical diagnosis using 68Ga-DOTATOC, increased specific activities can be
required for clinical application of DOTA-peptides with potential pharmaco-
logical side effects [7].
The application of C-18 cartridges for final purification seems to be an
essential step which provides high radiochemical purity independent of
labelling yield. Furthermore, additional quality control might be avoided.
Thus, a rapid, simple and chemically efficient processing of generator
produced 68Ga(III) was developed. The process guarantees safe preparation of
injectable 68Ga-DOTATOC (or other 68Ga labelled radiopharmaceuticals) for
routine application and can be successfully used in the clinical environment.
Simple equipment for routine preparation of injectable 68Ga-DOTATOC was
installed in various nuclear medicine departments equipped with a PET/CT.
More than 1000 patients with known neuroendocrine tumours were involved in
initial systematic studies to establish and evaluate a clinical protocol. Both
[68Ga]DOTATOC and [68Ga]DOTANOC were successfully used in a series of
human somatostatin receptor expressing tumour diagnoses with PET/CT. The
developed system represents a simple and efficient way for the labelling of
DOTA conjugated biomolecules with generator produced 68Ga(III).
Moreover, a variety of other 68Ga labelled compounds might be synthesized for
many other applications. The current level of radiochemical development and
radiopharmaceutical investigation clearly indicates a significant potential for
the 68Ge/Ga generators, for applications in basic research and for routine
application in state of the art nuclear medicine. In particular, for countries or
medical centres not yet running medical cyclotrons and/or not yet owning a
sophisticated organic radiopharmaceutical production infrastructure, the avail-
ability of the 68Ge/Ga generator might help to initiate PET chemistry develop-
ments and patient diagnoses using PET.
As the developed scheme guarantees safe preparation of injectable 68Ga
labelled DOTA conjugates for routine application (easy to automate) it has
been successfully used in clinical environments. Using the authors’ system, over
the last year several German clinical centres have been involved in establishing
clinical protocols for application of 68Ga-DOTATOC and similar compounds in
more than 500 patients. The clinical impact is extraordinarily high. Using PET
and in particular PET/CT, the diagnosis of neuroendocrine tumours and
378
SESSION 13
Diagnosis:
Therapy:
379
ROESCH et al.
radiochemistry for this generator and have shown the potential of longer lived
radioarsenic to image the targeting parameters of monoclonal antibodies.
In addition, in particular if targeting vectors such as monoclonal
antibodies are used instead of small peptides, the authors recently proposed the
radioarsenic isotopes 72As (T½ = 26 h, 88% ββ+ branching) and 74As (T½ = 17.78 d,
29% ββ+ branching). As a proof of principle, they successfully tested the
hypothesis that a new chimeric IgG3 monoclonal antibody ch3G4 (Tarvacin®)
directed against anionic phospholipids and labelled with radioactive arsenic
isotopes can be used for the vascular targeting and molecular imaging of solid
tumours in rats in vivo. For generators using no carrier added 72Se, the authors
described the distillation of AsCl3, while Se remains in non-volatile compounds
in the residue, and developed a solid phase extraction system with 72Se fixed as
metallic Se. Systematic chemical investigations on the labelling chemistry of no
carrier added radioarsenic are currently being developed prior to the
application of 72As labelled compounds [9–12].
Thus, the potential of PET radionuclide generator systems seems to fit
excellently with the strategy of the IAEA in facilitating the development of
modern medical imaging technologies in the developing countries.
REFERENCES
380
SESSION 13
381
.
RADIOPHARMACY
(Session 14)
Chairpersons
N.G. HARTMAN
United Kingdom
E. JANEVIK-IVANOVSKA
The Former Yugoslav Republic of Macedonia
.
NUCLEAR PHARMACY PRACTICES IN THE UNITED
STATES OF AMERICA
K. OZKER
Medical College of Wisconsin,
Milwaukee, Wisconsin,
United States of America
Email: ozker@mcw.edu
Abstract
There are more than 450 nuclear pharmacies in the United States of America.
Approximately 80% of these are centralized nuclear pharmacies operated by three
major companies: Cardinal Health, Tyco Healthcare/Mallinckrodt and GE Healthcare.
There are 88 independent facilities and two additional companies specialized in PET
radiopharmaceuticals: CTI/PETNET and Eastern Isotopes. Institutional nuclear phar-
macies, representing 20% of the radiopharmacies in the USA, prepare multidose radio-
pharmaceuticals in university or hospital settings. All commercial nuclear pharmacies
are licensed by a state board of pharmacy and operate under the supervision of an
authorized nuclear pharmacist. The Nuclear Regulatory Commission requires that all
nuclear pharmacists be certified pharmacists, completing an approved programme
consisting of 200 h of didactic and 500 h of practical training. Recently, requirements for
aseptic compounding and dispensing of radiopharmaceuticals have been developed by
the United States Pharmacopoeia (USP <797>). The Society of Nuclear Medicine
communicated with the USP in late 2004 regarding unique situations specific to the
preparation of radioactive compounds (such as radiation exposure, shielding require-
ments and contamination risks) that would make it difficult to comply fully with the new
USP regulations. The USP <797> Sterile Compounding Committee subsequently
approved revisions and exemptions for radiopharmaceutical compounding.
385
OZKER
386
SESSION 14
(1) Low risk conditions are where all compounding with aseptic manipula-
tions occur entirely with ISO Class 5 (old class 100) or better air quality
using only sterile ingredients, products, components and devices.
(2) Medium risk conditions include multiple individual or small doses of
sterile products that are compounded or pooled to prepare a
compounded sterile product that will be administered either to multiple
patients or to one patient on multiple occasions. USP <797> implies that
medium risk compounding must also take place in an ISO Class 5 or
better air environment.
(3) High risk conditions include the use of non-sterile ingredients,
components or devices and sterile ingredients, components or devices
which are exposed to air quality inferior to ISO Class 5. In this case, the
compound must subsequently be sterilized in an ISO Class 5
environment.
387
OZKER
level (old class 10 000) of cleanliness. Buffer zone surfaces must be smooth,
impermeable, readily cleanable and capable of sanitization. Low and medium
risk environments must have controlled ISO level Class 5 cleanliness as well as
a buffer zone and an additional ante area (where gowning and hand washing
are carried out) which does not necessarily need to be separated by a physical
wall. High risk environments must have the same features as the low and
medium risk sites but the additional ante area must be separated from the
buffer zone by a physical barrier [4, 5].
USP <797> regulations are currently enforceable by the Food and Drug
Administration and the State Board of Pharmacy, Medicine and Nursing.
Healthcare institutions must begin to comply with the requirements of USP
<797> over a specific timeline. The Society of Nuclear Medicine communicated
with the USP in late 2004 about unique situations specific to the preparation of
radioactive compounds (such as radiation exposure, shielding requirements
and contamination risks) that would make it difficult to comply fully with the
USP regulations. The USP Sterile Compounding Committee met in October
2004 and approved the following revisions to USP <797> [6]:
(1) Currently official <797> requires positive pressure for all sterile
compounding, but that is wrong for radioactive and other hazardous
drugs.
(2) Direct visual inspection of highly radioactive CSPs is not required.
(3) The 99mTc/99Mo generator systems shall be stored and eluted (operated)
under conditions recommended by their manufacturers and applicable
state and federal regulations.
(4) Three or fewer sterile products may be prepared in lower than ISO Class
5 air when there is no direct contact contamination and administration
begins within 1 h and is completed within 12 h of preparation.
388
SESSION 14
REFERENCES
389
.
REGULATORY ASPECTS OF HOSPITAL
RADIOPHARMACY AND CLINICAL TRIALS
A.A. SOYLU
Nuclear Medicine Department,
Faculty of Medicine, Ankara University
Ankara, Turkey
Email: ayfersoylu@yahoo.com
Abstract
1. INTRODUCTION
391
SOYLU
2. CLINICAL TRIAL
3. IMP
4. RADIOPHARMACEUTICALS
392
SESSION 14
once or a few times in their lifetime. In general, these products spend a short
time in the body and no biological effect is expected to occur by the intro-
duction of a radiopharmaceutical to a human being. On-site preparation and
immediate use may be other aspects of radiopharmaceuticals differing from
regular drug products. In spite of these arguments, radiopharmaceuticals are
considered as medicinal products [3] and any radiopharmaceutical used in a
clinical trial is therefore subject to all the legislation regarding IMP.
The radioactive nature of radiopharmaceuticals also makes it necessary
to follow the regulations related to ionizing radiation. Euratom Directive 97/43,
entitled Protection of Individuals Against the Dangers of Ionizing Radiation in
Relation to Medical Exposure applies to exposure of healthy individuals or
patients voluntarily participating in medical or biological, diagnostic or
therapeutic, research programmes [4].
393
SOYLU
important considerations and should prevail over the interests of science and
society.
6. CLINICAL TRIALS
394
SESSION 14
8. CONCLUSION
395
SOYLU
REFERENCES
396
SESSION 14
397
.
STANDARDIZATION AND QUALITY CONTROL OF
AN IN-HOUSE FORMULATION OF 99mTc(V)-DMSA IN
TUMOUR IMAGING AND ASSESSMENT OF
TUMOUR BIOLOGY: WORK IN PROGRESS
+
Regional Centre for Radiopharmaceuticals (BRIT)
Abstract
399
CHOUDHURY et al.
1. INTRODUCTION
400
SESSION 14
Limited clinical trials in the form of a pilot study were carried out in the
patients of biopsy proven lung carcinoma during staging workup to evaluate
the tumour concentrating capability of the radiopharmaceutical and the in vivo
kinetics of the tumour. Whole body imaging was performed with a gamma
camera. 740 MBq of the prepared 99mTc(V)-DMSA was injected intravenously
and images were aquired immediately, at 1, 2 and 3 h. SPECT of the region of
primary malignancy was performed. The radiochemical purity was assessed by
chromatography concomitantly.
401
CHOUDHURY et al.
3. RESULTS
Different batches of both the kits were used for chromatographic studies.
It was observed that the unbound (free) 99mTcO4 (technetium pertechnetate) in
both kits was less than 5%. This was also substantiated in the imaging studies.
Biodistribution and initial clinical studies showed predominantly renal pelvi-
calyceal excretion and bladder concentration. No tracer concentration was
seen in the salivary glands, thyroid gland or stomach, suggesting the absence of
significant unbound 99mTcO4 in the preparation. Radiochemical purity in both
the kits was more than 90% with the method of formulation used. However,
the incubation time taken to achieve this purity was variable in both the kits.
The kit supplied by BRIT (India) took 15 min of incubation time (Fig. 1),
whereas the Amersham kit required about 1 h of incubation time (Fig. 2) to
achieve the same purity. Significant amounts of DMSA(III) were found in the
latter preparation after 15 min of incubation (Fig. 3). On bubbling with hospital
grade oxygen for 20 min, the desired radiochemical purity was achieved in 15
min (Fig. 4). Both kits were stable for at least 3 h (Figs 5–7 and Tables 1–3).
15 m in.post preparation
on-Kit 1
200
20
150
15
ounts
k count
100
10 Series2
50
0
1 3 5 7 9 11
cm s
402
SESSION 14
200000
150000
counts
100000 Series2
50000
0
1 3 5 7 9 11
cm s
15 m in post preparation-Kit 2
20
k counts
15
10 Series2
5
0
1 3 5 7 9 11
cm s
403
CHOUDHURY et al.
40
k counts
30
20 Series2
10
0
1 3 5 7 9 11
cm s
80
60
counts
40 Series2
20
0
1 3 5 7 9 11
cm s
404
SESSION 14
300000
Counts
200000
Series2
100000
0
1 3 5 7 9 11
cm s
12
10
8
Counts
6 Series2
4
2
0
1 3 5 7 9 11
cm s
405
CHOUDHURY et al.
* The above results reflect the overall performance of a number of samples of both the
kits from different batches. The pH of the preparations was 8.5–9.0. The Rf values for
99m
Tc(III)-DMSA and 99mTc(V)-DMSA were 0.3 and 0.6–0.7, respectively.
* The above results reflect the overall performance of a number of samples of both the
kits from different batches. The pH of the preparations was 8.5–9.0. The Rf values
for 99mTc(III)-DMSA and 99mTc(V)-DMSA were 0.3 and 0.6–0.7, respectively.
406
SESSION 14
4. DISCUSSION
407
CHOUDHURY et al.
99m
FIG. 9. Tc(V)-DMSA scan in the same patient post-treatment (concurrent chemo-
therapy and external radiation) showing residual disease.
408
SESSION 14
FIG. 10. Post-treatment CT scan of the chest in the same patient showing residual disease
corresponding to the scintigraphic abnormality.
409
CHOUDHURY et al.
99m
FIG. 11. Normal biodistribution of Tc(V)-DMSA in a radiochemically pure prepa-
ration.
410
SESSION 14
mostly in medullary carcinoma thyroid and bone metastasis [9–12]. The authors
are attempting to study tumour biology with an emphasis on detection,
response to treatment and follow-up. Initial clinical results have been encour-
aging.
5. CONCLUSION
ACKNOWLEDGEMENT
This study was supported by a grant ( IAEA E1.30.28 ) from the IAEA in
the framework of the coordinated research project entitled Standardization
and Quality Control of In-house Prepared Radiopharmaceuticals in Nuclear
Oncology.
REFERENCES
[1] OHTA, H., et al., A new imaging agent for medullary carcinoma of the thyroid, J
Nucl Med 25 (1984) 323-325.
[2] WESTERA, G., GADZE, A., HORST, W., A convenient method for the
preparation of 99mTc(V)-dimercaptosuccinic acid [99mTc(V)-DMSA], J Appl
Radiat Isot 36 (1985) 311-312.
[3] CHAUHAN, U.P.S., et al., Evaluation of a DMSA kit for instant preparation of
99m
Tc(V)-DMSA for tumour and metastasis scintigraphy, Nucl Med Biol 19 (1992)
825-830.
[4] BABBAR, A., KASHYAP, R., CHAUHAN, U.P.S., A convenient method for the
preparation of 99mTc labelled pentavalent DMSA and its evaluation as a tumour
imaging agent, J Nucl Med Biol 35 (1991) 100-104.
[5] RAMAMOORTHY, N., et al., Preparation and evaluation of 99mTc(V)-DMSA
complex: Studies in medullary carcinoma of thyroid, Eur J Nucl Med 12 (1987)
623-628.
411
CHOUDHURY et al.
412
DEVELOPMENT OF CENTRALIZED
RADIOPHARMACIES IN SPAIN: A SUCCESSFUL
EXPERIENCE IN EUROPE
I. OYARZÁBAL, R. JIMÉNEZ-SHAW
Molypharma, S.A.,
Madrid, Spain
Email: rjs@molypharma.es
Abstract
1. INTRODUCTION
In Spain, 650 000 nuclear medicine procedures are performed every year
in more than 120 nuclear medicine departments in public and private hospitals.
The doses to be administered to patients were traditionally prepared by
hospital personnel in small hot laboratories in the departments. However,
during the 1990s new legislation was developed that triggered the need for a
change in the model. The change from radiotracers to the new concept of
radiopharmaceuticals led to pharmaceutical requirements for the devel-
opment, fabrication, commercialization and preparation of radioactive
compounds and their administration to patients. Additionally, regulatory
authorities imposed quality assurance rules in nuclear medicine practices.
As regards dose preparation and administration, those legal require-
ments established new specific criteria derived from compliance with good
manufacturing practices (GMPs) in several areas such as the following:
413
OYARZÁBAL and JIMÉNEZ-SHAW
— 45% of the unit doses are prepared and supplied from 6 centralized
radiopharmacies;
— 20% of the unit doses are prepared in hospital radiopharmacies but
managed and operated by an external contractor;
— 35% of the unit doses are prepared in hospital radiopharmacies managed
by hospital personnel.
2. INNOVATIVE CONCEPT
414
SESSION 14
Each unit has been designed to meet both GMPs and radioactive regula-
tions. These are conflicting regulations, as GMPs call for the protection of the
product, requiring positive pressure to avoid possible contamination of the
product, while radioactive regulations establish protection for the operator and
the environment, requiring negative pressure. This apparent contradiction is
solved in the company’s units through having a pressure cascade, the laminar
air flow open cabins being in negative pressure in relation to the clean room
where they are located. The clean room is in positive pressure over the
reception and expedition rooms in order to avoid any possible contamination.
The laminar airflow open cabins are shielded to protect the operator and
include a dose calibrator activimeter connected to the computer system.
Labelling and unit dispensing is done within the cabins. Operators are
comfortably seated. Generators are eluted in a special cabinet.
The unit also has conventional laboratory equipment and rooms, quality
control, air conditioning and filters, as required by pharmaceutical and
radioactive regulations.
415
OYARZÁBAL and JIMÉNEZ-SHAW
To carry out its activities, the company has adopted a quality management
system based on processes in accordance with the standard UNE-EN-ISO
9001:2000 requirements. The quality management system has been certified by
AENOR, the Spanish Association of Standardization and Certification. The
company also holds a licence for use of the ‘Madrid Excelente’ seal of
guarantee, issued by the regional government of Madrid and based on the
EFQM model of management.
The quality management system has the same quality manual and
standard operating procedures for each unit. These standard operating
procedures include reception, inventory and registry of materials,
compounding and quality control of each radiopharmaceutical, expedition of
materials, and maintenance, verification and calibration of equipment. There
are also general standard operating procedures for audits, customers and
process flow management, etc.
Quality assurance performance is measured regularly by a set of indexes.
One of the most important indexes is punctuality. It measures the percentage of
on-time deliveries. This index is measured every month and it is compared with
the goal of achieving a 99% success rate. In some units, the average punctuality
index is better than 98%.
5. E-BUSINESS SYSTEM
Customers can place their orders easily through the internet. The only
software needed is an internet navigator, either Netscape or Explorer. Each
hospital can have several users with their own user name and password. The
access is made through a secure connection.
The order web page is user friendly. The only field that must be typed in is
the name of the doctor making the prescription. Scroll bars manage all the
416
SESSION 14
other fields. Once the product is selected from a different list for each
customer, it is only necessary to set activity at the calibration time and delivery
time and date. The system automatically proposes the agreed delivery time.
Each order can have different lines, each line being a different product.
Filling in the name in the patient field is an option for the customer. The
database is designed to comply with the confidentiality required for these types
of data. Some of Molypharma’s customers leave it blank and others put in
either the real name of the patient or another kind of patient identification,
such as a clinical history number. Each order is automatically downloaded to
the computer system located in each radiopharmacy, avoiding possible human
errors.
In addition to order management, this e-business system (known as
Molysic) can handle a lot of valuable information for the customer. Customers
can check the status of pending orders and orders already delivered and have
access to a set of predesigned queries such as a list of products and a list of
delivered activity by date and isotope, etc. This query is very useful for
complying with the requirements of radioactive regulatory authorities.
Customers can rapidly have access to the identity of each unit dose. Just
by typing the identification number of a single dose, the system delivers the
following information: order number, container identification, delivery note
number, worker identification (who prepared the dose), identification of eluted
and labelled vials, expiry date, volume, patient identification, etc. The system
provides all this information in an error free way, as a bar code reader controls
every step of preparation.
Molyfact is the invoicing side of the e-business system. It gets all the
information from Molysic and allows the company to invoice each hospital with
their own requests. It can invoice for each delivery or over some defined
period, such as a week, a month, and so on. It also provides each customer with
statistics about its consumption in euros, not in units.
417
OYARZÁBAL and JIMÉNEZ-SHAW
— Identifies each single dose via bar coded labels and all the data required
by current legislation.
— Minimizes the possibility of human error, owing to the extensive use of
bar codes on vials, containers, processes and operators.
7. HUMAN RESOURCES
418
SESSION 14
8. DISTRIBUTION
Unit dose syringes are conditioned inside a sterilized envelope with their
label. Several unit doses can be delivered in a type ‘A’ certified package. The
company has designed and certified that the package contains a 4 mm lead
shield covered by a stainless steel container (127 mm × 127 mm × 270 mm),
with handle, reinforced hinges and closure system, two-piece expanded
polystyrene padding, plastic drum, metallic lid, closure ring and safety
strapping.
The pharmacy also manages the waste of some hospitals. Hospital
personnel dispose of used syringes in a biohazard plastic container. This
container is situated within a protective shield. After the container is closed
and measured as an exempt radioactive package, it is sent to the pharmacy’s
waste room. Once it is considered non-radioactive, it is disposed of as conven-
tional biohazard waste by a specialized company.
In order to focus on the radiopharmacy business and to provide the best
possible service to the customer, physical distribution is subcontracted to a
specialized multimodal transport company that operates in several dangerous
goods sectors. The transport company has its own vans and drivers. This
scheme requires very close collaboration between the radiopharmacy company
and the transport company, and several common procedures have been
developed and implemented.
A hospital representative, usually a nuclear medicine technician, signs
delivery notes, indicating the actual time of receipt. This time is introduced
subsequently into the system to obtain the punctuality index.
9. CONCLUSION
419
OYARZÁBAL and JIMÉNEZ-SHAW
420
CHAIRPERSONS OF SESSIONS
421
SECRETARIAT OF THE SYMPOSIUM
PROGRAMME COMMITTEE
M. DONDI
M. HAJI-SAEID
R.S. KAMEL
M.R.A. PILLAI
N. RAMAMOORTHY (Chairperson)
K.K. SOLANKI
422
LIST OF PARTICIPANTS
423
LIST OF PARTICIPANTS
424
LIST OF PARTICIPANTS
425
LIST OF PARTICIPANTS
426
LIST OF PARTICIPANTS
427
LIST OF PARTICIPANTS
428
LIST OF PARTICIPANTS
429
LIST OF PARTICIPANTS
430
LIST OF PARTICIPANTS
431
LIST OF PARTICIPANTS
432
LIST OF PARTICIPANTS
433
LIST OF PARTICIPANTS
Fresvig, M. GE Healthcare,
Amersham Health,
P.O. Box 4220 Nydalen,
0401 Oslo, Norway
Fax: +4723186025
Email: marianne.fresvig@ge.com
434
LIST OF PARTICIPANTS
435
LIST OF PARTICIPANTS
436
LIST OF PARTICIPANTS
Husbyn, M. GE Healthcare,
Amersham Health,
P.O. Box 4220 Nydalen,
0401 Oslo, Norway
Fax: +4723186025
Email: mette.husbyn@ge.com
Jahren, G. GE Healthcare,
Amersham Health,
P.O. Box 4220 Nydalen,
0401 Oslo, Norway
Fax: +4732186026
Email: grete.jahren@ge.com
437
LIST OF PARTICIPANTS
Jiménez-Shaw, R. Molypharma,
Dr. Severo Ochoa 37-4-1,
28100 Alcobendas,
Madrid, Spain
Fax: +34914905740
Email: rjs@molypharma.es
438
LIST OF PARTICIPANTS
439
LIST OF PARTICIPANTS
440
LIST OF PARTICIPANTS
441
LIST OF PARTICIPANTS
442
LIST OF PARTICIPANTS
443
LIST OF PARTICIPANTS
444
LIST OF PARTICIPANTS
445
LIST OF PARTICIPANTS
446
LIST OF PARTICIPANTS
447
LIST OF PARTICIPANTS
448
LIST OF PARTICIPANTS
449
LIST OF PARTICIPANTS
450
LIST OF PARTICIPANTS
451
LIST OF PARTICIPANTS
452
LIST OF PARTICIPANTS
453
LIST OF PARTICIPANTS
454
LIST OF PARTICIPANTS
455
LIST OF PARTICIPANTS
456
LIST OF PARTICIPANTS
457
LIST OF PARTICIPANTS
458
LIST OF PARTICIPANTS
459
.
AUTHOR INDEX
461
AUTHOR INDEX
462
AUTHOR INDEX
463
AUTHOR INDEX
464