cerebralperfusionimagingtracersforspectjnm.snmjournals.org/content/36/3/359.full.pdf2.pardridgewm,landawem.tracerkineticmodelofblood-brainbarrier...

6
ticals, since correlation found between PET or micro spheres and SPEC!' blood flow agents has demonstrated that the relationship is curvilinear. Recently, several re searchers suggested that the rising asymptotic plateau is caused by washout from high-flow regions (3,4). The up take, extraction fraction and retention of the agents used vaiy among different regions of normal and abnormal brain (1) and the fixation can be altered by local or regional parameters. The kinetic behavior and trapping mechanism specific to each tracer must be well understood for the cm ical application and interpretation of SPECT images. Tech netium-99m-d,1HMPAO (exametazime) and @Fc-1,1 ECD (bicisate) are both available and easy to label, however, HMPAO has poor in vitro stability (RP = 85% after 30 min) (5) and susceptibility to radiolysis, whereas ECD is much morestable(RP = 96%at 6 hr)(6). These technetium-amines demonstrate in vivo instability following blood contact and have a short arterial input of the bioavailable compound (7-9). The moiety of compoundschangesin circulating blood, which results in relative'y poor extraction (E) by the brain (HMPAO 77%, ECD 60%) (Table 1). The retention in the brain parenchymais linked to the enzymatic reactions with glutathione (GSH) for HMPAO (10,11) and with ste reospecific deesterification to acid derivatives for ECD (12— 15). These mechanisms have slow turnover and yield non diffusible polar metabolites. The first-passextraction rate is flow-dependentfor ECD, similar to HMPAO, and results in decreased uptake in the higher flow of a normal range (16). Conversely, extraction is higherwhen the flow is reduced. Due to a low backflux from brain (k2), the retention of ECD is higher than HMPAO (Table 1). Backdiffusionof HMPAO is more pro nounced in high flow regions and a higher extraction in low flow regions results in poor contrast. Technetium-99m- ECD images hold a slightly better contrast between high and low flow regions. ECD has some washout from the brain and negligible redistribution (17,18). HMPAO has slow clearance and liver accumulation contrary to ECD, which has rapid blood clearance with renal excretion of acid metabolites (EC) yielding a better signal-to-noise ratio. IMP, which is not readily available in every country and is cumbersome to label, was the first iodo-amine (19) used for brain imaging with a high brain extraction rate (96%) J NuciMed1995;36:359-363 R adionucide imaging and semiquantitationof cerebral perfusion by SPEC!' are important issues in patients with neurological and psychiatric diseases. The search for an ideal brain SPECT radiopharmaceuticalis complicated and competes with other imaging modalities such as dy namic CT and MRI. Unfortunately, currently available radiopharmaceuticals are far from ideal. The ideal agent should be commercially available, easy to label, possess in vivo and in vitro stability, have short arterialinput to facilitate intervention (stimulation)studies and a high extraction fraction at the first pass. Such an agent should reach a steady state in the brain quickly and be able to penetrate intact blood-brain barriers regardless of the blood cell-plasma protein partition. They also must have high passive retention in the brain independent of a metabolic or enzymatic system or a specific binding site. An ideal agent would also reflect regional distribution, which is linear to the cerebral blood flow over a wide range and remain fixed in the brain with a constant pattern to permit SPECT imaging with minimal extra brain activity. The ideal radiopharmaceutical should be neutral, ii pophilic (log p octanol/water between 1.0—2.5) and of small size (under 500 dalton) (1). All of the technetium-amines and iodo-amines discussed in this editorial[hexemethylpro pyleneamine oxime (HMPAO), bicisate (1,1-ECD) or iso propyl iodoamphatamine (IMP)J meet these criteria at different levels oflipophilicity, which determines biodistri bution. At higher levels of lipophilicity, there is a trade-off between binding to the brain cells and plasma proteins in favor of the latter (2). A @‘Fc-labeled compound that behaves like IMP would represent a major advance for routinebrainperfusionimaging.However, a consensus has not yet been reached on the best agent. A clear understandingof the absence of a trueflow agent is crucial before comparing the available radiopharmaceu ReceivedOct18,1994;acceptedOct.24,1995. Forcorrespondence orreprints onnt@Jean-LieMcmlii, MD,PhD,Med@ine Nucieaire, Ho@ Avicenne, 99009Bobigny, France. Cerebral Pertu@on ImagingTraces forSPECT• Morethetal. 359 Cerebral Perfusion Imaging Tracers for SPECT: Which One to Choose? J.L. Moretti, M. Caglar and P. Weinmann HopitalAvicenne, CHU Leonard de Vsni4 Bob@zy, France by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from

Upload: phamhanh

Post on 14-Apr-2018

220 views

Category:

Documents


3 download

TRANSCRIPT

ticals, since correlation found between PET or microspheres and SPEC!' blood flow agents has demonstratedthat the relationship is curvilinear. Recently, several researchers suggested that the rising asymptotic plateau iscaused by washout from high-flow regions (3,4). The up

take, extraction fraction and retention of the agents usedvaiy among different regions of normal and abnormal brain(1) and the fixation can be altered by local or regionalparameters. The kinetic behavior and trapping mechanismspecific to each tracer must be well understood for the cmical application and interpretation of SPECT images. Technetium-99m-d,1HMPAO (exametazime) and @Fc-1,1ECD(bicisate) are both available and easy to label, however,HMPAO has poor in vitro stability (RP = 85% after 30 min)(5) and susceptibility to radiolysis, whereas ECD is muchmorestable(RP = 96%at6 hr)(6). These technetium-aminesdemonstrate in vivo instability following blood contact andhave a short arterial input of the bioavailable compound(7-9). The moiety of compoundschangesin circulatingblood, which results in relative'y poor extraction (E) by thebrain (HMPAO 77%, ECD 60%) (Table 1). The retention inthe brain parenchymais linked to the enzymatic reactionswith glutathione (GSH) for HMPAO (10,11) and with stereospecific deesterification to acid derivatives for ECD (12—15). These mechanisms have slow turnover and yield nondiffusible polar metabolites.

The first-passextractionrate is flow-dependentfor ECD,similar to HMPAO, and results in decreased uptake in thehigher flow of a normal range (16). Conversely, extractionis higherwhen the flow is reduced. Due to a low backfluxfrom brain (k2), the retention of ECD is higher thanHMPAO (Table 1). Backdiffusionof HMPAO is more pronounced in high flow regions and a higher extraction in lowflow regions results in poor contrast. Technetium-99m-ECD images hold a slightly better contrast between highand low flow regions. ECD has some washout from thebrain and negligible redistribution (17,18). HMPAO hasslow clearance and liver accumulation contrary to ECD,which has rapid blood clearance with renal excretion ofacid metabolites (EC) yielding a better signal-to-noiseratio.

IMP, which is not readily available in every country andis cumbersome to label, was the first iodo-amine (19) usedfor brain imagingwith a high brain extraction rate (96%)

J NuciMed1995;36:359-363

R adionucide imagingand semiquantitationof cerebralperfusion by SPEC!' are important issues in patients

with neurological and psychiatric diseases. The search foran ideal brain SPECT radiopharmaceuticalis complicatedand competes with other imaging modalities such as dynamic CT and MRI. Unfortunately, currently availableradiopharmaceuticals are far from ideal.

The ideal agent should be commercially available, easyto label, possess in vivo and in vitro stability, have shortarterialinput to facilitate intervention (stimulation)studiesand a high extraction fraction at the first pass. Such anagent should reach a steady state in the brain quickly andbe able to penetrate intact blood-brain barriers regardlessof the blood cell-plasma protein partition. They also musthave high passive retention in the brain independent of ametabolic or enzymatic system or a specific binding site.An ideal agent would also reflect regional distribution,which is linear to the cerebral blood flow over a wide rangeand remain fixed in the brain with a constant pattern topermit SPECT imaging with minimal extra brain activity.

The ideal radiopharmaceutical should be neutral, iipophilic (log p octanol/water between 1.0—2.5)and of smallsize (under 500 dalton) (1). All of the technetium-aminesandiodo-aminesdiscussed in this editorial[hexemethylpropyleneamine oxime (HMPAO), bicisate (1,1-ECD) or isopropyl iodoamphatamine (IMP)J meet these criteria atdifferent levels oflipophilicity, which determines biodistribution. At higher levels of lipophilicity, there is a trade-offbetween binding to the brain cells and plasma proteins infavor of the latter (2). A @‘Fc-labeledcompound thatbehaves like IMP would represent a major advance forroutinebrainperfusionimaging.However, a consensus hasnot yet been reached on the best agent.

A clear understandingof the absence of a trueflow agentis crucial before comparingthe available radiopharmaceu

ReceivedOct 18,1994;acceptedOct.24,1995.Forcorrespondenceorreprintsonnt@Jean-LieMcmlii,MD,PhD,Med@ine

Nucieaire,Ho@ Avicenne,99009Bobigny,France.

CerebralPertu@onImagingTracesforSPECT•Morethetal. 359

Cerebral Perfusion Imaging Tracers for SPECT:Which One to Choose?

J.L. Moretti, M. Caglar and P. Weinmann

HopitalAvicenne, CHU Leonard de Vsni4 Bob@zy, France

by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from

TABLE IComparisonofBrainSPECTImagingAgents

nism and metabolism of the tracer. This washin/washoutbalance is clearly demonstrated in the study performed byNishizawa et al. (24) who observed a temporal change ofradioactivity among normal brain structures. Likewise,IMP uptake is different between white and gray matter andfavors the latter. The first possible explanation for this isthe first come, first served principle. Since the penetratingcapillaries enter the gray matter, more tracer is available tobe extracted by gray matter before reaching the whitematter. The second explanation is the difference in theextraction coefficient of IMP between gray and white matter (21). Rapinet al. demonstratedby autoradiographythatthe activity in the brain redistributeswith time in favor ofthe white matter possibly due to backflux from gray towhite matter (25). For this reason, it is crucial to performearly imaging at 20 min. Afterwards, polar metabolitessuch as p-iodobenzoic acid (PIBA) will appear in theplasma in non-negligibleamounts with time (26), diffusingpassively through a damaged blood-brain barrier. Therefore the delayed images have to be acquired at 3 hr.

One of the most active and exciting fields of nuclearmedicine has been evaluatingcerebralperfusion in severalpathologies. There are certain conditions where differentlipophilic tracers give similar images such as drug addiction, dementia, interictal epilepsy, psychiatric disorders

+ and head trauma. Ictal, postictal epilepsy, stimulations and

stroke are not among these conditions.

ICTAL AND POSflCTAL EPILEPSY

Interictal perfusion SPECT was performed with iodoamines (27) andTc-amines (28)yielding a sensitivity of 60%to75%whichcorrelateswellwithEEGstereolocalization.Fluctuations in perfusion and metabolism in the interictalperiod do not allow the detection of more than 75% epileptic hypoactive foci with perfusion SPECF or FDG-PET(29). During the ictal phase, higher metabolic activity coupled with high perfusion results in increased uptake in theepileptic foci. True ictal studies are difficult to perform invitro with unstable HMPAO, rendering it impossible tostore in the neurology department. In this case, compounds of higher stability such as IMP or ECD are moreappropriate. In the early postictal phase, cerebral bloodflow remains elevated decoupling from the rapidly dccliiiing glucose metabolism (30). Blood flow remains elevatedlong enough to allow lipophilictracers to detect the hyperactive foci. Since ECD has a shorter arterial input, it appears to be more appropriate than IMP. Sensitivity increases by 10% during the immediate postictal phase (31).

S11MULA11ONIn normal cerebral autoregulation, cerebral blood flow

increases in response to neuronal metabolism; SPED.' hasdemonstrated sufficient sensitivity to detect changes in activity related to stimulation of metabolic activity (32).These activation tasks must generate detectable and reproducible changes in cerebral blood flow which are sustained

CharecteristlcIMPHMPAOECDLogP1.401.901.64Purity96%85%96%Stabllfty6hr30mm6hrHead

uptake (%lD)6.0 ±15.2 ±16.3 ±1.9BUI1137773E%907760ki0.430.26

0.27t0.290.25tk20.0140.60*

0.59t0.220.19tA

= [email protected]*0.46t1.31*0.31tk30.930.58*

[email protected]@ak2ik3700.9r

[email protected]@R%98%50%@

49%t72%75%tE%88365k

37.7k43.2*45tK0.420.13*

[email protected]*O.l9@RediStributiOn++100t(mln)201010Meanwashoutrateof1%2%10%steady

state(hr)Heterogeneouswashout + 0

*SeeReferences49,50and51. SeeReference48for IMPdatatSee References16 and 51.kl = unidirectIonalinfluxconstantcorrespordngtoflowx extraction

(firstpass)(Fx g1m@1;@=back-flwinh/rT@,k1/k2=pardtion@e1ficientinthecoitex@k3= transformationconstant(1/mm),a k3@2

= conversion rate, R = retention fr@ion = k3/k2 + k3, K = steady

state influxconstant= K1.@ + K@,/E = extr@ion at first pass,E =E*R Extractionat steadystate,BUI = bran u@akeindex@logP =dedmaiioga@thmofthepartitioncoefficientoctanolonwater,t = timetoreachthesteadystate,W = washoutrateaftersteadystate(hr1) frombrain.

(Table 1). Its uptake linearly follows a wide range of flowassessed by microspheres (20). The gradual increase in thebrain uptake of IMP (50%at 5 miii and 90% at 20 min) islinked to its clearance from the lung, which functions as abuffer releasing the tracer into the circulation (21). Thestereoselective retention mechanism of IMP in brain tissueinvolves a possible conversion to hydrophilic metabolites(22), and an affinity to high capacity and relatively nonspecific binding sites. Intracellular pH can play a role in manyinstances: (1) rate of exchange through the BBB; (2)strength of binding to the nonspecific sites of the brain; and(3)metabolicconversionrate of aminesthrough the enzymatic system linked with binding sites. When blood pH isdecreased due to local lactate discharge, the uptake of IMPis notably decreased (23).

Brainretention of IMPreflects the balance of the washinandwashout ofthe tracerin individualstructures.Washoutfrom brain is influenced by blood flow, retention mecha

360 TheJournalofNuclearMediane•Vol.36•No.3•March1995

by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from

during the period after injection, while the tracer reachessteady state. In the activation studies, the tracer distribution has to reflect high flow rates, like IMP, in order toquantify the stimulation accurately. The trapping processof the tracer should not be saturable in test/retest studiesusing split dose technique within a short interval. In thetest/retest studies, assumption is that the original distribution pattern is still the same during the second SPECFimage acquiredafteractivationon the same day. Due to theheterogeneous washout pattern, a test/retest protocolwould introduce significant errors, therefore a two dayprotocol is preferable (33).

As the radioactivity of d,1 HMPAO changes with timeafterpreparation(in vitro and in vivo), even if equal dosesof radioactivity are injected, one cannot be sure of thesame bioavailabiity for brain. Using HMPAO in a test/retest activation study requires a long stimulation of thesame intensity. ECD has the same uptake and retentioncharacteristics of HMPAO, but it is more stable and moreconvenient to handle. Like HMPAO, however it underestimates high flow areas (5).

CEREBROVASCULARDISEASE

Tomographic images of cerebral perfusion with 11-pophilic radiotracers are extremely useful in early detection of acute and chronic stroke. SPECF is superiorto Cl'in detecting ischemia since it reflects alterations in perfusion before structuralchanges occur. This is an advantagefor determining the appropriate therapy, which must beinitiated as early as possible in acute stroke.

In stroke, two differentiatedregions are observed in thebrain: one the central infarct core known as the structuralabnormality, which can be imaged by CF and two a peripheralarea that reflects pen-infarct ischemia, known asthe penumbral zone (34). SPECT perfusion imaging candemonstrateboth of these regions dependingon the tracerused. There is a correlation between the penumbralzoneand the severity of the neurological deficit which may berelated to outcome (35).

Because of the high extraction rate in low-flow regionsand the high level of perfusion in ischemic regions due tovasodilation, HMPAO cannot be reliable to detect periinfarct ischemia in the subacute phase (4—15days). If images are acquired at 4 hr, clearance of the plasma activityand leakage of the tracer from the cells allows imagingofhypoactive peri-infarct ischemia (36). On the contrary,early IMP images demonstrate both structural abnormalities (40% decrease) and the penumbral zone (15% decrease). With time, even if the central area remains unchanged, redistribution occurs in the peripheral zoneresulting in a partiallyfilled delayed images. The degree ofredistribution is correlated with the local cerebral bloodflow and regional consumption of oxygen but not with theregional oxygen extraction fraction which reflects viability(37). IMP redistribution indicates a tissue with low CBFand a remainingmetabolism (38). This implies that blood

flow is inadequaterelative to the energy/metabolicdemandfor oxygen and substrate (misery perfusion). In these instances, slower metabolism and washout of IMP wouldcause retentionin the pen-infarctzone. The only definitiveconclusions made on redistribution IMP SPEC!' images iswhether non-necrotic cells are present or not. The redistribution of IMP was formerly related to a good clinicaloutcome (39).

With ECD in the acute phase of stroke, when the esterase enzyme system is still functioning, a higher extractionand retention of the tracer results in low blood flow areas(40). In the subacute phase, hypoxia may alter the functionof esterase, resulting in minimalretention, even if the extraction is preserved. The hypoactive area will be largerand deeper on delayed images, demonstrating increasedclearance from the ischemic areas (41,42). Diminished activity of ECD in subacute stroke may be due to the lack of

oxygen and enzyme activity, which causes an accumulation of unmetabolizedtracercandidate to leak out from theperipheral ischemic zone. By contrast, at low blood flow,since the GSH system is more resistant to hypoxia andcontinues to function, cells retain HMPAO in the ischemicarea.

During the period of luxury perfusion after recanalization, there is transientdecouplingof metabolic demandandcerebral blood flow in the subacute phase of ischemic regions. Increased glucose consumption is observed, indicatinganaerobic glycolysis and production of lactate accompaniedby localacidosisallofwhichdecreasetheuptakeof IMP.Inthis condition, the behavior of the perfusion tracers is different. Most of the time HMPAOshows a focal areaof hyperactivity, whereas IMP and ECD demonstrate hypoactivezones and IMPis relatedto low extractionin acidicpH andECD caused by low retention, which is linked to alteredesterase function in hypoxia (43,44).

In reversible ischemic stroke, structuralimaginghas litlie to offer and perfusion imaginghas low sensitivity (60%within the first 24 hr (45), declining further with time). Toincrease sensitivity, it is necessary to assess vasodilatoryreserve by an acetazolamide stimulation test as demonstrated by us with IMP (46,47). The weak contrast obtamed with HMPAO and ECD in very mild ischemia is adrawback incorrectly delineating lesions. Because activityof these @“@Tc-labeledamines is not linear at high flowrates, the response to acetazolamide is blunted as is lesioncontrast.

CONCLUSION

Although IMP because of its linearity with flow andcapacity to depict mild ischemic regions seems to remainthe reference in assessing cerebrovascular diseases, ECD,due to its availability and improved stability, will likelybecome widely used even though it is far from being anideal chemical microsphere agent. Technetium-99m-ECDis a powerful diagnostic tool to facilitate critical management decisions in neurologic and psychiatric diseases. It is

361Cerebral Perfusion ImagingTraces for SPECT •Morettiat al.

by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from

necessary to be cautious, however, in using this tracer inextreme conditions oflow and high brain blood flow and intest/retest examinations. Iodune-123-IMPhas not been replaced by Tc-labeled amines. The search continues for anoptimal brain tracer of perfusion and viability as a reliablefirst step for functional and receptor imaging. Although thetask to find the ideal agent is difficult, the challenge isworthwhile. With early diagnosis of cerebrovascular disease, there is an opportunity for improved outcome.

REFERENCES1. KungHF, OhmoinoY, KungMP.Currentandfutureradiopharmaceuticals

forbrain imagingwith singlephotonemissioncomputed tomography.SeminNuciMed 1990;20,4:290-302.

2. PardridgeWM, Landaw EM. Tracer kineticmodelof blood-brainbarriertransportof plasmaprotein-bondligands.I ClinInvest1984;74:745—752.

3. GemmellHG,EvansNi'S,BessoniAO,etal.Regonalcerebralbloodflowimaging:a quantitativecomparisonof technetium-99m-HMPAOSPECFwith C'502PET.I NuciMed1990;31:1595-1600.

4. YonekuraY, NishizawaS, Mukai1, Ctal. SPECFwith(@“Tc).d,1-hex.amethyl-propylene amine oxime (HM-PAO) comparedwith regional carebral blood flow measuredby PET: effects of linearisation. I Cereb BloodFlowMetal, 1988;8:S82-S89.

5. LucignaniG,RosettiC,FerrarioP,etat.InvivometabolismandkineticsofTc-99m HMPAO. EurJ NucI Med 1990:249-255.

6. Knudsen GM, Andersen AR, Sommier FE, Videbaek C, Hasselbach S,Paulson OB. Brain extraction and distribution of @Fc-bicisatein humansand in rats. I CerebBlood Flow Met 1994;14(suppl.1):S12-S18.

7. Matsuda H, Oba H, Seki H, Ctal. Determination offlow and rate constantsin a kinetic model of [@“[email protected] oxime in thehumanbrain.I CerebBloodFlowMetal, 1988;8:S61-S68.

8. TurbergenK, CorlijaM, VolkertWA, HolmesRA.Sensitivityof technetium-99m.d,1-HMPAO to radiolysis in aqueous solutions.JNuclMed 1991;32:111—115.

9. SuzukiS. SakaiF, Akutsu1, TazakiY. TracerKineticsof ‘@I.IMP,Tc-99m-HM-PAO and Tc-99m-ECD: measurements oftemporal changes inarterial andjugular venous radioactivity. I CerebBlood Flow Metab 1991;11(suppl.2):S774.

10.LassenNA, AndersenAR, FribergL, PaulsonOB. The retentionof[@rcJ.d,1.HM.PAOinthehumanbrainafterintracarotidboiusinjection:a kineticanalysis.I CerebBloodFlowMetab1988;8:S13-S22.

11.NeirinckxRD,BurkeiF, HarrisonRC,ForsterAM,AndersenAR,LassenA. The retentionmechanismof technetium-99m-FIM-PAO:intracellularreaction with glutathione. I Cei@bBlood Flow Metab 1988;8:S4-S12.

12. VallabhajosulaS, ZimmermanRE, PicardM, et aLTechnetiwn.99mECD:a newbrainunagingagent:invivokineticsandbiodistributionstudiesinnormalhumansubjects.INucI Med1989;30:599-604.

13.HotmanLB,HeRmanPS,GoldshmithSi, etal.Biodistril,ution,dosünetiyand clinical evaluation of technetium-99methyl cysteinatedimer in normalsubjectsand in patientswith chronic cerebral infarction. JNuclMed 1989;30:1018—1024.

14.DevousMD, SrKellyPayne3,LoweJL,LeroyRF.Comparisonoftechnetium-99m-ECDto xenon-133SPECFin normalcontrolsandin patientswith mild to moderate regional cerebral blood flow abnormalities.I NucIMed 1993;34:754—761.

15.LeveiHe3,DemonceauG,WalovitchRC.Intrasubjectcomparisonbetweentechnetium-99m-ECDandtechnetium-99m-HMPAOin healthyhumansubjects.INuci Med199233:480-484.

16.FnbergL, MdersenAR, LassenNA, Hohn 5, DamM. Retentionof@Tc-bicisatein the humanbrain after intracarotid injection.JCe,@bBlood

FlowMetal.,1994;14(suppl.1):519—S27.17.WalovitchRC,FranceschiM, PicardM, etal.Metabolismof @Fc-L,L

ethylcysteinatedimerin healthyvoiunteers.Newvplzwmacology199130:283-292.

18.WalovitchRC,CheesmanEH, MaheuLi, HallKM. Studiesoftheretention mechanismof the brain perfusion imagingagent @Fc-bicisate(@cECD).I CerebBloodFlowMetab1994;14(suppl:1):54-S11.

19.WinchellHS,BaldwinRM,LinTH. Developmentof1-123labeledaminesfor brain studies localization ofl-123 iodophenyl alkyl aminesin rat brain.JNucl Med 1980;21:940-946.

20. KuhI DE, Barrio JR. Huang SC, et aL Quantifying local cerebralblood flow

by N isotropyl p 123 iodoamphetamine(IMP) tomography. I Nuci Med1982;196—203.

21. YonekuraY, Nishizawa5, Mukai1, etat.Functionalmappingofflowandback-diffusion rate of N-isopropyl-p-iodoamphetamine in human brain. INuclMed1993;34:839-844.

22. Kung HF, Blau M. Regional intracellular pHshift: a proposednew mechanismforradio-pharmaceutical uptakein brain andother tissues 1NucI Med1980;21:147—152.

23. Oldendorf WH. Lipid solubility and drug penetration of the blood-brainbarrier.P@@ocSocEaplBioMed1974;147:813—816.

24. Nishizawa 5, Tanada 5, Yonekura Y, et al. Re@onaldynamics of N-isotropyI.(@I)-iodo-amphétaminein humanbrain.I CerebBboodMetab1989;3@U50-156.

25. RapinJR, Duterte D, Le Poncin-Lalitte M, LageronA, MonmaurP, Rips R,LassenNA. lodoamphetaminederivativeastracersfor cerebralbloodflowor not? Autoradiography an autochistoradiographie studies. I Cereb BloodMetab19833(suppl1):S105-S106.

26. Ronald M, Baldwin RM, Wu JL. In vivo chemistiy of iofetamine HCLiodine.123(IMP).JNucl Med 1988;29:122-124.

27. LeeBL,MarkandON,WellmanHN,etal.HIPDM-SPECTinpatientswithmedically intractable complex partial seizures. Ictal study. Arch Newvl1988;45:397—402.

28. Stefan HKC, Biersack HJ, Reichmann K. Initial experience with @“Tchexamethyl-propyleneamineoxiine (HMPAO SPECFÜIpatientawith focalepilepsy).EpilepsyRes1987;1:134—138.

29. Theodore W, Newmark M, Sato 5, et al. F-18-fluorodeoxyglucosePET inrefractory complex partial seizures.Ann Neurol 1983;14:429-437.

30. Theodore WH, Fishbein D, Dubinsky R. Patternsof cerebral glucosemetabolism in patientswith partial seizures.Neurology 1988;38:1201-1206.

31. RoweCC,BerkovicSF,SiaSIB, etal.LocalizationofepilepticfociwithpostictalSPECT.AnnNeuml1989;26:660-668.

32. Ring HA, GeorgeM, Costa D@ Ell Pi. The use of cerebralactivationprocedureswith SPECT.EurlNuclMed 1991;18:133-.141.

33. HohnS, MadsenPL, SparlingB, LassenNA. Useof @Fc-bicisateinactivation studiesby [email protected] 1994;14(suppi1):5115—S120.

34. Raynaud C, Rancurel G, Samson Y, et al. Patholophysiologic study ofchronicinfarctswith 1-123isopropyliodo-amphetamine(IMP):theimportanceofperiinfarctarea.Stroke1987;18:21-29.

35. MountzJM, Model JO, FosterNL, et al. Prognosticationof recoveiyfollowingstrokeusingthecomparisonofCF andtechnetium-99mHMPAOSPECr. JNuclMed 1990@,31:61-66.

36. HayashidaK, Nishimura 1, Imakita S, Uehara 1. Filling out phenomenonwith technitium-99mHMPAO SPEC!' at the site of mild cerebral ischemia.JNuclMed1989;30:591-598.

37. TsukudaM, KuwabaraY, IchiyaY, etaLEvaluationofthesignificanceofredistributionin 1-123IMP SPECFin cerebrovasculardisorders—acornparative study with PET. EurlNud Med 1989;15:746-749.

38. Matsuda H, Tsuji 5, Oba H, Ct al. Autoradiographic analysis of iodoamphetaminerodistril,ulion in experimentalbrain ischemia.JNuclMed 1990;31:660—667.

39. Defer 0, Moretti JL, CesarP. SergentA, RaynaudC, DegosD. Early anddelayedSPEC!'usingN-isopropyfp-iodoamphetamineiodine123in carebral ischernia.Arch Neurol 1987;44:715-718.

40. GreenbergJH, Araki N, Karp A. Cormlation between @Fc-bidsateandregional CBF measuredwith iodo-[14C] antipyrine in a primate focal isch.erniamodel.I CerebBloodMetal,1994;14(suppl1):536-543.

41. MorettiJL,Defer0, TamgacF, WeinmannP,5dm c, CesarP.Comparison of brain SPECF using @Fc-bicisata(L,L-ECD) and [‘@I]IMPincortical and subcortical strokes. I Cereb Blood Metab 1994;14(suppl1):584-590.

42. MorettiJL,TamgacF, WeinmannP,etal.EarlyanddelayedbrainSPECFwith @Fc-ECDand‘@I.IMPin subacutestrokes.I NuclMed 1994;35:1444—1449.

43. ShishidoF, UemuraK, MurakamiM, et al. Cerebraluptakeof @Fcbicisate in patients with cerebrovasculardiseasein comparison with CBFandCMRO2measuredby @tronemissiontomography.I CerebBloodMetal,1994;14(suppl1):S66—S75.

44. NakagawaraJ, Nakamura ii, Takeda R, et al. Assessmentof postichemicreperfusion anddiamox activation test in stroke using @Fc-ECDSPECF.I CerebBloodMetal, 1994;14(suppl1)S49-S57.

45. HartinannA. Prolongeddisturbancesof regionalcerebralbloodflowintransient ischernic attacks. Stroke 1985;16:932-939.

46. Devous MD Sr, &tjer HH, Ajmani AK, SamsonDS, Bonte FJ. SPECF

362 TheJournalofNudearMedicine•Vol.36•No.3•March1995

by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from

measurements of cerebrovascular reserve in patients with hemodynamicrisk. I NuclMed 1988;29:911.

47. CesaroP, MoreniJL, CaronJP,et al. TomoscintigraphiecérébraleutilisantI'iodoisopropylamphétamine123-I:intérétenpathologicischémiquecérébral.La PresseMed 1985;144:205.

48. Yonekura Y, Mukai T, lwasaki Y, et al. Kinetic analysisof N-isopropyl-piodoamphetaminein humanbrainIAbstractl.JNuclMed 1991;3l:991.

49. AndersenAR, FribergH, LassenNA, KristensenK, NeirinckxRD.As

Scatter(Continuedfrompage3A)

sessmentof the arterial input curve for (@“1'cJ-d,1-HMPA0by rapid octanol extraction. I CerebBlood Flow Metal, 1988;8:S23-S30.

50. Murase K, Tanada5, Fujita H, Sakaki 5, Hamamoto K. Kinetic behaviorof technctium-99m-HMPAO in the human brain and quantification of carebral blood flow using dynamic SPECf. I NucI Med 199233:135-143.

51. TanadaS,MuraseK, lnoue1, etal.DeterminationofintracerebralkineticsofTc-99methylcysteinatedimer(ECD)withdynamicbrainSPECflAbstracti. J NuclMed 1991;991.

developmentof ‘231-MIBG,‘‘In-pentetreotideor otherreceptor-specificradiolabeledligands. Idid not tell them ofthe men and women who work in the middle ofthe night to produceanddeliver those magic bullets, or ofthose who work day after day in hospitals and officesobtaining images and data. Nor did I describe the wonder ofa gamma camera or my delightwhen watchingthree-dimensionalimage acquisition,volume reconstructionand fusion withMRIor CT images.Thereis neverenoughtimeto relatethe fascinationofwatchingthebrainthink,theheartpump,or themarvelsof otherorganfunction.

Nuclear medicine is more than a medical specialty. It is a wonder to behold.

Stanley J. Goldsmith, MD, Editor-in-ChiefTheJournalofNuclearMedicine

—March1995

363CerebralPerfusionImagingTracesfor SPECT•Morethet al.

by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from

1995;36:359-363.J Nucl Med.   J.L. Moretti, M. Caglar and P. Weinmann  Cerebral Perfusion Imaging Tracers for SPECT: Which One to Choose?

http://jnm.snmjournals.org/content/36/3/359.citationThis article and updated information are available at:

  http://jnm.snmjournals.org/site/subscriptions/online.xhtml

Information about subscriptions to JNM can be found at:  

http://jnm.snmjournals.org/site/misc/permission.xhtmlInformation about reproducing figures, tables, or other portions of this article can be found online at:

(Print ISSN: 0161-5505, Online ISSN: 2159-662X)1850 Samuel Morse Drive, Reston, VA 20190.SNMMI | Society of Nuclear Medicine and Molecular Imaging

is published monthly.The Journal of Nuclear Medicine

© Copyright 1995 SNMMI; all rights reserved.

by on June 2, 2018. For personal use only. jnm.snmjournals.org Downloaded from