神經醫學造影放射藥物發展

林口長庚醫院 分子影像中心 魏孝萍,林昆儒,閻紫宸 醫師

內部網頁原文下載 (PDF),進階閱讀:台大核醫科的 E-learning

前言

核子醫學使用半衰期適中的放射性同位素標幟藥物,配合先進造影設備及影像重組技術,已廣泛用於臨床檢查生理的改變與疾病的發展。由於所使用的放射性同位素衰變模式的差異因而發展出 正子掃描 (PET SCAN) 以及單光子放射電腦斷層造影 (single-photon emission computed tomography; SPECT)。用於 正子掃描 (PET SCAN) 的放射藥物所標幟的放射核種包括碳-11、氮-13、氧-15及氟-18等,衰變時會釋出正子; 正子掃描 (PET SCAN) 即以偵測正子與電子發生互毀反應 (annihilation) 釋出的 511 keV光子,由於偶合電路的設計使PET具備高靈敏度與高解析度的造影優勢。SPECT則使用鎝-99m、碘-123、鉈-201、鎵-67、銦-111等衰變時釋放加馬射線的放射核種標幟的放射藥物, SPECT攝影機需要.直儀 (collimator) 來消除散射加馬射線,造影的靈敏度與解析度不及PET,但設備與放射藥物的費用較 正子掃描 (PET SCAN) 低廉,對於當前核醫檢查仍佔有重要地位。

血腦屏障

早期核醫影像診斷腦部疾病僅限於檢查血腦屏障 (blood-brain barrier; BBB)的完整性,使用的放射藥物必須依賴疾病或外傷所引起的的BBB破損才能進入腦內並被檢出。1980年代發展的局部腦血流灌注造影 (brain perfusion imaging) 開始用到能透過BBB、隨腦血流分佈並滯留腦內的放射藥物,包括SPECT造影使用的鎝-99m-HMPAO [1]、鎝-99m-ECD [2]、碘-123-IMP [3],以及PET使用的氧-15水 [4] 等。隨著迴旋加速器 (cyclotron) 的普遍化,被譽為世紀分子的氟-18-FDG (氟化去氧葡萄糖) 也大量用於PET檢查疾病引起的腦部葡萄糖攝取與代謝差異 [5]。

神經受體 (receptor)及轉運體 (transporter) 造影

過去十年在發展中樞神經系統特定神經受體 (receptor)及轉運體 (transporter) 造影用放射藥劑有顯著進展。由於早期的PET應用以神經醫學研究為主,因此許多神經受體造影多由PET開始,再延伸到SPECT造影劑的發展與應用。腦多巴胺神經系統的放射性造影劑研發尤其蓬勃,此與帕金森氏病 (Parkinson’s disease; PD) 以及精神分裂症 (schizophrenia) 的病因及治療機制有關。氟-18-FDOPA [6]及放射性同位素標幟古柯鹼 (cocaine) 衍生物被用來檢查與帕金森氏病及動作障礙疾病有關的腦多巴胺神經退化;氟-18-FDOPA PET影像顯示多巴胺神經細胞合成多巴胺的生化機轉,而放射性同位素標幟古柯鹼類似物則為選擇性結合於多巴胺神經節前細胞膜上的轉運體,兩者攝取機制不同,但都能顯現多巴胺神經節前細胞的功能是否完整。這些放射性同位素標幟古柯鹼類似物包括用於PET造影的碳-11-FECIT [7]、氟-18-CFT [8]、氟-18-FPCIT [9]及氟-18-FECNT [10] 等,以及用於SPECT造影的碘-123-IPT [11]、碘-123-β-CIT [12]、碘-123-PE2I [13]、碘-123-FPCIT [14]及鎝-99m-TRODAT-1 [15]等;其中碘-123-FPCIT已於歐洲由GE HealthCare以DatSacn之名上市,鎝-99m-TRODAT-1 已於國內上市,國外由GE HealthCare進行臨床試驗中。

腦多巴胺神經系統

針對腦多巴胺神經節後細胞膜上的受體造影,可分為D1受體造影劑及D2/D3受體造影劑兩大類,其中又以D2/D3受體造影劑的研究最為普遍,此與D2受體為主要精神分裂症治療藥的作用位置有很大關聯。文獻發表的D1受體造影劑只有PET造影用的碳-11-SCH 23390 [16]及碳-11-NNC 756 [17];D2/D3受體造影劑則包括用於PET造影的碳-11-raclopride [18]、碳-11-NMSP [19]、氟-18-FMB [20]、氟-18-FIDA-2 [21]、氟-18-DMFP [22]等,以及用於SPECT造影的碘-123-IBZM [23]、碘-123-IBF [24]、碘-123-epidepride [25]、碘-123-FIDA-2 [21]等。

情緒與精神狀態有關的血清胺造影

與情緒與精神狀態有關的血清胺 (serotonin) 神經系統的造影研究,近年亦有顯著發展,許多憂鬱症治療藥物的作用即為抑制血清胺轉運體或調控血清胺受體的機能,因此以核分子影像研究血清胺神經系統的發展如雨後春筍。研究血清胺轉運體的放射性造影劑包括用於PET的碳-11-McN 5652 [26]、碳-11-β-CIT [27]、碳-11-nor-βCIT [28]、碳-11-ADAM [29]、碳-11-DAPA [30]、碳-11-AFM [31]、碳-11-DASB [32]、碳-11-MADAM [33]、氟-18-FADAM [34]等,以及用於SPECT造影的碘-123-β-CIT [35]、碘-123- nor-βCIT [36]、碘-123-ADAM [37]等。腦血清胺神經受體造影可分為 5-HT1A受體及5-HT2A受體兩大類造影劑。5-HT1A受體造影劑包括用於PET的碳-11-WAY 100635 [38]、碳-11-DWAY 100635 [39]、氟-18-MPPF [40]、氟-18-DMPPF [41]等;文獻報導鎝-99m-DWAY以體外自動顯影 (in-vitro autoradiography) 顯現5-HT1A受體的分佈,但無後續體內造影的進展。至於5-HT2A受體造影劑則包括用於PET的碳-11-NMSP [42]、碳-11-MDL 100907 [43]、氟-18-setoperone [44]、氟-18-altanserin [45]等,以及用於SPECT造影的碘-123-5-iodo-R91150 [46]。

失憶症 (dementia) 有關造影研究

與失憶症 (dementia) 有關造影研究近年集中在與阿爾茲海漠氏症 (Alzheimer's disease) 病理息息相關的 β-amyloid plaques及tangles的造影。氟-18-FDDNP [47]、碳-11-PIB [48]、碳-11-SB-13 [49]的陸續出現為分子影像診斷阿爾茲海漠氏症帶來曙光,其中碳-11-PIB已授權給GE HealthCare作為未來藥物治療阿爾茲海漠氏症的療效評估指標。由於碳-11-PIB的物理半衰期只有20分鐘,不利於較長時間的造影程序,因此未來勢必會有氟-18標幟藥物的後續發展。在SPECT造影劑的研發方面,以賓州大學發展的碘-123-IMPY [50]最具發展潛力。

隨著能表現各種神經學疾病的動物模式的陸續建立,加上高解析度的動物造影用PET、SPECT結合CT、MRI影像設備與技術的發展,核醫腦造影已不再侷限於臨床檢查的工具。藉由不斷推陳出新的放射性造影劑研發,未來藉由動物造影研究發展出用於人類診斷及治療的新技術已經成為分子影像醫學的願景。

參考文獻

  1. Ell PJ, Hocknell JM, Jarritt PH, et al. A 99Tcm-labelled radiotracer for the investigation of cerebral vascular disease. Nucl Med Commun 1985;6:437-441.
  2. Vallabhajosula S, Zimmerman RE, Picard M, et al. Technetium-99m ECD: a new brain imaging agent: in vivo kinetics and biodistribution studies in normal human subjects. J Nucl Med 1989;30:599-604.
  3. Greenberg JH, Kushner M, Rango M, Alavi A, Reivich M. Validation studies of iodine-123-iodoamphetamine as a cerebral blood flow tracer using emission tomography. J Nucl Med 1990;31:1364-1359.
  4. Mintun MA, Raichle ME, Martin WR, Herscovitch P. Brain oxygen utilization measured with O-15 radiotracers and positron emission tomography. J Nucl Med 1984;25:177-187.
  5. Phelps ME, Huang SC, Hoffman EJ, Selin C, Sokoloff L, Kuhl DE. Tomographic measurement of local cerebral glucose metabolic rate in humans with (F-18) 2-. uoro-2-deoxy-D-glucose: validation of method. Ann Neurol 1979;6:371-388.
  6. Firnau G, Garnett ES, Chirakal R, Sood S, Nahmias C, Schrobilgen G. [18F].uoro-L-dopa for the in vivo study of intracerebral dopamine. Int J Rad Appl Instrum [A] 1986;37:669-675.
  7. Antonini A, Moresco RM, Gobbo C, et al. Striatal dopaminergic denervation in early and late onset Parkinson's disease assessed by PET and the tracer [11C]FECIT: preliminary .ndings in one patient with autosomal recessive parkinsonism (Park2). Neurol Sci 2002;23 Suppl 2:S51-52.
  8. Laakso A, Bergman J, Haaparanta M, Vilkman H, Solin O, Hietala J. [18F]CFT [(18F)WIN 35,428], a radioligand to study the dopamine transporter with PET: characterization in human subjects. Synapse 1998;28:244-250.
  9. Lundkvist C, Halldin C, Ginovart N, Swahn CG, Farde L. [18F] β-CIT-FP is superior to [11C] β-CIT-FP for quantitation of the dopamine transporter. Nucl Med Biol 1997;24:621-627.
  10. Davis MR, Votaw JR, Bremner JD, et al. Initial human PET imaging studies with the dopamine transporter ligand 18F-FECNT. J Nucl Med 2003;44:855-861.
  11. Malison RT, Vessotskie JM, Kung M-P, et al. Striatal dopamine transporter imaging in nonhuman primates with iodine-123-IPT SPECT. J Nucl Med 1995;36:2290-2297.
  12. Shaya EK, Scheffel U, Dannals RF, et al. In vivo imaging of dopamine reuptake sites in the primate brain using single photon emission computed tomography (SPECT) and iodine-123 labeled RTI-55. Synapse 1992;10:169-172.
  13. Kuikka JT, Baulieu JL, Hiltunen J, et al. Pharmacokinetics and dosimetry of iodine-123 labelled PE2I in humans, a radioligand for dopamine transporter imaging. Eur J Nucl Med 1998;25:531-534.
  14. Abi-Dargham A, Gandelman MS, DeErausquin GA, et al. SPECT imaging of dopamine transporters in human brain with iodine-123-. uoroalkyl analogs of β-CIT. J Nucl Med 1996;37:1129-1133.
  15. Kung HF, Kim HJ, Kung MP, Meegalla SK, Plossl K, Lee HK. Imaging of dopamine transporters in humans with technetium-99m TRODAT-1. Eur J Nucl Med 1996;23:1527-1530.
  16. Chan GL, Holden JE, Stoessl AJ, et al. Reproducibility of the distribution of carbon-11-SCH 23390, a dopamine D1 receptor tracer, in normal subjects. J Nucl Med 1998;39:792-797.
  17. Karlsson P, Farde L, Halldin C, et al. PET examination of [11C]NNC 687 and [11C]NNC 756 as new radioligands for the D1-dopamine receptor. Psychopharmacology (Berl) 1993;113:149-156.
  18. Farde L, Ehrin E, Eriksson L, et al. Substituted benzamides as ligands for visualization of dopamine receptor binding in the human brain by positron emission tomography. Proc Natl Acad Sci USA 1985;82:3863-3867.
  19. Wagner HN Jr, Burns HD, Dannals RF, et al. Imaging dopamine receptors in the human brain by positron tomography. Science 1983;221:1264-1266.
  20. Nikolaus S, Larisch R, Beu M, et al. In vivo measurement of D2 receptor density and af.nity for 18F-(3-N-methyl)benperidol in the rat striatum with a PET system for small laboratory animals. J Nucl Med 2003;44:618-624.
  21. Mozley PD, Stubbs JB, Kim HJ, et al. Dosimetry of a D2/D3 dopamine receptor antagonist that can be used with PET or SPECT. J Nucl Med 1995;36:1322-1331.
  22. Grunder G, Siessmeier T, Piel M, et al. Quantification of D2-like dopamine receptors in the human brain with 18F-desmethoxyfallypride. J Nucl Med 2003;44:109-116.
  23. Kung HF, Pan S, Kung MP, et al. In vitro and in vivo evaluation of [123I]IBZM: a potential CNS D-2 dopamine receptor imaging agent. J Nucl Med 1989;30:88-92.
  24. Kung MP, Kung HF, Billings J, Yang Y, Murphy RA, Alavi A. The characterization of IBF as a new selective dopamine D-2 receptor imaging agent. J Nucl Med 1990;31:648-654.
  25. Kessler RM, Mason NS, Votaw JR, et al. Visualization of extrastriatal dopamine D2 receptors in the human brain. Eur J Pharmacol 1992;223:105-107.
  26. Suehiro M, Scheffel U, Ravert HT, Dannals RF, Wagner HN Jr. [11C](+)McN5652 as a radiotracer for imaging serotonin uptake sites with PET. Life Sci 1993;53:883-892.
  27. Farde L, Halldin C, Muller L, Suhari T, Karlsson P, Hall H. PET-study of [11C]β-CIT binding to monoamine transporters in the monkey and human brain. Synapse 1994; 16: 93–103.
  28. Bergstrom KA, Halldin C, Hall H, et al. In vitro and in vivo characterisation of nor-β-CIT: a potential radioligand for visualization of the serotonin transporter in the brain. Eur J Nucl Med 1997;24:596-601.
  29. Vercouillie J, Tarkiainen J, Halldin C, et al. Precursor synthesis and radiolabeling of [11C]ADAM: a potent radioligand for the serotonin transporter exploration by PET. J Labelled Comp Radiopharm 2001;44:113-120.
  30. Huang Y, Hwang DR, Zhu Z, et al. Synthesis and pharmacological characterization of a new PET ligand for the serotonin transporter: [ 11C]5-bromo-2-[2-(dimethylaminomethylphenylsulfanyl)] phenylamine ([11C]DAPA). Nucl Med Biol 2002;29:741-751.
  31. Huang Y, Bae S-A, Zhu Z, Guo N, Hwang DR, Laruelle M. Fluorinated analogues of ADAM as new PET radioligands for the serotonin transporter: synthesis and pharmacological evaluation. J Labelled Comp Radiopharm 2001;44:S18-S20
  32. Houle S, Ginovart N, Hussey D, Meyer JH, Wilson AA. Imaging the serotonin transporter with positron emission tomography: initial human studies with [11C]DAPP and [11C]DASB. Eur J Nucl Med 2000;27:1719-1722.
  33. Halldin C, Lundberg J, Sovago J, et al. [11C]MADAM, a new serotonin transporter radioligand characterized in the monkey brain by PET. Synapse 2005;58:173-183.
  34. Shiue GG, Fang P, Shiue CY. Synthesis of N,N-dimethyl-2-(2-amino-4-[18F]fluorophenylthio) benzylamine as a serotonin transporter imaging agent. Appl Radiat Isot 2003;58:183-191.
  35. Kuikka JT, Bergstrom KA, Vanninen E, Laulumaa V, Hartikainen P, Lansimies E. Initial experience with single-photon emission tomography using iodine-123-labelled 2βcarbomethoxy-3β-(4-iodophenyl) tropane in human brain. Eur J Nucl Med 1993;20:783-786.
  36. Hiltunen J, Akerman KK, Kuikka JT, et al. Iodine-123 labeled nor-beta-CIT as a potential tracer for serotonin transporter imaging in the human brain with single-photon emission tomography. Eur J Nucl Med 1998;25:19-23.
  37. Oya S, Choi SR, Hou C, et al. 2-((2-((Dimethyl amino)methyl)phenyl)thio) -5-iodophenylamine (ADAM): an improved serotonin transporter ligand. Nucl Med Biol 2000;27:249-254.
  38. Mathis CA, Simpson NR, Mahmood K, Kinahan PE, Mintun MA. [11C]WAY 100635: a radioligand for imaging 5-HT1A receptors with positron emission tomography. Life Sci 1994;55:PL403-407.
  39. Pike VW, Halldin C, McCarron JA, 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 1998;25:338-346.
  40. Shiue CY, Shiue GG, Mozley PD, et al. p-[18F]-MPPF: a potential radioligand for PET studies of 5-HT1A receptors in humans. Synapse 1997; 25:147-154.
  41. Defraiteur C, Lemaire C, Luxen A, Plenevaux A. Radiochemical synthesis and tissue distribution of p-[18F]DMPPF, a new 5-HT1A ligand for PET, in rats. Nucl Med Biol 2006;33:667-675.
  42. Wong DF, Wagner HN Jr, Dannals RF, et al. Effects of age on dopamine and serotonin receptors measured by positron tomography in the living human brain. Science 1984;226:1393-1396.
  43. Lundkvist C, Halldin C, Ginovart N, et al. [11C]MDL 100907, a radioligland for selective imaging of 5-HT2A receptors with positron emission tomography. Life Sci 1996;58:PL187-192.
  44. Blin J, Pappata S, Kiyosawa M, Crouzel C, Baron JC. [18F]setoperone: a new high-affinity ligand for positron emission tomography study of the serotonin-2 receptors in baboon brain in vivo. Eur J Pharmacol 1988;147:73-82.
  45. Lemaire C, Cantineau R, Guillaume M, Plenevaux A, Christiaens L. Fluorine-18-altanserin: a radioligand for the study of serotonin receptors with PET: radiolabeling and in vivo biologic behavior in rats. J Nucl Med 1991;32:2266-2272.
  46. Busatto GF, Pilowsky LS, Costa DC, et al. Initial evaluation of 123I-5-I-R91150, a selective 5-HT 2A ligand for single-photon emission tomography, in healthy human subjects. Eur J Nucl Med 1997; 24:119-124.
  47. Agdeppa ED, Kepe V, Liu J, et al. Binding characteristics of radio. uorinated 6-dialkylamino-2-naphthylethylidene derivatives as positron emission tomography imaging probes for β-amyloid plaques in Alzheimer's disease. J Neurosci 2001;21:RC189.
  48. Klunk WE, Engler H, Nordberg A, et al. Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B. Ann Neurol 2004; 55:306-319.
  49. Kung MP, Hou C, Zhuang ZP, Skovronsky D, Kung HF. Binding of two potential imaging agents targeting amyloid plaques in postmortem brain tissues of patients with Alzheimer's disease. Brain Res 2004;1025:98-105.
  50. Kung MP, Hou C, Zhuang ZP, et al. IMPY: an improved thioflavin-T derivative for in vivo labeling of beta-amyloid plaques. Brain Res 2002;956:202-210