Pediatric PET Imaging - part 8 potx

59 244 0
Pediatric PET Imaging - part 8 potx

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

ligands have high affinity for extrastriatal receptors and are displaced from the receptor sites by amphetamine-induced dopamine release (27,97,98). Even though dynamic receptor imaging has so far been used only for detection of striatal dopamine, it is theoretically possible to develop ligands that can detect release of other neurotransmitters in other brain areas. Such developments will go a long way in enhancing our under- standing of human cognitive control. Conclusion Neurotransmitter imaging has resulted in a considerable amount of new information concerning the pathogenesis of a number of neuro- logic and psychiatric conditions that include schizophrenia, addiction, Parkinson’s disease, Alzheimer’s disease, ADHD, epilepsy, anxiety, and affective disorders. Further, the use of these techniques in the diag- noses of subclinical Alzheimer’s and Parkinson’s disease in asympto- matic patients can help in early diagnosis and intervention. In addition, localization of epileptic foci by GABAreceptor imaging has been shown to improve postsurgical clinical outcome. Neurotransmitter imaging for drug evaluation has aided in the development of new com- pounds that target specific receptors that are dysregulated in various disorders. Evolving molecular imaging techniques, like dynamic recep- tor imaging, offer even more exciting possibilities. These techniques can identify and localize areas of the brain where specific neuro- transmitters are released during a task performance or symptom provocation. It will greatly expand our understanding of the funda- mental alterations in neurochemistry in psychiatric and neurologic disorders. In addition, these methods will provide empirical data that can be used to formulate novel therapeutic strategies for treatment and prevention of the disorders that are associated with altered neurotransmission. References 1. Jacobs AH, Li H, Winkeler A, et al. PET-based molecular imaging in neu- roscience. Eur J Nucl Med Mol Imaging 2003;30:1051–1065. 2. Blankenberg FG. Molecular imaging: the latest generation of contrast agents and tissue characterization techniques. J Cell Biochem 2003;90: 443–453. 3. Gjedde A, Wong DF, Rosa-Neto P, Cumming P. Mapping neuroreceptors at work: on the definition and interpretation of binding potentials after 20 years of progress. Int Rev Neurobiol 2005;63:1–20. 4. Laruelle M. Imaging synaptic neurotransmission with in vivo binding competition techniques: a critical review. J Cereb Blood Flow Metab 2000; 20:423–451. 5. Alpert NM, Badgaiyan RD, Livini E, Fischman AJ. A novel method for non- invasive detection of neuromodulatory changes in specific neurotransmit- ter systems. NeuroImage 2003;19:1049–1060. A.J. Fischman and R.D. Badgaiyan 397 6. Badgaiyan RD, Alpert NM, Fischman AJ. Detection of striatal dopamine release during an implicit motor memory task. Annual meeting of the Society of Nuclear Medicine, Philadelphia, 2004. 7. Badgaiyan RD, Alpert NM, Fischman AJ. De tection of striatal dopamine released during an explicit motor memory task. Annual meeting of the Society of Nuclear Medicine, Toronto, 2005. 8. Badgaiyan RD, Alpert NM, Fischman AJ. Detection of striatal dopamine release during a motor planning task in human volunteers. Brain’03, XXIst International Symposium on Cerebral Blood Flow, Metabolism and Func- tion (abstract 706). Calgarg, 2003. 9. Badgaiyan RD, Fischman AJ , Alpert NM. Striatal dopamine release during unrewarded motor task in human volunteers. NeuroReport 2003;14:1421– 1424. 10. Badgaiyan RD, Fischman AJ, Alpert NM. Striatal dopamine release during unrewarded motor t ask in human volunteers. Neuroreport 2003;14:1421– 1424. 11.Koepp MJ, Gunn RN, Lawrence AD, et al. Evidence for striatal dopamine release during a video game. Nature 1998;393:266–268. 12.Pappata S, Dehaene S, Poline JB, et al. In vivo detection of striatal dopamine release during reward: a PET study with ((11)C)raclopride and a single dynamic scan approach. NeuroImage 2002;16:1015 –1027. 13. Morris ED, Alpert NM, Fischman AJ. Comparison of two compartmental models for describing receptor ligand kinetics and receptor availability in multiple injection PET studies. J Cereb Blood Flow Metab 1996;16:841–853. 14.Choy G, Choyke P, Libutti SK. Current advances in molecular imaging: noninvasive in vivo bioluminescent and fluorescent optical imaging in cancer research. Mol Imaging 2003;2:303–312. 15.Maria Moresco R, Messa C, Lucignani G, et al. PET in psychopharmacol- ogy. Pharmacol Res 2001;44:151–159. 16. Wagner HNJ, Dannals RF, Frost JJ, et al. Imaging neuroreceptors in the human brain in health and disease. Radioisotopes 1985;34:103–107. 17. Wong DF, Gjedde A, Wagner HNJ. Quantification of neuroreceptors in the living human brain. I. Irreversible binding of ligands. J Cereb Blood Flow Metab 1986;6:137–146. 18. Wong DF, Young D, Wilson PD, Meltzer CC, Gjedde A. Quantification of neuroreceptors in the living human brain: III. D2–like dopamine receptors: theory, validation, and changes during normal aging. J Cereb Blood Flow Metab 1997;17:3 16–330. 19. Mintun MA, Raichle ME, Kilbourn MR, Wooten GF, Welch MJ. A quanti- tative model for the in vivo assessment of drug binding sites with positron emission tomography. Ann Neurol 1984;15:217–227. 20. Laruelle M, al-Tikriti MS, Zea-Ponce Y, et al. In vivo quantification of dopamine D2 receptor parameters in nonhuman primates with (123I)iodobenzofuran and single photon emission computerized tomo- graphy. Eur J Pharmacol 1994;263:39–51. 21. Saelens JK, Simke JP, Neale SE, Weeks BJ, Selwyn M. Effects of haloperidol and d-amphetamine on in vivo 3H-spiroperiodol binding in the rat fore- brain (1). Arch Int Pharmacodyn Ther 1980;246:98–107. 22. Chugani DC, Ackermann RF, Phelps ME. In vivo (3H)spiperone binding: evidence for accumulation in corpus striatum by agonist-mediated recep- tor internalization. J Cereb Blood Flow Metab 1988;8:291–303. 23. Laruelle M, Huang Y. Vulnerability of positron emission tomography radiotracers to endogenous competition. New insights. Q J Nucl Med 2001;45:124–138. 398 Chapter 21 Neurotransmitter Imaging 24. Christian BT, Narayanan TK, Shi B, Mukherjee J. Quantitation of striatal and extrastriatal D-2 dopamine receptors using PET imaging of ((18)F) fallypride in nonhuman primates. Synapse 2000;38:71–79. 25. de Paulis T. The discovery of epidepride and its analogs as high-affinity radioligands for imaging extrastriatal dopamine D(2) receptors in human brain. Curr Pharm Des 2003;9:673–696. 26.Mukherjee J, Christian BT, Dunigan KA, et al. Brain imaging of 18F- fallypride in normal volunteers: Blood analysis, distribution, test-retest studies, and preliminary assessment of sensitivity to aging effects on dopamine D -2/D-3 receptors. Synapse 2002;46:170–188. 27. Mukherjee J, Christian BT, Narayanan TK, Shi B, Collins D. Measurement of d-amphetamine-induced effects on the binding of dopamine D-2/D-3 receptor radioligand, 18F-fallypride in extrastriatal brain regions in non- human primates using PET. Brain Res 2005;1032:77–84. 28. Abi-Dargham A. Do we still believe in the dopamine hypothesis? New data bring new evidence. Int J Neuropsychopharmacol 2004;7(suppl 1):S1–5. 29. Wong DF, Wagner HNJ, Tune LE, et al. Positron emission tomography reveals elevated D2 dopamine receptors in drug-naive schizophrenics. Science 1986;234:1558–1563. 30.Marzella PL, Hill C, Keks N, Singh B, Copolov D. The binding of both (3H)nemonapride and (3H)raclopride is increased in schizophrenia. Biol Psychiatry 1997;42:648–654. 31.Farde L , Wiesel FA, Stone-Elander S, et al. D2 dopamine receptors in neu- roleptic-naive schizophrenic patients. A positron emission tomography study with (11C)raclopride. Arch Gen Psychiatry 1990;47:213–219. 32.Guo N, Hwang DR, Lo ES, Huang YY, Laruelle M, Abi-Dargham A. Dopamine depletion and in vivo binding of PET D1 receptor radioligands: implications for imaging studies in schizophrenia. Neuropsychopharma- cology 2003;28:1703–1711. 33. Seeman P. Brain dopamine receptors in schizophrenia: PET problems. Arch Gen Psychiatry 1988;45(6):598–600. 34. Seeman P, Guan HC, Niznik HB. Endogenous dopamine lowers the dopamine D2 receptor density as measured by (3H)raclopride: implications for positron emission tomography of the human brain. Synapse 1989;3:96–97. 35. Xu ZC, Ling G, Sahr RN, Neal-Beliveau BS. Asymmetrical changes of dopamine receptors in the striatum after unilateral dopamine depletion. Brain Res 2005;1038:163–170. 36. Laruelle M. The role of endogenous sensitization in the pathophysiolo gy of schizophrenia: implications from recent brain imaging studies. Brain Res Brain Res Rev 2000;31:371–384. 37. Breier A, Su TP, Saunders R, et al. Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron emission tomography method. Proc Natl Acad Sci U S A 1997;94:2569–2574. 38. Laruelle M, Abi-Dargham A, van Dyck CH, et al. Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc Natl Acad Sci U S A 1996;93:9235–9240. 39. Abi-Dargham A, Gil R, Krystal J, et al. Increased striatal dopamine trans- mission in schizophrenia: confirmation in a second cohort. Am J Psychia- try 1998;155:761–767. 40. Laruelle M, Abi-Dargham A, Gil R, Kegeles L, Innis R. Increased dopamine transmission in schizophrenia: re lationship to illness phases. Biol Psychia- try 1999;46:56–72. A.J. Fischman and R.D. Badgaiyan 399 41. Voruganti L, Slomka P, Z abel P, et al. Subjective effects of AMPT-induced dopamine depletion in schizophrenia: correlation between dysphoric responses and striatal D(2) binding ratios on SPE CT imaging. Neuropsy- chopharmacology 2001;25:642–650. 42. Fischman AJ. Role of (18F)-dopa-PET imaging in assessing movement dis- orders. Radiol Clin North Am 2005;43:93–106. 43. McGowan S, Lawrence AD, Sales T, Quested D, Grasby P. Presynaptic dopaminergic dysfunction in schizophrenia: a positron emission tomo- graphic (18F)fluorodopa study. Arch Gen Psychiatry 2004;61:134–142. 44. Bannon MJ. The dopamine transporter: role in ne urotoxicity and human disease. Toxicol Appl Pharmacol 2005;204:355–360. 45. Fischman AJ, Bonab AA, Babich JW, et al. Rapid detection of Parkinson’s disease by SPECT with altropane: a selective ligand for dopamine trans- porters. Synapse 1998;29:128–141. 46. Lee CS, Samii A, Sossi V, et al. In vivo positron emission tomographic evi- dence for compensatory changes in presynaptic dopaminergic nerve ter- minals in Parkinson’s disease. Ann Neurol 2000;47:493–503. 47. Marshall V, Grosset D. Role of dopamine transporter imaging in routine clinical practice. Mov Disord 2003;18:1415–1423. 48. DiMaio S, Grizenko N, Joober R. Dopamine genes and attention-deficit hyperactivity disorder: a review. J Psychiatry Neurosci 2003;28:27–38. 49.Madras BK, Miller GM, Fischman AJ. The dopamine transp orter: relevance to attention deficit hyperactivity disorder (ADHD). Behav Brain Res 2002;130:57–63. 50. Dougherty DD, Bonab AA, Spencer TJ, Rauch SL, Madras BK, Fischman AJ. Dopamine transporter density in patients with attention deficit hyper- activity disorder. Lancet 1999;354(9196):2132–2133. 51.Cheon KA, Ryu YH, Kim YK, Namkoong K, Kim CH, Lee JD. Dopamine transporter density in the basal ganglia assessed with (123I)IPT SPET in children with attention deficit hyperactivity disorder. Eur J Nucl Med Mol Imaging 2003;30:306–311. 52. Dresel S, Krause J, Krause KH, et al. Attention deficit hyperactivity disor- der: binding of (99mTc)TRODAT-1 to the dopamine transporter before and after methylphenidate treatment. Eur J Nucl Med 2000;27:1518–1524. 53. Vles JS, Feron FJ, Hendriksen JG, Jolles J, van Kroonenburgh MJ, Weber WE. Methylphenidate down-regulates the dopamine receptor and trans- porter system in children with attention deficit hyperkinetic disorder (ADHD ). Neuropediatrics 2003;34:77–80. 54. Volkow ND, Wang GJ, Fowler JS, et al. Cardiovascular effects of methylphenidate in humans are associated with increases of dopamine in brain and of epinephrine in plasma. Psychopharmacology (Berl) 2003; 166:264–270. 55. Volkow ND, Fowler JS, Wang GJ, Goldstein RZ. Role of dopamine, the frontal cortex and memory circuits in drug addiction: insight from imaging studies. Neurobiol Learn Mem 2002;78:610–624. 56. Daws LC, Callaghan PD, Moron JA, et al. Cocaine increases dopamine uptake and cell surface expression of dopamine transporters. Biochem Biophys Res Commun 2002;290:1545–1550. 57. Kahlig KM, Binda F, Khoshbouei H, et al. Amphetamine induces dopamine efflux through a dopamine transporter channel. Proc Natl Acad Sci U S A 2005;102:3495–3500. 58. Volkow ND, Chang L , Wang GJ, et al. Loss of dopamine transporters in methamphetamine abusers recovers with protracted abstinence. J Neurosci 2001;21:9414–9418. 400 Chapter 21 Neurotransmitter Imaging 59. McCann UD, Wong DF, Yokoi F, Villemagne V, Dannals RF, Ricaurte GA. Reduced striatal dopamine transporter density in abstinent methamphet- amine and methcathinone users: evidence from positron emissio n tomo- graphy studies with (11C)WIN-35,428. J Neurosci 1998;18:8417–8422. 60. Wilson JM, Kalasinsky KS, Levey AI, et al. Striatal dopamine nerve termi- nal markers in human, chronic methamphetamine users. N at Med 1996;2: 699–703. 61.Parsey RV , Mann JJ. Applications of positron emission tomography in psy- chiatry. Semin Nucl Med 2003;33:129–135. 62.Merlet I, Ostrowsky K, Costes N, et al. 5–HT1Areceptor binding and intrac- erebral activity in temporal lobe epilepsy: an (18F)MPPF-PET study. Brain 2004;127:900–913. 63. Toczek MT, Carson RE, Lang L, et al. PET imaging of 5–HT1Areceptor binding in patients with temporal lobe epilepsy. Neurology 2003;60: 749–756. 64. Juhasz C, Chugani DC, Muzik O, et al. Alpha-methyl-L-tryptophan PET detects epileptogenic cortex in children with intractable epilepsy. Neurol- ogy 2003;60:960–968. 65.Farde L, Ginovart N, Ito H, et al. PET-characterization of (carbonyl- 11C)WAY-100635 binding to 5–HT1Areceptors in the primate brain. Psy- chopharmacology (Berl) 1997;133:196–202. 66.P arsey RV , Mann JJ. Applications of positron emission tomography in psy- chiatry. Semin Nucl Med 2003;33:129–135. 67. Sihver W, Langstrom B, Nordberg A. Ligands for in vivo imaging of nico- tinic receptor subtypes in Alzheimer brain. Acta Neurol Scand Suppl 2000;176:27–33. 68. Nordberg A, Hartvig P, Lilja A, et al. Decreased uptake and binding of 11C- nicotine in brain of Alzheimer patients as visualized by positron emission t omography. J Neural Transm Park Dis Dement Sect 1990;2:215–224. 69.Nordberg A, Lundqvist H, Hartvig P, Lilja A, Langstrom B. Kinetic analy- sis of regional (S)(-)11C-nicotine binding in normal and Alzheimer brains— in vivo assessment using positron emission tomography. Alzheimer Dis Assoc Disord 1995;9:21–27. 70.Nordberg A, Amberla K, Shigeta M, et al. Long-term tacrine treatment in three mild Alzheimer patients: effects on nicotinic receptors, cerebral blood flow, glucose metabolism, EEG, and cognitive abilities. Alzheimer Dis Assoc Disord 1998;12:228–237. 71. Yoshida T, Kuwabara Y, Ichiya Y, et al. Cerebral muscarinic acetylcholiner- gic receptor measurement in Alzheimer’s disease patients on 11C-N- methyl-4—piperidyl benzilate—comparison with cerebral blood flow and cerebral glucose metabolism. Ann Nucl Med 1998;12:35–42. 72.Maziere M, Hantraye P, Prenant C, Sastre J, Comar D. Synthesis of ethyl 8–fluoro-5,6–dihydro-5–(11C)methyl-6–oxo-4H-imidazo (1,5–a) (1,4)benzodiazepine-3–carboxylate (RO 15.1788–11C): a specific radioli- gand for the in vivo study of central benzodiazepine receptors by positron emission tomography. Int J Appl Radiat Isot 1984;35:973–976. 73. Hammers A. Flumazenil positron emission tomography and other ligands for functional imaging. Neuroimaging Clin North Am 2004;14:537–551. 74.Matheja P, Ludemann P, Kuwert T, et al. Disturbed benzodiazepine recep- tor function at the onset of temporal lobe epilepsy—lomanzenil-binding in de-novo TLE. J Neurol 2001;248:585–591. 75. Mitterhauser M, Wadsak W, Wabnegger L, et al. Biological evaluation o f 2¢-(18F)fluoroflumazenil ((18F)FFMZ), a potential GABAreceptor ligand for PET. Nucl Med Biol 2004;31:291–295. A.J. Fischman and R.D. Badgaiyan401 76.Heiss WD, Graf R, Fujita T, Ohta K, Bauer B, Lottgen J, Wienhard K. Early detection of irreversibly damaged ischemic tissue by flumazenil positron emission tomography in cats. Stroke 1997;28:2045–2051; discussion 2051 – 2052. 77. Savic I, Pauli S, Thorell JO, Blomqvist G. In vivo demonstration of altered benzodiazepine receptor density in patients with generalised epilepsy. J Neurol Neurosurg Psychiatry 1994;57:797–804. 78. Koepp MJ, Richardson MP, Brooks DJ, et al. Cerebral benzodiazepine receptors in hippocampal sclerosis. An objective in vivo analysis. Brain 1996;119:1677–1687. 79. Debets RM, Sadzot B, van Isselt JW, et al. Is 11C-flumazenil PET superior to 18FDG PET and 123I-iomazenil SPECT in presurgical evaluation of tem- poral lobe epilepsy? J Neurol Neurosurg Psychiatry 1997;62:141–150. 80. Juhasz C, Chugani DC, Muzik O, et al. Relationship of flumazenil and glucose PET abnormalities to neocortical epilepsy surgery outcome. Neurology 2001;56:1650–1658. 81. Bouvard S, Costes N, Bonnefoi F, et al. Seizure-related short-term plastic- ity of benzodiazepine receptors in partial epilepsy: a (11C)flumazenil-PET study. Brain 2005;128:1330–1343. 82. Savic I, Svanborg E, Thorell JO. Cortical benzodiazepine receptor changes are related to frequency of partial seizures: a positron emission tomogra- phy study. Epilepsia 1996;37:236–244. 83. Machulla HJ, Heinz A. Radioligands for brain imaging of the kappa-opioid system. J Nucl Med 2005;46:386–387. 84. Bencherif B, Wand GS, McCaul ME, et al. Mu-opioid receptor binding measured by (11C)carfentanil positron emission tomography is related to craving and mood in alcohol dependence. Biol Psychiatry 2004;55:255– 262. 85.Cohen RM, Andreason PJ, Doudet DJ, Carson RE, Sunderland T. Opiate rece ptor avidity and cerebral blood flow in Alzheimer’s disease. J Neurol Sci 1997;148:171–180. 86.Prevett MC, Cunningham VJ, Brooks DJ, Fish DR, Duncan JS. Opiate recep- tors in idiopathic generalised epilepsy measured with (11C)diprenorphine and positron emission tomography. Epilepsy Res 1994;19:71–77. 87. Bartenstein PA, Prevett MC, Duncan JS, Hajek M, Wieser HG. Quantifica- tion of opiate receptors in t wo patients with mesiobasal temporal lobe epilepsy, before and after selective amygdalohippocampectomy, using positron emission tomography. Epilepsy Res 1994;18:119–125. 88. McConathy J, Owens MJ, Kilts CD, et al. Synthesis and biological evalua- tion of (11C)talopram and (11C)talsupram: candidate PET ligands for the norepinephrine transporter. Nucl Med Biol 2004;31:705–718. 89. Ressler KJ, Nemeroff CB. Role of norepinephrine in the pathophysiology of neuropsychiatric disorders. CNS Spectr 2001;6:663–666, 670. 90. Klimek V, Stockmeier C, Overholser J, et al. Reduced levels of norepi- nephrine transporters in the locus coeruleus in major depression. J Neu- rosci 1997;17:8451–8458. 91.Morris ED, Fisher RE, Alpert NM, Rauch SL, Fischman AJ. In vivo imaging of neuromodulation using positron emission tomography: optimal ligand characteristics and task length for detection of activation. Hum Brain Map 1995;3:35–55. 92. Fisher RE, Morris ED, Alpert NM, Fischman AJ. In vivo imaging of neurom odulatory synaptic transmission using PET: A review of relevant neurophysiology. Hum Brain Map 1995;3:24–34. 402 Chapter 21 Neurotransmitter Imaging 93. Dewey SL, MacGregor RR, Brodie JD, et al. Mapping muscarinic receptors in human and baboon brain using (N-11C- methyl)-benztropine. Synapse 1990;5:213–223. 94. Seeman P, Guan HC, Niznik HB. Endogenous dopamine lowe rs the dopamine D2 receptor density as measured by (3H)raclopride: implica- tions for positron emission tomography of the human brain. Synapse 1989;3:96–97. 95. Friston KJ, Malizia AL, Wilson S, Cunningham VJ, Jones T, Nutt DJ. Analy- sis of dynamic radioligand displacement or “activation” studies. J Cereb Blood Flow Metab 1997;17:80–93. 96. Gunn RN, Lammertsma AA, Hume SP, Cunningham VJ. Parametric imaging of ligand-r eceptor binding in PET using a simplified reference region model. Neuroimage 1997;6:279–287. 97. Mukherjee J, Yang ZY, Lew R, et al. Evaluation of d-amphetamine effects on the binding of dopa mine D-2 receptor radioligand, 18F-fallypride in nonhuman primates using positron emission tomography. Synapse 1997; 27:1–13. 98. Chou YH, Halldin C, Farde L. Effect of amphetamine on e xtrastriatal D2 dopamine receptor binding in the primate brain: a PET study. Synapse 2000;38:138–143. A.J. Fischman and R.D. Badgaiyan 403 Section 4 Other Applications 22 Cardiovascular Applications Miguel Hernandez-Pampaloni An improved understanding of the pathophysiology of myocardial ischemia combined with the development of new diagnostic modali- ties has substantially modified the concepts of myoca rdial blood flow (MBF) and left ventricular function in coronary artery disease (CAD). Positron emission tomography (PET) has emerged as a unique tool to characterize physiologic and pathologic processes, and it already plays a significant role in different areas of clinical medicine, including car- diology. Cardiac PET is based on the properties of positron emitters and radiation detection to provide a noninvasive and in vivo assess- ment of regional myocardial perfusion and metabolism. Different study protocols have been largely used in the adult population to detect and grade the severity of coronary artery disease during the last two decades using cardiac PET technology. Further, cardiac PET using fluorine-18 ( 18 F) 2-fluoro-2-deoxyglucose (FDG) is considered the gold standard imaging modality for the assessment of myocardial viability (1) and is well recognized as providing accurate information on the long-term prognosis of the patients with chronic coronary ischemic disease (2). Positron emission tomography offers unique capabilities for nonin- vasive assessment of regional myocardial function and can disclose information of utmost importance for the more accurate understand- ing of pathophysiologic processes and for the optimal management of the diseased patient. By detecting the very early functional regional abnormalities before the development of more severe structural changes, PET imaging can be useful in providing improved and pre- ventive care to the patient. Thus, cardiac PET imaging can offer a more comprehensive understanding of the normal myocardial physiology and early recognition of functional abnormalities. This is even more important in pediatric cardiology where the early detection of myocar- dial dysfunction may be helpful in choosing the appropriate manage- ment for the prevention of long-term consequences. Recent surgical and technical advances in pediatric cardiology make even more impor- tant an accurate detection of potentially treatable coronary abnor- malities. Positron emission tomography imaging’s high spatial and 407 temporal resolution provides better image quality, especially important in small pediatric hearts, and it allows the quantification in absolute terms of the MBF and regional myocardial metabolism. Single photon emission computed tomography (SPECT) imaging quality is usually limited by poor resolution and the low usable activity of thallium 201 ( 201 Tl), whereas images obtained after the administration of technetium- labeled compounds are compromised by the high liver activity in close proximity to the small heart. This chapter describes the basic principles of PET applied to the study of the heart, and presents the current and potential future clini- cal applications of cardiac PET in pediatric patients. Principles of Emission Tomography Applied to the Cardiovascular System Positron emission tomography imaging features are based on the physical properties of the positron decay. A positron has the same characteristics as an electron, except for its positive charge. Positrons are emitted from unstable nuclei (that have an excess of protons) that dispose of their excess charge by emitting a positron. At the end of the positron range, after losing its kinetic energy, a positron combines with an electron and the two particles annihilate. The annihilation coinci- dence detection of the two colinear 511-keV gamma rays photons, emitted in diametrically opposite directions by opposing scintillator detectors, is the essence of the PET imaging formation (3). To detect the location of the annihilation event, detectors are placed on opposite sides of the source and are connected in a coincidence detection circuit. When a given event is recorded simultaneously, positron annihilation is assumed to have taken place on the line between the detectors, and hence the location can be accurately determined. Radiopharmaceuticals Cardiac PET studies are performed with radiopharmaceuticals speci- fically synthesized to assess determined cardiac functions or biochemi- cal processes and with radiopharmaceuticals that have applications in other disciplines. The radiolabeled positron-emitting tracers used in cardiac PET studies are produced by a cyclotron or by a generator system. Currently, different processes of the heart have been studied using different radiopharmaceuticals (Table 22.1). The radiation expo- sure to PET radiotracers is lower compared to other radionuclides used for nuclear cardiology studies (Table 22.2). Evaluation of Myocardial Blood Flow The development of suitable radiotracers and appropriate mathemati- cal models applied to PET imaging has been shown to allow for the noninvasive and accurate quantification of regional MBF. Different radiotracers have been used for measuring MBF, including nitrogen- 13–labeled ammonia ( 13 NH 3 ) (4–6), oxygen-15 ( 15 O)-labeled water ( 15 O- 408 Chapter 22 Cardiovascular Applications [...]... 11.00 0.0320 0.0 58 6.90 0.0130 0.041 4.90 0.0067 0.027 4.10 0.0043 0.019 0. 68 0.00 28 0.015 0.34 0.0022 Kidney Kidney Bladder wall Newborn (3.5 kg) 1-yr-old (10 kg) 5-yr-old ( 18 kg) 10-yr-old (31 kg) 15-yr-old (56 kg) Adult (70 kg) Dose equivalent per body weight 0.34 0 .89 12.22 0.43 7.67 0.14 0.30 5.44 0.07 0.20 4.56 0.05 0.14 0.76 0.03 0.11 0. 38 0.02 18 F-FDG 0.310 0.100 0.077 0.050 0.0 38 0.030 Bladder... inflammatory processes (35) Different technetium-99m (99mTc)-labeled compounds are being studied for potential clinical use in patients with FUO, such as 99mTc-hexamethylpropylene-amine-oxime (HMPAO)-labeled leukocytes (36), 99mTc-ciprofloxacin (37), and 99mTc-labeled monoclonal antibodies ( 38, 39) FDG -PET Scan Mechanisms of FDG Uptake by the Cells Currently, 1 8- uoro-2-deoxyglucose (FDG) is the most clinically... incorporation of the radiotracer into M Hernandez-Pampaloni Glucose (18F-FDG) Lactate Hexobinase Lactate Glucose-6-phosphate Pyruvate Glycogen Mitochondria PDH Triglycerides Phospholipids Acyl-Camitine Camitine TCA Acyl-CoA α-GP CPTI β-oxidation Acety1-CoA Acyl-CoA CO2 Myocyte Free Fatty Acids (11C-Palmitate) CO2 11C-Acetate CO2 Figure 22.1 Radiotracers that can be used by PET to investigate different myocardial... program for the evaluation of cardiac PET studies: initial results in the detection and localization of coronary artery disease using nitrogen-13–ammonia J Nucl Med 1993;34(6): 9 68 9 78 33 Tillisch J, et al Reversibility of cardiac wall-motion abnormalities predicted by positron tomography N Engl J Med 1 986 ;314(14) :88 4 88 8 34 Knuuti MJ, et al Myocardial viability: fluorine- 1 8- deoxyglucose positron emission... Cyclotron Generator Cyclotron 110 min 20 min 0.2 mm 0. 28 mm Cyclotron Cyclotron H2O) (7–9), the potassium analogue rubidium -8 2 (82 Rb) (10), copper-62 (62Cu)-pyruvaldehyde bis(N4-methylthio-semicarbazone (62Cu-PTSM) (11,12), gallium- 68 (68Ga)-labeled albumin microspheres (13), and potassium 38 (38K) (14) The choice of a specific radiotracer is finally frequently determined by different factors besides their physical... As a potassium analogue, 82 Rb is retained in the myocardium and equilibrates with the cellular potassium pool Because of the dependence on the flow rate and the metabolic state, Table 22.2 Dosimetry in cardiac pediatric nuclear medicine 99m Newborn (3.5 kg) 1-yr-old (10 kg) 5-yr-old ( 18 kg) 10-yr-old (31 kg) 15-yr-old (56 kg) Adult (70 kg) Critical organ 201 13 Tc-sestamibi Tl N-ammonia Effective dose... Currently, 13NH3, 15O-H2O, and 82 Rb are the most widely used PET perfusion tracers Generator-produced 82 Rb has the advantages of not requiring a cyclotron and having a very short half-life ( 78 seconds), making it attractive for one-session rest/stress imaging 82 Rb has a low first-pass myocardial extraction fraction (50% to 60%) that results in a nonlinear uptake in relation to blood flow, particularly at... 15O-labeled water and PET J Nucl Med 1999;40(11): 184 8– 185 6 10 Herrero P, et al Noninvasive quantification of regional myocardial perfusion with rubidium -8 2 and positron emission tomography Exploration of a mathematical model Circulation 1990 ;82 (4):1377–1 386 11 Tadamura E, et al Generator-produced copper-62-PTSM as a myocardial PET perfusion tracer compared with nitrogen-13–ammonia J Nucl Med 1996;37(5):729–735... (87 ) Therefore, equivocal radiographic findings can be accurately characterized with FDG -PET There is also increasing concern about the risk of radiation (88 ), radiocontrast-induced nephropathy (89 ), and allergic reactions (90) to patients imaged with CT Furthermore, FDG -PET is able to detect early inflammatory and infectious lesions when anatomic imaging modalities reveal no abnormalities (91) FDG -PET. .. Coll Cardiol 1 989 ;14(3):639–652 8 Araujo LI, et al Noninvasive quantification of regional myocardial blood flow in coronary artery disease with oxygen-15-labeled carbon dioxide inhalation and positron emission tomography Circulation 1991 ;83 (3) :87 5– 88 5 9 Kaufmann PA, et al Assessment of the reproducibility of baseline and hyperemic myocardial blood flow measurements with 15O-labeled water and PET J Nucl Med . 11.00 0.0320 0.310 1-yr-old (10kg) 0.0 58 6.90 0.0130 0.100 5-yr-old (18kg) 0.041 4.90 0.0067 0.077 10-yr-old (31kg) 0.0274.10 0.0043 0.050 15-yr-old (56kg) 0.019 0. 68 0.00 28 0.0 38 Adult (70 kg) 0.015. ( 82 Rb) (10), copper-62 ( 62 Cu)-pyruvaldehyde bis(N4-methylthio-semicarbazone ( 62 Cu-PTSM) (11,12), gallium- 68 ( 68 Ga)-labeled albumin microspheres (13), and potassium 38 ( 38 K) (14). The choice. kg) 0 .89 12 .22 0.34 1.64 1-yr-old (10kg) 0.43 7.67 0.14 0.69 5-yr-old (18kg) 0.30 5.44 0.07 0.41 10-yr-old (31kg)0.204.56 0.05 0.26 15-yr-old (56kg)0.14 0.76 0.03 0.20 Adult (70 kg)0.11 0. 38 0.02

Ngày đăng: 11/08/2014, 06:21

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan