Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 7.
Published in final edited form as: Eur J Nucl Med Mol Imaging. 2011 Dec 8;39(3):544–547. doi: 10.1007/s00259-011-2005-8

Suggested pathway to assess radiation safety of 11C-labeled PET tracers for first-in-human studies

Paolo Zanotti-Fregonara 1, Robert B Innis 1,
PMCID: PMC3791506  NIHMSID: NIHMS517099  PMID: 22160195

Dear Sir,

Before a new radioactive tracer can be used in humans, many national as well as local Institutional Radiation Safety Committees often require that a biodistribution study be conducted in animals. The aim of these time-consuming and expensive studies is to assess the dose of absorbed radiation in animals so that the human dose can be extrapolated. Because of their physical similarity to humans (organ size and body weight), monkeys are arguably the best animal species to estimate radiation burden in human subjects. The extrapolation can be performed with different methods [1], for example by converting the percent administered activity in the monkey organ to percent administered activity in the same human organ. Basically, the human residence time for a given organ is derived by performing the animal to human organ mass ratio multiplication for each time point and then by integrating that time-activity curve to derive a hypothetical area under the curve for that human organ. This process is then repeated for each major source organ. The resulting residence times are then used in conjunction with an anthropomorphic human phantom to calculate the human dose. This method assumes that the metabolism of radiopharmaceuticals is similar between the two species and varies only as a function of organ mass. However, as we will describe below, it is our contention that results obtained in this manner from monkeys are misleading. This commentary will suggest a more accurate alternative that nevertheless maintains a wide safety margin.

To investigate this issue, we identified a total of nine 11C-labeled tracers for which both monkey and human absorbed doses were reported. We compared the absorbed doses extrapolated from monkeys for that particular radioligand and the mean dose from human biodistribution data for all nine radiotracers.

The results of these studies showed that, in general, doses extrapolated from monkeys overestimated the human effective dose (Table 1). Looking at all nine tracers, we found that the mean dose extrapolated from monkeys was 7.3±1.6 µSv/MBq, while the mean dose measured in actual human studies for the same tracers was 5.7±1.2 µSv/MBq (paired t test: p<0.001). Furthermore, such overestimates were not constant, but showed unpredictable variations; indeed, the percentage difference ranged from −72 to +11%. The most likely reason for such discrepancies is metabolic differences between different species. For instance, for 11C-(R)-rolipram, biodistribution in monkey poorly estimated biodistribution in humans [2]. Compared to humans, monkeys had a higher liver uptake, no hepatobiliary excretion, and higher urinary elimination. This led to an approximately 40% overestimate of the effective dose [2]. For 11C-PBR28, monkey data overestimated the effective human dose by 60%, with exposures to individual organs both over- and underestimated [3].

Table 1.

Comparison between the effective dose extrapolated from monkeys for nine different tracers and the actual effective dose measured in humans

Tracer Dose from monkeys
(µSv/MBq)
Reference Dose from humans
(µSv/MBq)
Reference % difference as compared
to monkey estimatesa
% difference as compared
to the human meanb
11C-BTA-1 7.4 Parsey et al. (2005) [7] 4.3 Thees et al. (2007) [8] −72.1 −25.1
11C-(R)-Rolipram 6.6 Sprague et al. (2008) [2] 4.8 Sprague et al. (2008) [2] −37.5 −16.4
11C-DASB 6.2 Tipre et al. (2004) [9] 7.0 Lu et al. (2004) [10] 11.4 21.9
11C-PBR28 10.3 Brown et al. (2007) [3] 6.6 Brown et al. (2007) [3] −56.1 14.9
11C-CUMI-101 6.9 Parsey (unpubl.) 5.3 Hines et al. (2011) [11] −30.2 −7.7
11C-MePPEP 6.6 NIMH/SNIDD [12] 4.6 Terry et al. (2010) [13] −43.5 −19.9
11C-dLop 9.4 Liow et al. (2009) [14] 7.8 Seneca et al. (2009) [15] −20.5 35.8
11C-(R)-PK11195 5.3 Our unpubl. data 5.0 Hirvonen et al. (2010) [16] −6.0 −13.0
4.6 Kumar et al. (2010) [17] −13 −19.9
11C-raclopride 6.7 Herscovitch et al. (1997) [18] 6.3 Slifstein et al. (2006) [19] −6.3 9.7
6.7 Ribeiro et al. (2005) [20] 0 16.6
Mean±SD 7.3±1.6 5.7±1.2 −29.0±26.5 0±21.7
a

Obtained for each tracer j by: (human dosej − monkey dosej)*100/human dosej

b

Obtained for each tracer j by: (human dosej − mean human dose)*100/mean human dose

Moreover, the doses measured in human studies were quite constant across the different tracers (range 4.3–7.8 µSv/MBq) (Table 1). This suggests that when predicting the human dose for a given tracer, a simple mean human value from all published studies is a better predictor of the effective dose than the value derived from monkeys for that specific tracer.

We also searched the literature and found 21 other 11C-labeled tracers for which dosimetry in human scans had been calculated. With the exception of 11C-WAY-100635, whose effective dose was about 12–16 µSv/MBq, all other tracers had very similar, low estimates (range 3.0–6.8 µSv/MBq, mean 5.1 µSv/MBq) that closely agree with the average dose of 5.8 µSv/MBq found in the first nine tracers (Table 2).

Table 2.

Effective dose measured in humans for 21 different 11C-labeled tracers

Radiopharmaceutical Target Effective dose
(µSv/MBq)
Reference
11C-Glucose Cell metabolism 3.0a Graham et al. (1998) [21]
3.8b
11C-NPA Dopamine D2/3 receptor 3.2 Laymon et al. (2009) [22]
11C-ABP688 Metabotropic glutamate receptor subtype 5 (mGluR5) 3.7 Treyer et al. (2008) [23]
Methyl-11C-thymidine Cancer imaging 3.8 Thierens et al. (1994) [24]
11C-GSK931145 Glycine transporter 1 4.0a Bullich et al. (2011) [25]
4.9b
11C-MeAIB Amino acid transport 4.0 Tolvanen et al. (2006) [26]
11C-PD153035 Epidermal growth factor receptor 4.0a Liu et al. (2009) [27]
5.2b
11C-MP4B Butyrylcholinesterase 4.2 Virta et al. (2008) [28]
11C-Choline Tumors and proliferative disorders 4.4 Tolvanen et al. (2010) [29]
11C-Carfentanil μ-Opiate receptor 4.6 Newberg et al. (2009) [30]
11C-Docetaxel Cancer imaging 4.7 van der Veldt et al. (2010) [31]
11C-PIB β-Amyloid plaque 4.7 Scheinin et al. (2007) [32]
11C-MPGA GABA receptor 4.8 Santens et al. (1998) [33]
11C-Acetate Cancer imaging 4.9 Seltzer et al. (2004) [34]
11C-Flumazenil GABAA receptor complex 5.0 Nugent et al. (2004) [35]
11C-Methionine Cancer imaging 5.2c Deloar et al. (1998) [36]
5.0d
11C-PIB β-Amyloid plaque 5.3 O’Keefe et al. (2009) [37]
11C-NNC112 Dopamine D1 receptor 5.7 Cropley et al. (2006) [38]
11C-PE2I Dopamine transporter 6.4 Ribeiro et al. (2007) [39]
11C-Mirtazapine Central adrenoceptor 6.8 Marthi et al. (2003) [40]
11C-WAY-100635 Serotonin1A receptor 12.2a Parsey et al. (2005) [6]
16.0b
Mean±SD 5.1±2.2
Range 3.0–16.0
Median 4.7

GABA γ-aminobutyric acid

a

Male

b

Female

c

Caucasian

d

Japanese

Based on the findings reviewed here we conservatively recommend that the upper limit of injected activity can be derived from the maximum reported effective dose in humans. For 11C ligands, the maximum effective dose (16 µSv/MBq) was found for 11C-WAY-100635 (Table 2). Although to our knowledge the International Commission on Radiological Protection (ICRP) has not proposed any radiation dose limits for volunteer subjects in human radiation experiments, including radionuclide metabolism studies and testing of new diagnostic agents, Institutional Radiation Safety Committees usually do establish upper limits. For instance, at the National Institutes of Health (NIH), the upper limit of effective dose for subjects participating in research radiation studies is 50 mSv [4]. Thus, the maximum injectable activity of 11C-WAY-100635 would be 3,125 MBq (84 mCi). It should be noted that even an activity more than 8 times lower (370 MBq or 10 mCi) would be sufficient to perform a study with an acceptable imaging quality. That is, we think that 370 MBq of any 11C-labeled radiopharmaceutical can be safely used for first-in-human studies without needing to conduct prior animal biodistribution studies.

Nevertheless, it is possible that even when the effective dose is low, a given tracer may deliver an excessive absorbed dose to a specific organ. To address this issue, Gatley [5] performed simulation studies to estimate an intravenously injected quantity of several 11C-labeled compounds that could not exceed a regulatory limit of 50 mSv on absorbed doses for individual organs. Upper limits on organ cumulative activities were estimated by assuming that 11C-Iabeled compounds are instantaneously distributed in the plasma and then transferred solely and irreversibly to a single organ. This approach would allow the assessment of the “worst case” estimate (although such an estimate would be obviously unrealistic), in order to conservatively plan initial human positron emission tomography (PET) imaging studies. Using an organ limit of 50 mSv, Gatley showed that a preliminary study with up to 130 MBq of 11C could be performed in humans without reaching this limit. This preliminary study would yield 11C arterial plasma data, which could in turn be used to give a refined upper limit on radiation absorbed doses [5].

In the case of 11C-WAY-100635, the limiting organ is the bladder, with an estimated dose of 0.167 mGy/MBq in men and 0.220 mGy/MBq in women [6]. Even for this tracer, an activity of up to 300 MBq in men and 227 MBq in women can be administered in a single injection before reaching 50 mSv, an organ limit established by the US Food and Drug Administration for radioligands studied under a Radioactive Drug Research Committee (21CFR361.1).

To safely perform first-in-human studies of new 11C-labeled tracers, we suggest the following design:

  1. Begin with whole-body scanning in a single human subject using 370 MBq (10 mCi). If we confirm that the radioactivity is fairly widely distributed in the body, we would know that no organ is likely to receive the theoretical maximal irradiation hypothesized by Gatley; therefore, higher activities may be injected.

  2. To determine whether the radioligand is worth pursuing, then do five to ten kinetic scans centered on the organ of interest (e.g., brain) using doses up to 740 MBq (20 mCi).

  3. If the radioligand looks promising, complete the human dosimetry study by acquiring a total of six to ten wholebody scans at 370 MBq (10 mCi) each.

Please note that 10 mCi is adequate for whole-body imaging because we quantify large regions, i.e., the whole organ. Higher injected activities (e.g., 20 mCi) are necessary for brain imaging because we measure subregions of brain and, more importantly, because we seek to measure rapidly declining concentrations of the parent radioligand in plasma.

Taken together, these data suggest that monkey dosimetric studies with 11C-labeled tracers are not only unreliable, but also unnecessary, as such studies can be safely performed in humans. We suggest that the very first human subject have a whole-body scan to confirm a fairly broad distribution in the body. Subsequent subjects may have dedicated imaging of the target organ as well as arterial blood sampling for rigorous compartmental modeling. Only after the tracer is deemed to be reasonably useful would additional whole-body studies be performed to estimate radiation doses from a larger sample size (five to ten subjects). One advantage of this pathway is that the radiation exposure to the larger sample of five to ten subjects would be avoided if the tracer is thought to lack utility based on rigorous compartmental analyses. Finally, our recommendation is made for 11C-labeled tracers because we had dosimetry data from biodistribution data in monkeys for many radioligands (i.e., nine). Nevertheless, similar arguments might also be made for 18F-labeled tracers.

References

  • 1.Sparks RB, Aydogan B. Comparison of the effectiveness of some common animal data scaling techniques in estimating human radiation dose. In: Stelson AS, editor. Sixth International Radiopharmaceutical Dosimetry Symposium; Associated Universities; Oak Ridge. 1999. pp. 705–716. [Google Scholar]
  • 2.Sprague DR, Fujita M, Ryu YH, Liow JS, Pike VW, Innis RB. Whole-body biodistribution and radiation dosimetry in monkeys and humans of the phosphodiesterase 4 radioligand [(11)C](R)-rolipram: comparison of two-dimensional planar, bisected and quadrisected image analyses. Nucl Med Biol. 2008;35:493–500. doi: 10.1016/j.nucmedbio.2008.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Brown AK, Fujita M, Fujimura Y, Liow J-S, Stabin M, Ryu YH, et al. Radiation dosimetry and biodistribution in monkey and man of 11C-PBR28, a PET radioligand to image inflammation. J Nucl Med. 2007;48:2072–2079. doi: 10.2967/jnumed.107.044842. [DOI] [PubMed] [Google Scholar]
  • 4.NIH RSC report: improving informed consent for research radiation studies, 10/17/2001.
  • 5.Gatley SJ. Estimation of upper limits on human radiation absorbed doses fromcarbon-11-labeled compounds. J Nucl Med. 1993;34:2208–2215. [PubMed] [Google Scholar]
  • 6.Parsey RV, Belanger MJ, Sullivan GM, Simpson NR, Stabin MG, Van Heertum R, et al. Biodistribution and radiation dosimetry of 11C-WAY100,635 in humans. J Nucl Med. 2005;46:614–619. [PubMed] [Google Scholar]
  • 7.Parsey RV, Sokol LO, Bélanger MJ, Kumar JS, Simpson NR, Wang T, et al. Amyloid plaque imaging agent [C-11]-6-OH-BTA-1: biodistribution and radiation dosimetry in baboon. Nucl Med Commun. 2005;26:875–880. doi: 10.1097/00006231-200510000-00005. [DOI] [PubMed] [Google Scholar]
  • 8.Thees S, Neumaier B, Glatting G, Deisenhofer S, von Arnim CA, Reske SN, et al. Radiation dosimetry and biodistribution of the beta-amyloid plaque imaging tracer 11C-BTA-1 in humans. Nuklearmedizin. 2007;46:175–180. [PubMed] [Google Scholar]
  • 9.Tipre DN, Fujita M, Chin FT, Seneca N, Vines D, Liow JS, et al. Whole-body biodistribution and radiation dosimetry estimates for the PET dopamine transporter probe 18F-FECNT in non-human primates. Nucl Med Commun. 2004;25:737–742. doi: 10.1097/01.mnm.0000133074.64669.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lu JQ, Ichise M, Liow JS, Ghose S, Vines D, Innis RB. Biodistribution and radiation dosimetry of the serotonin transporter ligand 11C-DASB determined from human whole-body PET. J Nucl Med. 2004;45:1555–1559. [PubMed] [Google Scholar]
  • 11.Hines CS, Liow JS, Zanotti-Fregonara P, Hirvonen J, Morse C, Pike VW, et al. Human biodistribution and dosimetry of 11C-CUMI-101, an agonist radioligand for serotonin-1a receptors in brain. PLoS One. 2011;6:e25309. doi: 10.1371/journal.pone.0025309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.NIMH/SNIDD tracer database initiative. [Accessed 17 Mar 2011]; http://pdsp.med.unc.edu/snidd/ [Google Scholar]
  • 13.Terry GE, Hirvonen J, Liow JS, Seneca N, Tauscher JT, Schaus JM, et al. Biodistribution and dosimetry in humans of two inverse agonists to image cannabinoid CB1 receptors using positron emission tomography. Eur J Nucl Med Mol Imaging. 2010;37:1499–1506. doi: 10.1007/s00259-010-1411-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Liow JS, Kreisl W, Zoghbi SS, Lazarova N, Seneca N, Gladding RL, et al. P-glycoprotein function at the blood-brain barrier imaged using 11C-N-desmethyl-loperamide in monkeys. J Nucl Med. 2009;50:108–115. doi: 10.2967/jnumed.108.056226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Seneca N, Zoghbi SS, Liow JS, Kreisl W, Herscovitch P, Jenko K, et al. Human brain imaging and radiation dosimetry of 11C-N-desmethyl-loperamide, a PET radiotracer to measure the function of P-glycoprotein. J Nucl Med. 2009;50:807–813. doi: 10.2967/jnumed.108.058453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hirvonen J, Roivainen A, Virta J, Helin S, Någren K, Rinne JO. Human biodistribution and radiation dosimetry of 11C-(R)-PK11195, the prototypic PET ligand to image inflammation. Eur J Nucl Med Mol Imaging. 2010;37:606–612. doi: 10.1007/s00259-009-1298-3. [DOI] [PubMed] [Google Scholar]
  • 17.Kumar A, Muzik O, Chugani D, Chakraborty P, Chugani HT. PET-derived biodistribution and dosimetry of the benzodiazepine receptor-binding radioligand (11)C-(R)-PK11195 in children and adults. J Nucl Med. 2010;51:139–144. doi: 10.2967/jnumed.109.066472. [DOI] [PubMed] [Google Scholar]
  • 18.Herscovitch P, Schmall B, Doudet D, Carson R, Eckelman WC. Biodistribution and radiation dose estimates for [C-11]raclopride. J Nucl Med. 1997;38:224P. [Google Scholar]
  • 19.Slifstein M, Hwang DR, Martinez D, Ekelund J, Huang Y, Hackett E, et al. Biodistribution and radiation dosimetry of the dopamine D2 ligand 11C-raclopride determined from human whole-body PET. J Nucl Med. 2006;47:313–319. [PubMed] [Google Scholar]
  • 20.Ribeiro MJ, Ricard M, Bourgeois S, Lièvre MA, Bottlaender M, Gervais P, et al. Biodistribution and radiation dosimetry of [11C] raclopride in healthy volunteers. Eur J Nucl Med Mol Imaging. 2005;32:952–958. doi: 10.1007/s00259-005-1783-2. [DOI] [PubMed] [Google Scholar]
  • 21.Graham MM, Peterson LM, Muzi M, Graham BB, Spence AM, Link JM, et al. 1-[Carbon-11]-glucose radiation dosimetry and distribution in human imaging studies. J Nucl Med. 1998;39:1805–1810. [PubMed] [Google Scholar]
  • 22.Laymon CM, Mason NS, Frankle WG, Carney JP, Lopresti BJ, Litschge MY, et al. Human biodistribution and dosimetry of the D2/3 agonist 11C-N-propylnorapomorphine (11C-NPA) determined from PET. J Nucl Med. 2009;50:814–817. doi: 10.2967/jnumed.108.058131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Treyer V, Streffer J, Ametamey SM, Bettio A, Bläuenstein P, Schmidt M, et al. Radiation dosimetry and biodistribution of 11C-ABP688 measured in healthy volunteers. Eur J Nucl Med Mol Imaging. 2008;35:766–770. doi: 10.1007/s00259-007-0638-4. [DOI] [PubMed] [Google Scholar]
  • 24.Thierens H, van Eijkeren M, Goethals P. Biokinetics and dosimetry for [methyl-11C]thymidine. Br J Radiol. 1994;67:292–295. doi: 10.1259/0007-1285-67-795-292. [DOI] [PubMed] [Google Scholar]
  • 25.Bullich S, Slifstein M, Passchier J, Murthy NV, Kegeles LS, Kim JH, et al. Biodistribution and radiation dosimetry of the glycine transporter-1 ligand (11)C-GSK931145 determined from primate and human whole-body PET. Mol Imaging Biol. 2011;13:776–784. doi: 10.1007/s11307-010-0398-6. [DOI] [PubMed] [Google Scholar]
  • 26.Tolvanen T, Någren K, Yu M, Sutinen E, Havu-Aurén K, Jyrkkiö S, et al. Human radiation dosimetry of [11C]MeAIB, a new tracer for imaging of system A amino acid transport. Eur J Nucl Med Mol Imaging. 2006;33:1178–1184. doi: 10.1007/s00259-006-0096-4. [DOI] [PubMed] [Google Scholar]
  • 27.Liu N, Li M, Li X, Meng X, Yang G, Zhao S, et al. PET-based biodistribution and radiation dosimetry of epidermal growth factor receptor-selective tracer 11C-PD153035 in humans. J Nucl Med. 2009;50:303–308. doi: 10.2967/jnumed.108.056556. [DOI] [PubMed] [Google Scholar]
  • 28.Virta JR, Tolvanen T, Någren K, Bruck A, Roivainen A, Rinne JO. 1-11C-methyl-4-piperidinyl-N-butyrate radiation dosimetry in humans by dynamic organ-specific evaluation. J Nucl Med. 2008;49:347–353. doi: 10.2967/jnumed.107.047233. [DOI] [PubMed] [Google Scholar]
  • 29.Tolvanen T, Yli-Kerttula T, Ujula T, Autio A, Lehikoinen P, Minn H, et al. Biodistribution and radiation dosimetry of [(11)C]choline: a comparison between rat and human data. Eur J Nucl Med Mol Imaging. 2010;37:874–883. doi: 10.1007/s00259-009-1346-z. [DOI] [PubMed] [Google Scholar]
  • 30.Newberg AB, Ray R, Scheuermann J, Wintering N, Saffer J, Schmitz A, et al. Dosimetry of 11C-carfentanil, a micro-opioid receptor imaging agent. Nucl Med Commun. 2009;30:314–318. doi: 10.1097/MNM.0b013e328329a0ec. [DOI] [PubMed] [Google Scholar]
  • 31.van der Veldt AA, Hendrikse NH, Smit EF, Mooijer MP, Rijnders AY, Gerritsen WR, et al. Biodistribution and radiation dosimetry of 11C-labelled docetaxel in cancer patients. Eur J Nucl Med Mol Imaging. 2010;37:1950–1958. doi: 10.1007/s00259-010-1489-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Scheinin NM, Tolvanen TK, Wilson IA, Arponen EM, Någren KA, Rinne JO. Biodistribution and radiation dosimetry of the amyloid imaging agent 11C-PIB in humans. J Nucl Med. 2007;48:128–133. [PubMed] [Google Scholar]
  • 33.Santens P, DeVos F, Thierens H, Decoo D, Slegers G, Dierckx RA, et al. Biodistribution and dosimetry of carbon-11-methoxyprogabidic acid, a possible ligand for GABA-receptors in the brain. J Nucl Med. 1998;39:307–310. [PubMed] [Google Scholar]
  • 34.Seltzer MA, Jahan SA, Sparks R, Stout DB, Satyamurthy N, Dahlborn M, et al. Radiation dose estimates in humans for (11) C-acetate whole-body PET. J Nucl Med. 2004;45:1233–1236. [PubMed] [Google Scholar]
  • 35.Nugent AC, Neumeister A, Drevets WC, Eckelman WC, Channing MA, Herscovitch P. Human biodistribution and dosimetry of the PET benzodiazepine receptor ligand [11C]flumazenil. J Nucl Med. 2004;45(Suppl):434P. [Google Scholar]
  • 36.Deloar HM, Fujiwara T, Nakamura T, Itoh M, Imai D, Miyake M, et al. Estimation of internal absorbed dose of L-[methyl-11C] methionine using whole-body positron emission tomography. Eur J Nucl Med. 1998;25:629–633. doi: 10.1007/s002590050265. [DOI] [PubMed] [Google Scholar]
  • 37.O’Keefe GJ, Saunder TH, Ng S, Ackerman U, Tochon-Danguy HJ, Chan JG, et al. Radiation dosimetry of beta-amyloid tracers 11C-PiB and 18F-BAY94-9172. J Nucl Med. 2009;50:309–315. doi: 10.2967/jnumed.108.055756. [DOI] [PubMed] [Google Scholar]
  • 38.Cropley VL, Fujita M, Musachio JL, Hong J, Ghose S, Sangare J, et al. Whole-body biodistribution and estimation of radiationabsorbed doses of the dopamine D1 receptor radioligand, (11)C-NNC 112, in humans. J Nucl Med. 2006;47:100–104. [PubMed] [Google Scholar]
  • 39.Ribeiro MJ, Ricard M, Lièvre MA, Bourgeois S, Emond P, Gervais P, et al. Whole-body distribution and radiation dosimetry of the dopamine transporter radioligand [(11)C]PE2I in healthy volunteers. Nucl Med Biol. 2007;34:465–470. doi: 10.1016/j.nucmedbio.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 40.Marthi K, Hansen SB, Jakobsen S, Bender D, Smith SB, Smith DF. Biodistribution and radiation dosimetry of [N-methyl-11C]mirtazapine, an antidepressant affecting adrenoceptors. Appl Radiat Isot. 2003;59:175–179. doi: 10.1016/s0969-8043(03)00156-8. [DOI] [PubMed] [Google Scholar]

RESOURCES