throbber
ARTICLE IN PRESS
`
`Applied Radiation and Isotopes 65 (2007) 301–308
`
`www.elsevier.com/locate/apradiso
`
`On the preparation of a therapeutic dose of 177Lu-labeled
`DOTA–TATE using indigenously produced 177Lu in medium
`flux reactor
`
`Tapas Das, Sudipta Chakraborty, Sharmila Banerjee
`
`
`
`, Meera Venkatesh
`
`Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai-400085, India
`
`Received 26 June 2006; received in revised form 1 September 2006; accepted 29 September 2006
`
`Abstract
`177Lu could be produced with a specific activity of 23,000 mCi/mg (850 GBq/mg) by neutron activation using enriched 176Lu (64.3%)
`target when irradiation was carried out at a thermal neutron flux of 1  1014 n/cm2/s for 21 d. 177Lu–DOTA–TATE could be prepared in
`high radiochemical yield (99%) and adequate stability using the 177Lu produced indigenously. The average level of radionuclidic
`impurity burden in 177Lu due to 177mLu was found to be 250 nCi of 177mLu/1 mCi of 177Lu (9.25 kBq/37 MBq) at the end of
`bombardment, which corresponds to 0.025% of the total activity produced. The maximum specific activity achievable via careful
`optimization of
`the irradiation parameters was found to be adequate for the preparation of a therapeutic dose of
`the
`radiopharmaceutical. The in-house preparation of this agent using 25 mg (17.41 nmole) of DOTA–TATE and indigenously produced
`177Lu (0.8 mg, 4.52 nmole), corresponding to peptide/Lu ratio of 3.85 yielded 98.7% complexation. Allowing possibility of decay due to
`transportation to users, it has been possible to demonstrate that at our end, a single patient dose of 150–200 mCi (5.55–7.40 GBq) can be
`prepared by using 250–333 mg of DOTA–TATE conjugate. This amount compares well with 177Lu–DOTA–TATE prepared for a typical
`peptide receptor radionuclide therapy (PRRT) procedure which makes use of 100 mg of the DOTA–TATE conjugate, which incorporates
`50 mCi (1.85 GBq) of 177Lu activity, thereby implying that in order to achieve a single patient dose of 150–200 mCi (5.55–7.40 GBq),
`300–400 mg of the conjugate needs to be used.
`r 2006 Elsevier Ltd. All rights reserved.
`
`Keywords: PRRT; 177Lu; DOTA–TATE; Somatostatin receptors
`
`1. Introduction
`
`Radiometallated peptides which exhibit high specificity
`for cognate receptors over-expressed on cancerous lesions,
`offer important potential as site-directed diagnostic and
`therapeutic radiopharmaceuticals (Boerman et al., 2000;
`Breeman et al., 2001; Britton, 1997; Eckelman and Gibson,
`1993). The highly specific biochemical and physiologic
`binding capabilities of the receptor-avid peptides can be
`exploited for using these agents as a vehicle to target the
`delivery of the radioactivity to the cells over-expressing
`similar type of receptors after radiolabeling the peptides
`with the radionuclide of interest. Such peptide analogs
`
`
`
`Corresponding author. Tel.: +91 22 2559 0616; fax: +91 22 2550 5345.
`E-mail address: sharmila@barc.gov.in (S. Banerjee).
`
`0969-8043/$ - see front matter r 2006 Elsevier Ltd. All rights reserved.
`doi:10.1016/j.apradiso.2006.09.011
`
`when labeled with therapeutic radionuclides are being
`actively investigated as agents for use in peptide receptor
`radionuclide therapy (PRRT) (Kwekkeboom et al., 2000,
`2003a, 2005; de Jong et al., 2005).
`Among the several classes of radiolabeled peptides,
`radiolabeled somatostatin analogs have been proved to be
`the most promising. Octreotide, a metabolically stable
`analog of native somatostatin (Pauwels et al., 1998), has
`been radiolabeled with 111In using a bifunctional chelating
`agent and the resultant radiolabeled peptide, 111In–DTPA-
`Octreotide (commonly known as OctreoScans) has been
`successfully employed in clinical diagnosis of somatostatin
`receptor positive tumors (Pauwels et al., 1998; Krenning
`et al. 1992, 1993). Efforts are being made to develop suitable
`therapeutic analogs having the potential to eradicate the
`cancerous lesions over-expressing somatostation receptors.
`
`Evergeen Ex. 1031
`1 of 8
`
`

`

`302
`
`T. Das et al. / Applied Radiation and Isotopes 65 (2007) 301–308
`
`ARTICLE IN PRESS
`
`Recently started clinical trials with (177Lu–DOTA–Tyr3)-
`Octreotide (known as 177Lu–DOTA–TOC) and (177Lu–
`DOTA–Tyr3)-Octreotate (known as 177Lu–DOTA–TATE)
`have shown very encouraging results in terms of tumor
`regression in patients suffering from different types of
`neuroendocrine tumors, which are known to over-express
`somatostatin receptors (Kwekkeboom et al., 2003a, b,
`2005; Forrer et al. 2005). However,
`it is reported that
`[DOTA–Tyr3]-Octreotate has a ninefold higher affinity for
`the somatostatin receptor subtype 2 as compared with
`[DOTA–Tyr3]-Octreotide and therefore, the later agent is
`expected to be more potent for carrying out PRRT in patients
`suffering from neuroendocrine tumors (Kwekkeboom et al.,
`2003a; Reubi et al. 2000).
`177Lu is presently being considered as a potential
`radionuclide for use in in vivo therapy, because of its
`favorable decay characteristics. 177Lu decays with a half-
`life of 6.73 d by emission of b- particles with maximum
`energies of 497 keV (78.6%), 384 keV (9.1%), and 176 keV
`(12.2%) to stable 177Hf (Firestone, 1996). The emission of
`suitable energy g photons of 113 keV (6.4%) and 208 keV
`(11%) (Firestone, 1996) with relatively low abundances
`provides
`the opportunity to carry out
`simultaneous
`scintigraphic studies, which helps to monitor the proper
`in vivo localization of the injected radiopharmaceutical as
`well as to perform dosimetric evaluations. An important
`aspect for the countries with limited reactor facility is the
`177Lu which provides
`comparatively long half-life of
`logistic advantage towards facilitating supply to locations
`far away from the reactors (Das et al., 2002). Besides this,
`the high thermal neutron capture cross-section (s ¼ 2100b)
`of (176Lu(n,g)177Lu) reaction (Firestone, 1996) ensures that
`177Lu can be produced with sufficiently high specific
`activity using the moderate flux reactors. In fact, the
`cross-section is the highest encountered among all (n,g)
`produced radionuclides presently used for therapy. These
`favorable nuclear parameters ensure that there will be no
`constraints with respect to large-scale isotope production.
`In connection with our ongoing research on the
`development of novel agents for radiotherapy of tumors
`(Banerjee et al., 2004; Das et al., 2004), production of
`177Lu,
`identified as
`the ideal
`radioisotope is being
`extensively optimized with an aim to achieving the
`maximum specific activity via a cost-effective route.
`This is an essential requirement for designing agents for
`PRRT since the primary criterion is to deliver sufficient
`number of radionuclides to the receptors over-expressed on
`the targeted tumor site without saturating the target
`(Mausner and Srivastava, 1993; Volkert and Hoffman,
`1999; Breeman et al., 2003).
`177Lu can be produced by two different routes, namely,
`by irradiation of Lu target and by irradiation of Yb target
`followed by radiochemical separation of 177Lu from Yb
`isotopes. Though the latter method enables production of
`no-carrier-added (NCA) 177Lu, the radiochemical separa-
`tion of 177Lu activity from irradiated Yb target is a difficult
`task owing to the similarity in the chemistry of the two
`
`adjacent members of the lanthanide series. Presence of Yb
`in substantial amounts, attributable to its low cross-
`section, besides reducing the effective specific activity of
`the product, will also compete with 177Lu in the complexa-
`tion procedure unless effectively eliminated from the
`irradiated target. This is one of the major impediment in
`
`the ready production of NCA 177Lu via 176Yb(n,g,b
`)177Lu
`route. Moreover, use of enriched targets with low activation
`cross-section (s ¼ 2.4b for
`176Yb(n,g)177Yb)
`(Firestone,
`1996) is not economical for isotope production, as a
`significant part of the target remains unutilized. Though,
`176Yb recovery is theoretically feasible, the aforementioned
`practical problems associated with the cumbersome separa-
`tion still remains.
`177Lu have
`in the production of
`Our experiences
`provided an insight towards envisaging 177Lu–DOTA–
`TATE as a radiopharmaceutical, which can be indigen-
`ously produced and supplied to local users as an agent for
`PRRT. The primary aim in the designing of this agent
`attempts to maximize the specific activity of the resultant
`radiolabeled preparation. Towards this, 177Lu production
`is envisaged via (n,g) using an enriched Lu2O3 target and
`efforts are directed to obtain 177Lu with maximum possible
`specific activity utilizing the moderate flux reactor available
`in our country. However, careful optimization of the
`irradiation parameters is required for obtaining 177Lu in
`adequate specific activity for PRRT applications, particu-
`larly when production is envisaged at high flux positions
`owing to the high thermal neutron capture cross-section of
`176Lu which eventually leads to considerable target burn
`up. The present paper describes optimization of 177Lu
`production with sufficiently high specific activity using a
`moderate flux reactor and successful preparation of patient
`dose of 177Lu–DOTA–TATE using the 177Lu indigenously
`produced at our end.
`
`2. Materials and methods
`
`Lutetium oxide (64.3% enriched in 176Lu, spectroscopic
`grade, 499.99% pure) was obtained from Isoflex, Russia.
`DOTA–TATE was obtained from PiChem, Finland,
`through International Atomic Energy Agency (IAEA) as
`a part of a coordinated research project
`(CRP). All
`chemicals and solvents used in the experiments were of
`AR grade and supplied by reputed chemical manufac-
`turers. Radionuclidic purity of 177Lu was ascertained by
`high resolution g-ray spectrometry using an HPGe detector
`(EGG Ortec/Canberra detector) coupled to a 4K multi-
`channel analyzer
`(MCA)
`system after
`radiochemical
`processing. 152Eu reference source used for energy and
`efficiency calibration of the detector was obtained from
`Amersham Inc., USA. All other radioactivity measure-
`ments were carried out using a well
`type NaI (Tl)
`scintillation counter after adjusting the 150 keV baseline
`and keeping a window of 100 keV, thereby utilizing the
`208 keV g photon of 177Lu. Whatman 3 mm chromato-
`graphy paper (UK) was used for paper chromatography
`
`Evergeen Ex. 1031
`2 of 8
`
`

`

`ARTICLE IN PRESS
`
`T. Das et al. / Applied Radiation and Isotopes 65 (2007) 301–308
`
`303
`
`studies. The high performance liquid chromatography
`(HPLC) system used was obtained from JASCO (PU
`1580), Japan, equipped with a PU 1575 UV/VIS detector.
`A well-type NaI (Tl) scintillation detector was coupled to
`the system for radioactivity measurements in the eluate. All
`the solvents used for HPLC analyses were of HPLC grade
`and purchased from reputed local manufacturers, degassed
`and filtered prior to use.
`
`2.1. Production and radiochemical processing of 177Lu
`
`177Lu was produced by thermal neutron bombardment
`on isotopcially enriched (64.3% in 176Lu) Lu2O3 target. A
`stock solution of enriched target was prepared by dissol-
`ving enriched Lu2O3 powder in 0.1 M HCl (1 mg/mL
`concentration). A known aliquot of this solution was taken
`in a quartz ampoule and carefully evaporated to dryness.
`The ampoule was subsequently flame sealed and irradiated
`after placing inside an aluminum can. Irradiations were
`carried out at different available flux positions (1.4 
`1013–1.0  1014 n/cm2/s) for different durations (7–21 d) in
`order to optimize the irradiation conditions towards
`achieving maximum specific activity of 177Lu. The irra-
`diated target was dissolved in 1 M HCl by gentle warming
`inside a lead-shielded plant. The resultant solution was
`evaporated to near dryness and reconstituted in de-ionized
`water. The pH of the 177LuCl3 solution thereby obtained
`was adjusted to 4 prior to complexation studies. A known
`aliquot was drawn for assay of radioactivity and determi-
`nation of radionuclidic purity.
`Radioactivity assay was carried out by measuring the
`ionization current obtained when an aliquot of the batch
`was placed inside a pre-calibrated well-type ion-chamber.
`Radionuclidic purity was determined by recording g-ray
`spectra of
`the appropriately diluted solution of
`the
`irradiated target using an HPGe detector connected to a
`4 K MCA system. Energy as well as efficiency calibration of
`the detector were carried out using a 152Eu reference source
`prior to the recording of g-ray spectra. Several spectra were
`recorded for each batch at regular time intervals. Samples
`measured initially for the assay of 177Lu were preserved for
`complete decay of 177Lu (8–10 T1/2 of 177Lu, i.e. for a period
`of 50–70 d) and re-assayed to determine the activity of long-
`lived 177mLu (T1/2 ¼ 160.5 d). Appropriately diluted sample
`
`solutions were counted for 1 h.
`
`2.2. Preparation of 177Lu–DOTA–TATE complex
`
`177Lu-labeled DOTA–TATE
`the preparation of
`For
`complex, a stock solution of the DOTA–TATE was prepared
`by dissolving DOTA–TATE conjugate in HPLC grade water
`with a concentration of 1 mg/mL. To a 25 mL aliquot of this
`stock solution, 25 mL of 177LuCl3 solution was added and the
`volume was made upto 200 mL using the 0.1 M ammonium
`acetate buffer of pH 5. The reaction mixture was incubated
`at 80 1C for 1 h after adjusting the pH to 4.5–5. Various
`parameters, such as, ligand concentration, incubation time,
`
`and temperature were varied extensively in order to arrive at
`the protocol for maximum complexation.
`
`2.3. Quality control techniques
`
`The characterization of the 177Lu-labeled DOTA–TATE
`conjugate as well as the determination of the complexation
`yield was carried out by paper chromatography (PC) as
`well as by HPLC techniques.
`
`2.3.1. Paper chromatography (PC)
`5 mL portions of the test solutions were applied at 1.5 cm
`from the lower end of the chromatography paper strips.
`The strips were developed in 50% aqueous acetonitrile,
`dried, cut into segments of 1 cm each and the radioactivity
`associated with each segment was measured in a NaI (Tl)
`detector.
`
`2.3.2. High performance liquid chromatography (HPLC)
`HPLC of the 177Lu-labeled conjugate was carried out
`using a dual pump HPLC unit with a C-18 reversed phase
`HiQ-Sil (5 mm, 25  0.46 cm) column. The elution was
`monitored both by detecting UV signals at 270 nm as well
`as by radioactivity signal using NaI (Tl) detector. Water
`(A), and acetonitrile (B) mixtures with 0.1% trifluoroacetic
`acid were used as the mobile phase and the following
`gradient elution technique was adopted for the separation
`(0–4 min 95% A, 4–15 min 95% A to 5% A, 15–20 min 5%
`A, 20–25 min 5% A to 95% A, 25–30 min 95% A). Flow
`rate was maintained at 1 mL/min.
`
`2.4. Maximization of specific activity of
`177Lu–DOTA–TATE complex
`
`In order to obtain 177Lu-labeled DOTA–TATE con-
`jugate in highest possible specific activity, experiments were
`carried out to achieve maximum complexation yield of
`177Lu-DOTA–TATE at
`the minimum possible [L]/[M]
`ratio. For this, 25 mg of the peptide (already optimized
`ligand amount) was allowed to react with different
`amounts of Lu ranging from 0.2 to 1.6 mg under the same
`reaction conditions described earlier.
`
`2.5. Stability of 177Lu–DOTA–TATE
`
`The in vitro stability of the 177Lu-labeled conjugate was
`ascertained by storing the radiolabeled product at room
`temperature and determining the radiochemical purity at
`different time intervals post-preparation employing the
`standard quality control techniques described earlier.
`
`3. Results and discussions
`
`3.1. Production of 177Lu
`
`177Lu was produced by thermal neutron bombardment
`on isotopically enriched (64.3% in 176Lu) Lu2O3 target in
`
`Evergeen Ex. 1031
`3 of 8
`
`

`

`304
`
`T. Das et al. / Applied Radiation and Isotopes 65 (2007) 301–308
`
`ARTICLE IN PRESS
`
`Dhruva reactor at our Institute. The radionuclidic purity of
`177Lu produced was determined by analyzing the g-ray
`spectrum and found to be 99.975%. A typical g-ray
`spectrum recorded after the radiochemical processing of
`the irradiated target exhibited major g photopeaks at 72,
`113, 208, 250, and 321 keV. All of these photopeaks
`correspond to that of 177Lu (Firestone, 1996). This was
`further confirmed from the decay as followed by monitor-
`ing peak area cps values at those peaks according to the
`half-life of 177Lu. It is well documented in the literature
`that there is a possibility of formation of 177mLu (T1/2
`¼ 160.5 d) on thermal neutron bombardment of Lu target
`(Neves et al., 2002; Pillai et al., 2003; Nir-El, 2004).
`However, g-ray spectrum of the irradiated Lu target after
`chemical processing did not show any significant peak
`corresponding to the photopeaks of 177mLu (128, 153, 228,
`378, 414, 418 keV) (Firestone, 1996). This can be explained
`from the fact that the radioactivity due to 177mLu produced
`will be insignificant compared to that of 177Lu, owing to its
`long half-life
`and comparatively
`low cross-section
`(s ¼ 7 barns) (Firestone, 1996) for its formation. The trace
`level of 177mLu produced was determined by recording the
`g-ray spectrum of a sample aliquot, initially having high
`radioactive concentration, after complete decay of 177Lu
`activity (8–10 T1/2 of 177Lu, i.e. for a period of 50–70 d).
`The average level of radionuclidic impurity burden in 177Lu
`due to 177mLu determined by this technique was found to
`be 250 nCi of 177mLu/1 mCi of 177Lu (9.25 kBq/37 MBq) at
`EOB, which corresponds to 0.025% of the total activity
`produced.
`In order to produce 177Lu with maximum possible
`specific activity using the moderate flux reactor available at
`our end, Lu2O3 target was irradiated at different available
`flux positions for different periods of time. Four different
`flux positions with thermal neutron flux ranging between
`1.4  1013 and 1.0  1014 n/cm2/s were available for irradia-
`tion. Since the normal irradiation schedule of this multi-
`facility reactor allowed irradiation of the target only in
`
`multiples of 7 d, irradiations were carried out for 7, 14, and
`the 177Lu obtained on
`21 d. The specific activity of
`irradiation of enriched target for varied time durations
`and at different
`thermal neutron flux positions are
`tabulated in Table 1. A maximum specific activity of
`23000 mCi/mg (850 GBq/mg) was achieved when irradia-
`tion was carried out at a thermal neutron flux of 1  1014 n/
`cm2/s for 21 d, which corresponds to 21% of
`the
`maximum achievable specific activity. It is evident from
`Table 1 that the specific activities of 177Lu obtained were
`significantly higher compared to the theoretically calcu-
`lated values accounting for only thermal neutron capture.
`A possible reason for practically obtaining an activity
`higher
`than theoretically calculated values could be
`attributed to the contribution from epithermal neutrons
`(resonance integral ¼ 1087b), which is not accounted for in
`theoretical calculations (Pillai et al., 2003; Nir-El, 2004;
`Knapp et al., 1995). However, the contribution from
`epithermal neutron alone could not provide a satisfactory
`explanation for obtaining 2.5–2.8 times higher specific
`activity of 177Lu than that of
`theoretically calculated
`values.
`Apart from the use of enriched target and choosing
`the highest available neutron flux position in a reactor,
`the other factor, which can be optimized for achieving the
`maximum possible specific activity of a radioisotope
`produced by neutron activation,
`is
`the duration of
`irradiation. In most cases of radioisotope production using
`(n,g) route, irradiation of the target 4–6 times of the half-
`life of the radioisotope being produced results in maximum
`specific activity. However, the same does not hold good in
`case of production of 177Lu by neutron capture, particu-
`larly, when the production is being carried out at a
`comparatively higher neutron flux position, owing to the
`high thermal neutron capture cross section of 177Lu. In this
`case, a careful optimization of the time of irradiation needs
`to be considered in order to obtain the highest specific
`activity (Pillai et al., 2003). In high flux positions of a
`
`Table 1
`Specific activity of 177Lu obtained at different thermal neutron flux for irradiation of different durations
`
`Neutron flux (n/cm2/s)
`
`Irradiation time (d)
`
`Activity obtained
`(mCi/mg) (GBq/mg)
`
`Theoretical activity
`(mCi/mg) (GBq/mg)
`
`Experimental/theoretical
`
`1.4  1013
`
`3.0  1013
`
`3.0  1013
`
`6.75  1013
`
`6.75  1013
`
`1.0  1014
`
`7
`
`7
`
`14
`
`7
`
`14
`
`21
`
`23227102
`85.973.8
`
`46357269
`171.579.9
`
`74607253
`276.079.4
`
`110007782
`407.0728.9
`
`1775071476
`656.8754.6
`
`231857658
`857.8724.3
`
`846
`31.3
`
`1814
`67.1
`
`2696
`99.8
`
`4081
`151.0
`
`6406
`237.0
`
`8622
`319.0
`
`2.74
`
`2.56
`
`2.77
`
`2.70
`
`2.77
`
`2.69
`
`Evergeen Ex. 1031
`4 of 8
`
`

`

`ARTICLE IN PRESS
`
`T. Das et al. / Applied Radiation and Isotopes 65 (2007) 301–308
`
`305
`
`3.2. Characterization of 177Lu–DOTA–TATE
`
`In PC using 50% aqueous acetonitrile, the activity
`corresponding to 177Lu–DOTA–TATE complex moved
`towards the solvent front with Rf ¼ 0.8–0.9, while un-
`complexed 177Lu remained at the point of spotting (Rf ¼ 0)
`under identical conditions. The PC patterns of 177LuCl3
`and 177Lu–DOTA–TATE complex are shown in Fig. 2.
`The 177Lu-labeled DOTA–TATE complex was also
`characterized by HPLC studies. Fig. 3(a) shows the typical
`HPLC pattern of 177Lu-labeled DOTA–TATE conjugate
`prepared under optimized conditions. Fig. 3(b) shows the
`HPLC pattern of 177LuCl3 under identical conditions. The
`actual extent of complexation achieved was also deter-
`mined from the HPLC studies.
`
`3.3. Optimization of complexation yield of
`177Lu–DOTA–TATE
`
`Various parameters such as, conjugate concentration,
`pH of the reaction mixture, reaction time and temperature
`were varied extensively in order to achieve maximum
`complexation yield. Keeping the reaction volume as
`200 mL, the amount of DOTA–TATE was varied from 5
`to 100 mg in order to determine the optimum ligand
`concentration required for obtaining maximum complexa-
`tion. It was observed that a minimum of 25 mg DOTA–
`TATE was required to obtain a complexation yield of
`99%. The reaction was carried out by incubating the
`reaction mixture at three different temperatures (room
`temperature, 50 and 80 1C) for different time periods
`(5 min, 15 min, 30 min, 1 h, and 2 h) in order to determine
`optimum reaction time and temperature. It was observed
`that, maximum complexation was achieved when the
`reaction mixture was incubated at 80 1C for a period of
`1 h. It
`is well documented in the literature that
`the
`
`177Lu-DOTA-TATE
`
`177LuCl3
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`% Radioactivity
`
`0
`
`1
`
`2
`3
`4
`5
`Migration from point of spotting (cm)
`
`6
`
`7
`
`Fig. 2. Paper chromatography patterns of 177LuCl3 and 177Lu–DOTA–
`TATE.
`
`Evergeen Ex. 1031
`5 of 8
`
`reactor, the target burn up will be considerably higher
`owing to the high thermal neutron capture cross section
`of 176Lu and hence, the usual assumption that the number
`of target atoms remains constant during the period of
`irradiation will not be valid in this case. Considering the
`fact that the number of target atoms is not fixed but a
`function of time, the commonly used differential equation
`used for the calculation of the activity produced i.e.
`
`dN 2=dt ¼ N 1sf N 2l
`
`(1)
`
`can be modified as
`
`sftsf N 2l,
`dN 2=dt ¼ N 0e
`(2)
`where N0 is the number of 176Lu atoms used as target (at
`t ¼ 0), N1 the number of 176Lu atoms at any time t, N2 the
`number of 177Lu atoms at any time t, l the decay constant
`of 177Lu, s the thermal neutron capture cross section of
`176Lu, f the thermal neutron flux of the reactor and t the
`time of
`irradiation. Solution of
`the above mentioned
`modified differential equation leads
`to the following
`mathematical expression, which enables calculation of the
`177Lu activity (A) produced at EOB, taking into account
`that the number of the target atoms is also a function of
`time of irradiation.
`½e
`sft e
`A ¼ N 0lsf
`l sf
`Fig. 1 shows the variation of the theoretically calculated
`specific activity of 177Lu obtained at EOB with irradiation
`time at 1  1014 n/cm2/s thermal neutron flux. It is evident
`that the activity of 177Lu produced will be maximum after
`21 d irradiation, beyond which, the activity will decrease
`owing to the high target burn up. Therefore, in order to
`obtain maximum specific activity of 177Lu using the
`maximum available flux position in our reactor, the time
`of irradiation was fixed as 21 d.
`
`ltŠ
`
`(3)
`
`0
`
`7
`
`14
`Duration of irradiation (d)
`
`21
`
`28
`
`325
`
`300
`
`275
`
`250
`
`225
`
`200
`
`175
`
`150
`
`125
`
`100
`
`Specific activity of177Lu (GBq/mg)
`
`Fig. 1. Variation of theoretically calculated specific activity of 177Lu
`obtained at EOB with time of irradiation at thermal neutron flux of
`1  1014 n/cm2/s.
`
`

`

`306
`
`T. Das et al. / Applied Radiation and Isotopes 65 (2007) 301–308
`
`ARTICLE IN PRESS
`
`is kept constant at this value and complexations were
`carried out using different amounts of Lu ranging from 0.2
`to 1.6 mg under the same reaction conditions described
`earlier. The complexation yield in each case was deter-
`mined by PC as well as by HPLC. The complexation yields
`obtained at different concentrations of Lu are given in
`Table 2. It is evident from Table 2, that a radiolabeling
`yield of 99% could be achieved using a ligand/metal ratio
`4. This implies that 0.8 mg of Lu can be incorporated in
`25 mg of the peptide without compromising the extent of
`complexation. Considering the maximum possible specific
`activity of 177Lu achievable at our end to be 23,000 mCi/mg
`(850 GBq/mg), 0.8 mg of Lu could provide 18.4 mCi
`(0.68 GBq) of activity. Therefore, a single patient dose of
`150–200 mCi (5.55–7.40 GBq) can be prepared by using
`200–275 mg of peptide. In order to carry out a comparative
`evaluation of the radiolabeled DOTA–TATE prepared by
`us, an analysis of a reported case (Kwekkeboom et al.,
`2003a, 2001) was carried out. It has been shown that
`177Lu–DOTA–TATE prepared for a typical PRRT pro-
`cedure uses 100 mg of
`the DOTA–TATE conjugate
`which incorporates 50 mCi (1.85 GBq) of 177Lu activity
`(Kwekkeboom et al., 2001). This implies that to achieve a
`single patient dose of 150–200 mCi
`(5.55–7.40 GBq),
`300–400 mg of the conjugate has been used. Considering
`the practical aspects of time required for Post-irradiation
`radiochemical processing, preparation and quality control
`of the radiopharmaceutical and subsequent transportation
`to the nuclear medicine facilities, 15 mCi (0.55 GBq) of
`177Lu activity in 25 mg of DOTA–TATE can be delivered to
`the nuclear medicine facilities. Therefore, the single patient
`dose of 150–200 mCi (5.55–7.40 GBq) can be prepared by
`using 250–333 mg of DOTA–TATE conjugate at our end,
`which is comparable or possibly less than the reported
`values.
`
`3.5. Stability of 177Lu–DOTA–TATE
`
`The stability of the 177Lu-labeled DOTA–TATE complex
`was studied by employing standard quality control techni-
`ques mentioned earlier. It was observed that the radi-
`olabeled conjugate retained its radiochemical purity after
`7 d of its preparation when stored at room temperature.
`
`Table 2
`Complexation yield of 177Lu–DOTA–TATE at different [L]/[M] ratio
`
`[DOTA–TATE]
`
`[Lu]
`
`[L]/[M]
`
`Complexation
`yield (%)
`
`25 mg
`(17.41 nmole)
`25 mg
`(17.41 nmole)
`25 mg
`(17.41 nmole)
`25 mg
`(17.41 nmole)
`
`0.2 mg
`(1.13 nmole)
`0.4 mg
`(2.26 nmole)
`0.8 mg
`(4.52 nmole)
`1.6 mg
`(9.04 nmole)
`
`15.41
`
`7.70
`
`3.85
`
`1.92
`
`99.2
`
`99.0
`
`98.7
`
`81.2
`
`1000
`
`800
`
`600
`
`400
`
`200
`
`0
`
`0
`
`300
`
`1200
`900
`600
`Retention time (s)
`
`1500
`
`1800
`
`CPS
`
`(a)
`
`400
`
`300
`
`200
`
`CPS
`
`100
`
`0
`
`0
`
`300
`
`(b)
`
`1200
`900
`600
`Retention time (s)
`
`1500
`
`1800
`
`Fig. 3. HPLC patterns of (a) 177Lu–DOTA–TATE and (b) 177LuCl3.
`
`maximum complexation is obtained when 177Lu labeling of
`the biomolecules are carried out at pH5 (Breeman et al.,
`2003; Milenic et al., 2002; Stimmel and Kull, 1998). In the
`present case,
`the maximum complexation yield was
`achieved at pH 4.5–5.
`
`3.4. Maximization of the specific activity of
`177Lu–DOTA–TATE
`
`Bearing in mind the importance of preparing radiola-
`beled agents intended for targeted use in binding with
`receptors over-expressed on tumors, efforts were directed
`to prepare the 177Lu-DOTA–TATE with maximum possi-
`ble specific activity, using the 177Lu available at our end.
`Towards achieving this, the concentration of the peptide
`conjugate used for a typical labeling procedure needs to be
`minimum which,
`in turn, would aim at the minimum
`possible ligand/metal ratio, in order that unlabeled peptide
`which would otherwise bind also with the targeted
`receptors will be minimum. The ligand/metal ratio can be
`minimized by keeping the concentration of the peptide
`constant and gradually increasing the amount of radio-
`metal in the reaction mixture. Since 25 mg of the peptide
`was found to be the optimum amount required for
`obtaining 99% complexation, the ligand concentration
`
`Evergeen Ex. 1031
`6 of 8
`
`

`

`ARTICLE IN PRESS
`
`T. Das et al. / Applied Radiation and Isotopes 65 (2007) 301–308
`
`307
`
`The complex prepared using maximum possible amount of
`Lu (0.8 mg) also showed excellent stability till 7 d when
`stored at identical conditions.
`
`4. Conclusion
`
`Production of 177Lu by the (n,g) route from enriched
`176Lu target with a specific activity of 23000 mCi/mg
`(850 GBq/mg) was achievable when irradiation was carried
`out at a thermal neutron flux of 1  1014 n/cm2/s for 21 d.
`177Lu–DOTA–TATE could be prepared in high radio-
`chemical yield and adequate stability using the 177Lu
`produced indigenously. The maximum specific activity
`achievable via careful optimization of
`the irradiation
`parameters was found to be adequate for the preparation
`of a therapeutic dose of the radiopharmaceutical. The in-
`house preparation of
`this agent using indigenously
`produced 177Lu was found to be a more cost-effective
`route over the method involving use of 177Lu from
`commercial sources. Clinical studies with this agent will
`be carried out after obtaining clearances from relevant
`regulatory authorities.
`
`Acknowledgments
`
`The authors sincerely acknowledge the support provided
`by the International Atomic Energy Agency (IAEA) in the
`form of a coordinated research project (CRP). The authors
`express their sincere thanks to Dr. V. Venugopal, Director,
`Radiochemistry and Isotope Group for his valuable
`suggestions and constant encouragement through out the
`course of the work. The sincere help received from our
`colleagues, Dr. S.V. Thakare and Mr. K.C. Jagadeesan for
`carrying out
`irradiations of
`lutetium oxide target
`is
`gratefully acknowledged.
`
`References
`
`Banerjee, S., Das, T., Chakraborty, S., Samuel, G., Korde, A., Srivastava,
`S., Venkatesh, M., Pillai, M.R.A., 2004. 177Lu–DOTA–lanreotide: a
`novel tracer as a targeted agent for tumor therapy. Nucl. Med. Biol.
`31, 753–759.
`Boerman, O.C., Oyer, W.J.G., Corstens, F.H.M., 2000. Radio-labeled
`receptor-binding peptides: a new class of radiopharmaceuticals. Semin.
`Nucl. Med. 30, 195–208.
`Breeman, W.A.P., de Jong, M., Kwekkeboom, D.J., Valkema, R., Kooij,
`P.P., Visser, T.J., Krenning, E.P., 2001. Somatostatin receptor-
`mediated imaging and therapy: basic science, current knowledge,
`limitations and future perspectives. Eur. J. Nucl. Med. 28, 1421–1429.
`Breeman, W.A.P., de Jong, M., Visser, T.J., Erion, J.L., Krenning, E.P.,
`2003. Optimising conditions for radiolabelling of DOTA-peptides with
`90Y, 111In and 177Lu at high specific activities. Eur. J. Nucl. Med. Mol.
`Imag. 30, 917–920.
`Britton, K.E., 1997. Towards the goal of cancer specific imaging and
`therapy. Nucl. Med. Commun. 18, 992–1005.
`Das, T., Chakraborty, S., Unni, P.R., Banerjee, S., Samuel, G., Sarma,
`H.D., Venkatesh, M., Pillai, M.R.A., 2002. 177Lu labeled cyclic
`polyaminophosphonates as potential agents for bone pain palliation.
`Appl. Radiat. Isot. 57, 177–184.
`
`de
`
`Das, T., Chakraborty, S., Banerjee, S., Mukherjee, A., Samuel, G., Sarma,
`H.D., Nair, C.K.K., Kagiya, V.T., Venkatesh, M., 2004. Preparation
`and preliminary biological evaluation of a 177Lu labeled sanazole
`derivative for possible use in targeting tumor hypoxia. Bioorg. Med.
`Chem. 12, 6077–6084.
`Jong, M., Breeman, W.A.P., Valkema, R., Bernard, B.F.,
`Krenning, E.P., 2005. Combination radionuclide therapy using
`177Lu- and 90Y-labeled somatostatin analogues. J. Nucl. Med. 46,
`13S–17S.
`Eckelman, W.C., Gibson, R.E., 1993. The design of site-directed radio-
`pharmaceuticals for use in drug discovery. In: Burns, H.D., Gibson,
`R.E., Dannals, R., Siegl, P.
`(Eds.), Nuclear Imaging in Drug
`Discovery, Development, and Approval. Birkhauser, Boston.
`Firestone, R., 1996. In: Shirley, V.S. (Ed.), Table of Isotopes, eighth ed.
`Wiley, New York.
`Forrer, F., Uusijarvi, H., Storch, D., Maecke, H.R., Mueller-Brand, J.,
`2005. Treatment with 177Lu–DOTATOC of patients with relapse of
`neuroendocrine tumors after treatment with 90Y–DOTATOC. J. Nucl.
`Med. 46, 1310–1316.
`Knapp, F.F., Ambrose, K.R., Beets, A.L., Luo, H., McPherson, D.W.,
`Mirzadeh, S., 1995. Nuclear medicine program progress report for
`quarter ending September 30, 1995. Oak Ridge National Laboratory,
`pp. 6–16.
`Krenning, E.P., Bakker, W.H., Kooij, P.P., Breeman, W.A., Oei, H.Y., de
`Jong, M., Reubi, J.C., Visser, T.J., Bruns, C., Ikekubo, K., 1992.
`S

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket