throbber
PROTOCOL
`
`Conjugation of DOTA-like chelating agents to peptides
`and radiolabeling with trivalent metallic isotopes
`
`Jane K Sosabowski & Stephen J Mather
`
`Nuclear Medicine Research Laboratory, St. Bartholomew’s Hospital, London, EC1A 7BE, UK. Correspondence should be addressed to J.K.S.
`(jane.sosabowski@cancer.org.uk) or S.J.M. (stephen.mather@cancer.org.uk).
`
`Published online 3 August 2006; doi:10.1038/nprot.2006.175
`
`Peptides can be labeled with various trivalent radiometals for imaging or targeted radionuclide-therapy applications. The peptide
`is first conjugated to a chelating agent that is able to form stable complexes with the radionuclide of interest. This conjugation step
`can be carried out as part of the solid-phase peptide synthesis, or it can be undertaken in the solution phase after synthesis and
`purification of the peptide. The latter route, described here, involves reacting a molar excess of the activated tri-tert-butyl ester-
`derivatized chelator with a designated free amino group of a peptide analog, in which all other reactive amines are protected, in
`the presence of a coupling agent. The conjugate molecule is then purified prior to deprotection and further purification by HPLC.
`The product can be radiolabeled by addition of a suitable metal salt, followed, if necessary, by removal of the unchelated metal.
`The entire process of conjugation, purification and radiolabeling should take approximately 12.5 h.
`
`INTRODUCTION
`A number of trivalent metal radionuclides have physical properties
`suitable for radioisotope imaging (indium-111 (111In), gallium-
`67/68 (67/68Ga) and yttrium-86 (86Y)) or for targeted radionuclide
`therapy (90Y and lutetium-177 (177Lu)). These metal radionuclides
`can be combined with a targeting biomolecule (such as a peptide or
`antibody) in order to diagnose, monitor or treat disease. To obtain
`a radiolabeled biomolecule with the required stability, the peptide
`or protein must first be conjugated to a suitable chelator in order
`to complex the metal. The requirements of chelators for trivalent
`metals (such as In, Y, Ga and Lu) for labeling peptides are generally
`the same as those for labeling proteins1. The complexes should
`be stable in biological systems and their chelating ability should
`not be impaired by reaction with the peptide. Most often,
`diethylenetriaminepentaacetic acid (DTPA) and 1,4,7,10-tetra-
`azacyclododecanetetraacetic acid (DOTA) are used. Of the metals
`mentioned, the DOTA complexes are more thermodynamically and
`kinetically stable than the DTPA complexes. The drawback of using
`DOTA complexes is that they require a degree of heating to aid their
`formation, whereas the less-stable DTPA complexes can form at
`room temperature (19–25 1C). However, peptides (unlike proteins)
`are generally stable to heating; therefore, for peptide-labeling
`applications, DOTA is preferred. As DOTA is a tetra acid, it has
`four potential sites for conjugation, which can lead to a mixture
`of cross-linked conjugates. Fortunately, active-ester tri-t-butyl-
`protected DOTA compounds are commercially available along
`with other useful bifunctional chelators. For this protocol, we
`have described the use of 1,4,7,10-tetraazacyclododecane-1,4,7-tris
`(t-butyl acetate)-10-acetic acid (DOTA-(tBu)3; Fig. 1) a compound
`that requires activation of its free carboxyl group for the conjuga-
`tion to occur. The low aqueous solubility of this compound and the
`need for deprotection after conjugation make it unsuitable for
`chelation with large water-soluble antibodies or proteins, but it is
`ideal for reacting peptides in non-aqueous solvents.
`It is necessary to ensure that the conjugation of a chelator to the
`peptide does not substantially lower the peptide’s receptor-binding
`affinity. It is advantageous to use peptides in which the active
`sequence that confers binding affinity is known, so that the
`
`972 | VOL.1 NO.2 | 2006 | NATURE PROTOCOLS
`
`conjugation can be directed away from this receptor binding site.
`If necessary, spacers can be used to increase the distance between
`the conjugation site and the receptor binding site. Usually, the
`peptide is synthesized in such a way that the receptor binding site is
`at the carboxy terminus and the chelator is conjugated to the
`amino-terminal group. This can be done either during fluorenyl-
`methoxycarbonyl (Fmoc) solid-phase peptide synthesis2–5 or in the
`solution phase after synthesis and purification of the peptide6,7.
`In the latter scenario, all free amino groups (such as those on
`lysine residues) other than the desired conjugation site should be
`tert-butyloxycarbonyl (BOC) protected, in order to prevent any
`undesirable side reactions. Chelators can also be conjugated to the
`free amino group of a lysine residue within the peptide sequence. As
`Fmoc solid-phase peptide synthesis is amply covered elsewhere3, this
`
`a
`
`O
`
`O
`
`O
`
`O
`
`b
`
`COOH
`
`COOH
`
`N
`
`N
`
`N
`
`N
`
`N
`
`N
`
`N
`
`N
`
`O
`
`O
`
`O
`
`OH
`
`COOH
`
`O
`
`Peptide
`
`Figure 1 | Chemical structures. (a) DOTA-tri-t-butyl ester. (b) The conjugated
`DOTA-peptide.
`
`natureprotocols
`
`/moc.erutan.www//:ptth
`
`
`
` puorG gnihsilbuP erutaN 6002 ©
`
`Evergeen Ex. 1026
`1 of 5
`
`

`

`PROTOCOL
`
`temperature (37–100 1C) and reaction time (10–60 min). The
`reaction kinetics vary depending on the metal8,9, and the peptide
`residues adjacent to the chelator can also have an effect. Therefore,
`the reaction conditions should be optimized for each peptide/
`chelator/metal combination.
`The amounts of the radioactive metal added will depend on the
`required specific activity (MBq nmol–1) of the product. For
`receptor imaging and therapy studies, it is usually the case that a
`high specific activity is required to prevent both saturation of the
`receptor and possible pharmacological side effects. Usually the
`peptide conjugate is in excess in comparison with the radionuclide,
`and the labeling will be highly efficient with almost 100% of the
`metal being complexed. If higher specific activity is required, then
`rigorous elimination of metal impurities (e.g., by performing the
`radiolabeling reaction in polypropylene instead of glass, using
`polypropylene weighing spatulas, and treating vials, pipette tips
`and so on with acid10) might become necessary, as well as some
`adjustments to the reaction conditions. As far as the reagents are
`concerned, it is important to start with high-grade materials with
`the lowest levels of metal impurities (for this reason, ammonium
`acetate buffer is used as opposed to sodium acetate). In addition,
`the buffers and solutions can be pretreated with Chelex chelating
`ion-exchange resin11 to remove trace multivalent metals. However,
`care should be taken with this approach if high radiolabeling
`efficiencies are required. It has been reported that treatment of
`radiolabeling reaction solutions with Chelex resin, while potentially
`increasing the specific activity of the labeled product, can also
`reduce the labeling efficiency. This is probably due to the presence
`of chelating material residue in the solvents, which has leached
`from the resin. To obtain radiolabeled products with the highest
`possible specific activities, the labeling efficiency might therefore
`have to be compromised and a post-labeling purification step (such
`as a Sep-Pak purification) might need to be introduced.
`This protocol describes the method required to conjugate a
`peptide to the DOTA chelator, and to label the conjugate with a
`radionuclide (such as 111In or 90Y) for use in radionuclide imaging
`or targeted therapy. In total, the entire process should take
`about 12.5 h.
`
`protocol provides a method for small-scale solution-phase conjuga-
`tion of the DOTA-tri-t-butyl ester to a peptide amino group.
`The conjugation method for peptides is quicker than that for
`proteins1, as the solubility of peptides in non-aqueous solvents
`(such as N,N-dimethylformamide (DMF) or N-methyl-2-pyrroli-
`dinone (NMP)) allows them to be reacted and purified more
`rapidly. In this protocol, a DOTA-tri-t-butyl ester is pre-incubated
`in NMP with equimolar amounts of
`the coupling agents,
`O-(7-azabenzotriazole-1-yl)-1,1,3,3,-tetramethyluronium hexa-
`fluorophosphate (HATU) and diisopropylethylamine (DIPEA),
`the last of which is an organic base. The peptide is added to this
`mixture in a peptide:chelator ratio of 1:3 to 1:4, which is suitable for
`small-scale reactions (using 1–5 mg peptide), such as those carried
`out early in the radiopharmaceutical development process. For
`reactions using a large amount of peptide, it is possible to use ratios
`close to 1:1 (ref. 7). After stirring for 4 h at room temperature, the
`product is purified by solid-phase extraction (or extracted into
`ethylacetate (EtOAc)) and the solvent is evaporated off under
`vacuum. The crude product is dissolved in a 94:4:2 (vol/vol)
`mixture of trifluoroacetic acid (TFA):thioanisole:water to remove
`the BOC-protecting groups. After 4 h, the residue is washed with
`ether, redissoved in acetonitrile (ACN)/water and purified by
`reversed phase (RP)-HPLC. The HPLC fractions are evaporated
`to dryness (overall yield, 60%). The final peptide product can then
`be dissolved in water or aqueous buffer (depending on the peptide)
`for radiolabeling.
`In the subsequent radiolabeling step, the metal chloride salt
`(dissolved in dilute HCl) is added to a reaction vial, followed by
`acetate buffer at pH 4–6. This pH range is chosen to ensure that the
`metal remains in solution during the subsequent labeling proce-
`dure. At pH 46, many metals form insoluble hydroxide complexes
`with water and become unavailable for binding to the chelator. The
`presence of weakly chelating ions in the buffer (such as acetate or
`citrate) will also reduce the likelihood of hydrolysis. Some anti-
`oxidants (such as ascorbic or gentisic acid) might also be added to
`prevent radiolysis of the peptide. The peptide is added to this weak
`metal acetate chelate solution and the vial is heated (usually in
`a heating block). The general reaction variables are pH (4–6),
`
`natureprotocols
`
`/moc.erutan.www//:ptth
`
`
`
` puorG gnihsilbuP erutaN 6002 ©
`
`MATERIALS
`REAGENTS
`. Peptide (typically 1–5 mg depending on scale; e.g., piCHEM, Bachem,
`BioSynthema)
`. DOTA-(tBu)3 (Macrocyclics, cat. no. B260)
`. NMP (Sigma-Aldrich)
`. HATU (Sigma-Aldrich)
`. DIPEA (Sigma-Aldrich)
`. Metal-free water (typical resistance, 18 mO cm–1; from an ELGA water-
`purification system)
`. TFA (Spectrophotometric grade; Sigma-Aldrich)
`. Thioanisole (Sigma-Aldrich)
`. ACN (HPLC grade; Sigma-Aldrich)
`. EDTA (Sigma-Aldrich)
`. Ammonium acetate (choose the grade with the lowest possible metal
`impurities; Sigma-Aldrich)
`. Glacial acetic acid (Sigma-Aldrich) ! CAUTION Highly corrosive.
`. Ammonia solution (35%; Sigma-Aldrich) ! CAUTION Vapors are irritating to
`respiratory system; corrosive.
`. Methanol (HPLC grade; Sigma-Aldrich)
`. Chelex-100 analytical grade resin, 100–200 mesh (Bio-Rad Laboratories)
`. Radionuclide as chloride salt in dilute hydrochloric acid.
`
`. 111In chloride (e.g., GE Healthcare, Tyco Healthcare, MDS Nordion)
`. 90Y chloride (e.g., MDS Nordion, Perkin Elmer)
`. 177Lu chloride (e.g., Perkin Elmer, IDB) ! CAUTION Radioactive; use
`appropriate radiation safety measures as laid out in local rules; standard
`shielding and radionuclide handling procedures should be employed; direct
`exposure to the radioactive dose should be kept to a minimum; individuals
`working with the material should monitor their radiation exposure with
`appropriate devices.
`. HPLC Solvent A: 0.1% TFA (vol/vol) in water (Sigma-Aldrich)
`. HPLC Solvent B: 0.1% TFA (vol/vol) in ACN (Sigma-Aldrich)
`EQUIPMENT
`. Calibrated pH meter
`. 1 ml Reacti-Vial with Reacti-Vial magnetic stirrer (Pierce and Warriner,
`cat. nos. 13221 and 16010)
`. Magnetic stirrer plate
`. C-18-Sep-Pak-Classic cartridge (Waters)
`. Metal-free round-bottomed polypropylene tubes, 2 ml (e.g., Nunc
`Cryo-tubes, Corning Inc.)
`. Centrifugal evaporator
`. RP-HPLC gradient system with UV and radiochemical detectors
`(Raytest GmbH)
`
`NATURE PROTOCOLS | VOL.1 NO.2 | 2006 | 973
`
`Evergeen Ex. 1026
`2 of 5
`
`

`

`PROTOCOL
`
`. Jupiter C18 HPLC column, 5 mm, 300 A˚ , 250  4.6 mm internal diameter
`(i.d.) or similar (Phenomenex, cat. no. 00G-4053-E0)
`. Chromatographic support material, silica-gel-impregnated glass-fiber
`(ITLC-SG, Pall Corp.) cut into strips approximately 2  10 cm in size with
`a faint pencil mark 1.5 cm from one end
`. Gamma counter (e.g., Perkin Elmer)
`REAGENT SETUP
`0.1 M ammonium acetate buffer (pH 6) Dissolve 7.708 g ammonium acetate in
`1 l water, add 300 ml acetic acid. Mix well and check that the pH is 5.5–6.
`! CAUTION Acetic acid is highly corrosive.
`0.1 M ammonium acetate containing 50 mM EDTA Dissolve 185 mg EDTA in
`10 ml of 0.1 M ammonium acetate buffer (pH 6).
`1 M ammonium acetate buffer (pH 5.5–6) Prepare a 1-M solution of
`ammonium acetate by dissolving 3.854 g in 50 ml water. Prepare a 1-M solution of
`acetic acid by making 3 ml acetic acid up to 50 ml. Add one part 1 M acetic acid
`to eight parts 1 M ammonium acetate. Mix well and check that the pH is 5.5–6.
`If an extremely high specific activity radiolabeled product is required, this buffer can
`be treated with Chelex-100 resin prior to use (see INTRODUCTION for a
`discussion of the merits of this approach). ! CAUTION Acetic acid is highly corrosive.
`1:1 mixture of 3.5% (vol/vol) ammonia solution and methanol To prepare
`100 ml, using an appropriate measuring cylinder, measure 45 ml water and
`
`make up to 50 ml using 35% (vol/vol) ammonia solution. Add this to 50 ml
`methanol and mix. m CRITICAL This solution must be prepared fresh on
`the day of analysis to avoid evaporation of the solvents.
`EQUIPMENT SETUP
`Preparation of metal-free vials and disposables For preparation of an
`extremely high specific activity radiolabeled peptide, place vials, tubes, pipette
`tips and so on in a 500-ml bottle containing 0.1 M HCl (prepared with
`metal-free water), and place on a mixer overnight. Pour off the HCl and rinse
`thoroughly with metal-free water. Refill the bottle with metal-free water and
`place on a mixer for a further 2 h. Remove the vials and disposables from the
`water, and dry in an oven.
`HPLC method for peptide purification Set up a general HPLC method
`for peptide separation on a C18 column, as in the following example:
`0–20 min, 5–60% Solvent B; 20–25 min, hold at 60% Solvent B; 25–30 min,
`60–90% Solvent B; 30–35 min, 90–5% Solvent B. It might be necessary to
`optimize the separation of the conjugated peptide from any unconjugated
`peptide or chelator. Inject the starting materials to determine their elution
`times. Inject small amounts of the crude product and decrease the
`solvent gradient if necessary to improve the peak resolution. Try not to
`decrease the gradient to o1% ACN per min, as this might cause peak
`broadening.
`
`PROCEDURE
`Conjugation reaction
`1|
`Calculate the amount of DOTA-(tBu)3 to give a threefold to fourfold molar excess over the amount of peptide to be used
`(1–5 mg).
`
`2| Add equimolar amounts of HATU and DIPEA along with NMP (approximately 30 ml NMP per mg peptide to be used) to the
`DOTA-(tBu)3 chelator, and incubate at room temperature for 20 min.
`
`3|
`
`Transfer the mixture to a 1-ml Reacti-Vial.
`
`4| Dissolve the peptide in NMP (30 ml mg–1 peptide) and add the same amount of DIPEA as was added to the chelator.
`
`5|
`
`Transfer this solution to the 1-ml Reacti-Vial, add a Reacti-Vial magnetic stirrer and stir for 4 h at room temperature.
`
`Sep-Pak purification
`6| Activate a C-18-Sep-Pak cartridge with methanol and then pre-equilibrate the column with water.
`
`7|
`
`8|
`
`Load the reaction mixture onto the column and wash with 5 ml water.
`
`Elute the peptide conjugate into a 1.5-ml microcentrifuge tube with 1 ml of 0.1% TFA (vol/vol) in ACN.
`
`9|
`Evaporate the solvent using a centrifugal evaporator (this should take approximately 15 min at room temperature).
`’ PAUSE POINT The protected peptide-conjugate crude material can be stored in a refrigerator overnight at 4 1C.
`
`Deprotection
`10| Dissolve the crude product in 282 ml TFA, 12 ml thioanisole and 6 ml water, and react for 4 h.
`
`11| Evaporate off the deprotection mixture using a centrifugal evaporator (this should take approximately 15–20 min at room
`temperature).
`12| Wash the residue with ice-cold ether (3  1 ml) and re-dissolve in 20% ACN (vol/vol) in water.
`
`HPLC purification
`13| Purify the peptide conjugate by RP-HPLC using a Jupiter C18 column (or similar) equilibrated on 5% Solvent B.
`m CRITICAL STEP The first time this step is carried out for any peptide, it might be necessary to optimize the separation method,
`before injection of the entire crude mixture. Refer to the advice given in the EQUIPMENT SETUP. Be aware that any residual
`unconjugated DOTA-(tBu)3 might have also come through the SPE column, so there might be some deprotected DOTA in this HPLC
`purification mixture. If there is more than one unprotected primary amine group on the peptide, this will lead to a mixture of
`products, which will again complicate the purification procedure. In this case, the products should be separated by HPLC, collected
`and structural analysis carried out in order to identify the required product.
`
`14| Inject the crude product onto the HPLC and run the separation method.
`
`974 | VOL.1 NO.2 | 2006 | NATURE PROTOCOLS
`
`natureprotocols
`
`/moc.erutan.www//:ptth
`
`
`
` puorG gnihsilbuP erutaN 6002 ©
`
`Evergeen Ex. 1026
`3 of 5
`
`

`

`PROTOCOL
`
`15| Collect the HPLC eluate containing the peptide conjugate and evaporate to dryness in a weighed microcentrifuge tube
`using a centrifugal evaporator. Re-weigh the microcentrifuge tube to determine the yield of conjugate.
`
`16| Redissolve the product in water (with added base or acid to achieve solubility if necessary) to a concentration of
`1 mg ml–1, divide into suitable sized aliquots (10–50 mg depending on application) in metal-free tubes and store at below
`–20 1C until required.
`’ PAUSE POINT The peptide conjugate can be stored for months or years under these conditions.
`
`Radiolabeling
`! CAUTION Use appropriate radiation safety measures at this stage.
`17| Thaw out an aliquot of the peptide conjugate.
`
`18| Pipette the required volume of radionuclide solution into a metal-free polypropylene screw-top tube (such as a 2-ml
`Corning cryotube). The quantity will depend on the amount of radioactivity needed for the desired application.
`
`19| Add a volume of 1 M ammonium acetate buffer (pH 5.5–6) equal to 10% of the volume of the radioactive isotope.
`
`20| Add a volume of peptide conjugate corresponding to the required specific activity (MBq nmol–1) of the radiolabeled
`product. Check the pH of the labeling reaction using pH indicator strips (it should be in the range of 4 to 6).
`m CRITICAL STEP If the pH is o4, it is probable that 1 M ammonium acetate buffer has been omitted from the reaction mixture or
`insufficient buffer has been added. This will reduce the rate of metal complexation and more buffer should be added to compensate
`for this. If the pH is 46, the pH of the ammonium acetate buffer is probably incorrect. This could lead to the formation of insoluble
`metal hydroxides.
`
`21| Mix well and heat the labeling reaction in a dry-block heater for up to 30 min at up to 98 1C. The exact conditions depend
`on the radioisotope (e.g., 111In requires more heating than 90Y) and the peptide (the temperature or duration of heating might
`have to be minimized if there are any thermolabile residues, e.g., if it is necessary to prevent the oxidation of methionine).
`
`22| Remove the vial from the heating block and allow to cool for a few minutes. Quench the reaction by adding 10% of the
`total reaction volume of 0.1 M ammonium acetate/50 mM EDTA solution.
`
`Measurement of radiolabeling efficiency by thin-layer chromatography (TLC)
`23| Pour 0.1 M ammonium acetate containing 50 mM EDTA solution into a glass beaker (10–15 cm tall) or similar container
`until it is 0.5 cm deep. Cover the beaker with a Petri dish lid, aluminum foil or similar.
`
`24| Repeat this procedure using a 1:1 ratio of 3.5% (vol/vol) ammonia/methanol solution in a separate container.
`
`25| Place a 1-ml spot of the radiolabeled peptide sample to be analyzed onto the centre of the pencil mark on each of two
`chromatographic strips and allow the spots to dry.
`
`26| Using forceps, gently place the strips upright in each beaker with the pencil mark at the lower end just above the solvent
`level. Cover the beaker and allow the solvent to run up the support material.
`
`27| When the solvent is about 5 mm from the top of the strip, remove it from the beaker using forceps and lay it on a clean
`piece of tissue to dry.
`
`28| Cut the strips in two equal parts across the short axis. Place the upper and lower halves into separate gamma counter tubes
`and measure the radioactive counts from each tube in a gamma counter.
`
`29| In the case of the ammonium acetate/EDTA strip, unbound radionuclide migrates with a retardation factor (Rf) of 1, while
`labeled peptide remains at the origin. Calculate the proportion of unbound radionuclide as follows:
`100%
`% unbound radionuclideðAÞ ¼
`
`Counts on upper half
`Counts on lower plus upper half
`
`30| In the case of the 1:1 ratio of 3.5% (vol/vol) ammonia/methanol strip, calculate the amount of insoluble metal hydroxides
`(colloidal material) present in the radiolabeling mixture as follows:
`
`natureprotocols
`
`/moc.erutan.www//:ptth
`
`
`
` puorG gnihsilbuP erutaN 6002 ©
`
`% insoluble hydroxidesðBÞ ¼
`
`Counts on lower half
`Counts on lower plus upper half
`
`100%
`
`The peptide labeling efficiency is therefore 100 – (A + B)%.
`
`NATURE PROTOCOLS | VOL.1 NO.2 | 2006 | 975
`
`Evergeen Ex. 1026
`4 of 5
`
`

`

`PROTOCOL
`
`Measurement of radiolabeling efficiency by HPLC
`31| Dilute radiolabeling reaction with water or saline. Inject on RP-HPLC using the same column and conditions
`as those used for the purification of the peptide conjugate.
`
` TIMING
`
`Steps 1–5: peptide conjugation, 5 h.
`
`Steps 9–15: deprotection and purification, 6.5 h.
`
`Steps 17–31: Radiolabeling and analysis, 1 h.
`
`? TROUBLESHOOTING
`Troubleshooting advice can be found in Table 1.
`
`TABLE 1 | Troubleshooting table.
`
`PROBLEM
`Poor peptide
`conjugate yields.
`
`POSSIBLE CAUSES
`If an EtOAc extraction step is used, the product can be lost. Use NMP as a reaction solvent instead of DMF, and use
`SPE purification instead of EtOAc extraction.
`
`Loss of peptide during HPLC purification. This can be due to peptide binding to free silanols on the C18 column.
`Use a column that tolerates low pH, such as Phenomenex Jupiter C18.
`
`Low labeling
`efficiency.
`
`Reaction mixture too dilute (i.e., peptide concentration too low).
`
`Reaction conditions not optimal for particular peptide-chelator-metal combination. Vary reaction time, temperature,
`volume and pH (do not exceed 6.5 due to formation of insoluble metal hydroxides).
`
`Contamination with metal ions. Make sure that all reagents and disposables are metal free. Use a polypropylene
`reaction vial for the radiolabeling reaction.
`
`Contamination of buffers with chelating material, such as Chelex. If high specific activities cannot be achieved
`without the use of Chelex, introduce a post-labeling Sep-Pak purification to remove the radiometal–Chelex complexes.
`
`High specific activity
`not achievable.
`
`Contamination with metal ions. Make sure that all reagents and disposables are metal free. Use a polypropylene
`reaction vial for the radiolabeling reaction.
`
`ANTICIPATED RESULTS
`Yields of conjugated peptide after deprotection and purification should be in the region of 60%. Radiolabeling efficiencies
`495% should be routinely achieved.
`
`COMPETING INTERESTS STATEMENT The authors declare that they have no
`competing financial interests.
`
`Published online at http://www.natureprotocols.com
`Rights and permissions information is available online at http://npg.nature.com/
`reprintsandpermissions
`
`1. Cooper, M.S., Sabbah, E. & Mather, S.J. Conjugation of chelating agents to
`proteins and radiolabelling with trivalent metallic isotopes. Nat. Protocols,
`published online 27 June 2006 (doi:10.1038/nprot.2006.49).
`2. Achilefu, S. et al. Novel bioactive and stable neurotensin peptide analogues
`capable of delivering radiopharmaceuticals and molecular beacons to tumors.
`J. Med. Chem. 46, 3403–3411 (2003).
`3. Chan, W.C. & White, P.D. (eds.) Fmoc Solid Phase Peptide Synthesis: A Practical
`Approach (Oxford University Press, Oxford, UK, 2000).
`4. De Leon-Rodriguez, L.M., Kovacs, Z., Dieckmann, G.R. & Sherry, A.D.
`Solid-phase synthesis of DOTA-peptides. Chemistry 10, 1149–1155
`(2004).
`
`5. Reubi, J.C. et al. Unsulfated DTPA- and DOTA-CCK analogs as specific high-affinity
`ligands for CCK-B receptor-expressing human and rat tissues in vitro and in vivo.
`Eur. J. Nucl. Med. 25, 481–490 (1998).
`6. Edreira, M., Melendez-Alafort, L. & Mather, S.J. Optimization of the small-scale
`synthesis of DOTA-Tyr3-octreotide. Nucl. Med. Commun. 23, 493–499 (2002).
`7. Heppeler, A. et al. Radiometal-labelled macrocyclic chelator-derivatised
`somatostatin analogue with superb tumour-targeting properties and potential for
`receptor-mediated internal radiotherapy. Chem. Eur. J. 5, 1974–1981 (1999).
`8. Breeman, W.A. et al. Radiolabelling DOTA-peptides with 68Ga. Eur. J. Nucl. Med.
`Mol. Imaging 32, 478–485 (2005).
`9. Breeman, W.A., De Jong, M., Visser, T.J., Erion, J.L. & Krenning, E.P. Optimising
`conditions for radiolabelling of DOTA-peptides with 90Y, 111In and 177Lu at high
`specific activities. Eur. J. Nucl. Med. Mol. Imaging 30, 917–920 (2003).
`10. Hainsworth, J.E. & Mather, S.J. Regressive DOTA labelling performance with
`indium-111 and yttrium-90 over a week of use. Eur. J. Nucl. Med. Mol. Imaging 32,
`1348 (2005).
`11. Bio-Rad Laboratories. Chelex 100 and Chelex 20 Chelating Ion Exchange Resin
`Instruction Manual (Bio-Rad Laboratories, Hercules, CA, 2000).
`
`natureprotocols
`
`/moc.erutan.www//:ptth
`
`
`
` puorG gnihsilbuP erutaN 6002 ©
`
`976 | VOL.1 NO.2 | 2006 | NATURE PROTOCOLS
`
`Evergeen Ex. 1026
`5 of 5
`
`

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