WO2012012612A2 - Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes - Google Patents

Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes Download PDF

Info

Publication number
WO2012012612A2
WO2012012612A2 PCT/US2011/044814 US2011044814W WO2012012612A2 WO 2012012612 A2 WO2012012612 A2 WO 2012012612A2 US 2011044814 W US2011044814 W US 2011044814W WO 2012012612 A2 WO2012012612 A2 WO 2012012612A2
Authority
WO
WIPO (PCT)
Prior art keywords
diels
tetrazine
alder adduct
residue
mmol
Prior art date
Application number
PCT/US2011/044814
Other languages
French (fr)
Other versions
WO2012012612A3 (en
Inventor
Joseph M. Fox
Matthew Hassink
Melissa Blackman
Zibo Li
Peter S. Conti
Original Assignee
University Of Delaware
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Delaware filed Critical University Of Delaware
Priority to EP11810394.4A priority Critical patent/EP2595967B1/en
Priority to US13/811,568 priority patent/US10434197B2/en
Priority to CN201180041210.XA priority patent/CN103221398B/en
Publication of WO2012012612A2 publication Critical patent/WO2012012612A2/en
Publication of WO2012012612A3 publication Critical patent/WO2012012612A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/08Six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • Positron emission tomography is a non-invasive imaging modality that utilizes positron-emitting radionuclides (C-ll, N-13, 0-15 and F-18).
  • F-18 PET has a number of attributes that make it clinically attractive, including 100% positron efficiency, a very high specific radioactivity, and a short half-life of ⁇ 110 min.
  • the short half-life of F-18 and the poor nucleophilicity of fluoride render it difficult to incorporate F-18 in complex molecules.
  • radiochemistry is a major limiting factor for the field of PET.
  • challenges exist for improving F-18 incorporation with respect to reaction rates, efficiency, and selectivity.
  • tetrazine-trans-cyclooctene ligation OTTCO ligation' as a method of bioconjugation that proceeds with fast reaction rates without need for catalysis
  • tra/is-Cyclooctene derivatives are readily prepared from c/ ' s- cyclooctenes using a photochemical flow-reaction that the inventors developed (M.
  • 3,6-diaryl-s-tetrazines offer an excellent combination of fast reactivity and stability for both the conjugate and starting material.
  • 3,6 - di(2-pyridyl)-s-tetrazines have been shown to display excellent characteristics.
  • the reaction between frans-cyclooctene and la proceeds with a rapid rate (k 2 ⁇ 2000 M 1 s "1 in 9: 1 MeOH :water), and is successful in cell media and cell lysate.
  • 3,6 -Di(2-pyridyl)- s-tetrazines can easily be functionalized as their amido derivatives ( lb), which display excellent st
  • the TTCO-ligation offers opportunities for the rapid conjugation of radionuclides to biomolecules, both of which are often available only at low concentration.
  • the invention provides a Diels-Alder adduct of a trans-cyclooctene with a tetrazine, wherein the adduct bears a substituent labeled with a radionuclide.
  • the invention provides a method of producing a PET or other image of an organ in an animal or human.
  • the method includes forming in the animal or human a Diels-Alder adduct of a trans-cyclooctene with a tetrazine, wherein the adduct bears a substituent labeled with a radionuclide.
  • the invention provides the compound according to structure 5
  • the invention provides the compound according to structure 9
  • the invention provides the compound according to structure 17
  • the invention provides the compound according to structure 13
  • the invention provides the compound according to structure 24 vides the compound according to structure 25
  • the invention provides the compound according to structure 26 provides the compound according to structure 27 BRIEF DESCRIPTION OF THE DRAWINGS
  • Figure 1 shows cell binding affinity studies of c(RGDyK) ( 12) and 19 F-15.
  • Figure 2 is a schematic representation of an automated synthesis module for performing fluorination ( ⁇ 200 mCi).
  • Figure 3A shows tumor and major organ uptake of 18 F-41.
  • the invention provides a number of ways to use TTCO-ligation for labeling, as will now be described. Labeling with 18 F will be described first, followed by a description of labeling with other radionuclides.
  • the inventors focused on the development of direct methods for 18 F-incorporation via reactions with fluoride ion, with an initial focus on the synthesis of 18 F-labeled tetrazines (Scheme 1). Attempts to convert nitrotetrazine derivatives 2a and 2b into 18 F-labeled substitution products 3 using 18 F-fluoride/kryptofix or 18 F-TBAF gave decomposition products and only traces of radiolabeled products. The inventors also combined mesylate 4 with fluoride, and in the most successful experiment ( 18 F-TBAF at 85°C for 15 min) 18 F-labeled product 5 was obtained in ⁇ 1% labeling yield.
  • Nosylate 8 reacted efficiently with TBAF to provide 19 F-9 in high yield.
  • the notation 19 F- as part of a structure name indicates that the compound has not been radiolabeled, and 18 F- indicates labeling .
  • the invention provides analogs of 18 F-9 in which the spacer group between 0 and 18 F is an alkylene group.
  • the alkylene group may be a C2 alkylene group (as in 18 F-9), or it may be any alkylene group from C3 to C20, more typically from C3 to CIO, and most typically from C3 to C5.
  • an analog of 18 F-9 uses an 18 F-CH 2 -phenyl moiety in place of the 18 F-CH 2 - CH 2 moiety of 18 F-9.
  • Such a compound may be prepared from the corresponding bromide, chloride, iodide, sulfonate, etc. by nucleophilic displacement with 18 F fluoride.
  • the inventors also used a photochemical flow reactor (M. Royzen, G. Yap, and J. Fox, J. Am. Chem. Soc. 2008, 130 ( 12), 3760) to synthesize 16, an analog of 7 with a functionalizable group.
  • reaction between la and 16 was even faster, in fact too fast to measure by UV-vis spectroscopy.
  • 3,6-di(2-pyridyl)-s-tetrazine e.g ., lb
  • known methods for example as described by M . L. Blackman, M. Royzen and J. M. Fox, J. Am. Chem. Soc. 2008, 130, 13518-13519 and R. Rossin, P. R. Verkerk, S. M. v. d . Bosch, R. C. M. Vulders, I. Verel, J . Lub and M . S. Robillard, Angew. Chem. Int. Ed.
  • the Diels-Alder conjugate 10 was found to be stable in water, and the benign isomerization to 11 was the only side reaction. Thus, 19 F-10 was the only product detected by : H NMR analysis immediately after the conjugation. In CD 3 CN/H 2 0, the rearrangement of 19 F-10 to 19 F-11 proceeded to 11% conversion after 4 hours, and >95% conversion after 48 hours. The stability of the radiolabeled conjugation product was monitored in PBS buffer and serum media for 4 hours, and no degradation products of 18 F-10 were observed.
  • the inventors report the construction of a 18 F labeled PET probe for imaging (such as cancer imaging) based on the tetrazine-trans-cyclooctene ligation.
  • the reaction speed for labeling tetrazine-RGD peptide 14 with 18 F-9 was then investigated .
  • 18 F-9 was synthesized from nosylate 8.
  • the conjugation of 18 F-9 and tetrazine-RGD was performed at room temperature for 5 min.
  • the conjugation was efficient and high yielding : starting with only 30 pg (78 ⁇ ) of tetrazine-RGD conjugate 14 and 2 mCi (5 ⁇ ) of 18 F-rrans-cyclooctene, the labeling yield was 90% after 5 min by HPLC analysis.
  • the stability of 18 F-tetrazine-RGD conjugate 15 was evaluated in PBS in the presence 5% EtOH. The stability was excellent: >95% of the tracer remained after incubation for 6 h as judged by radio-HPLC analysis.
  • Static microPET scans were performed for 18 F-tetrazine-RGD conjugate 15, via injection into athymic female nude mice bearing an U87MG tumor. High tumor accumulation was observed as early as 30 min time point. The tumor uptake was 4.6 ⁇ 0.2, 4.4 ⁇ 0.6, 4.2 ⁇ 0.6, and 2.7 ⁇ 0.5 %ID/g at 0.5, 1, 2, 4 h post injection, respectively. 18 F-15 was cleared through both liver and kidney. Up to 4 h post injection, there was still a fair amount of activity accumulated in the abdomen, which may be a result of the relative hydrophobicity of this system.
  • Blocking experiments were also performed by co-injecting 10 mg/kg of c(RGDyK) along with 18 F-15.
  • the tracer uptake in U87MG tumor dropped to 1.4 ⁇ 0.2, 1.0 ⁇ 0.3, 0.6 ⁇ 0.2, and 0.4 ⁇ 0.1 %ID/g at 0.5, 1, 2, 4 h post injection respectively.
  • the successful blocking confirmed the integrin ⁇ ⁇ ⁇ 3 specific binding of the tracer 18 F-15.
  • conjugation of 18 with 9 to give conjugate 19 is extremely rapid and extremely efficient, and conjugate 19 oxidized spontaneously in solution to form the aromatized compound 20.
  • the inventors have synthesized the diaminotetrazine 21 and derivative 22, which can be expected to show rapid conjugation with trans-cyclooctenes such as 9 while affording robust in vivo stability.
  • the invention provides efficient methods for conjugating tetrazine and irans-cyclooctenes to proteins.
  • the amine-reactive NHS- ester 13 can be readily converted into the thiol-reactive maleimide derivative 24.
  • the reactive functionalities of these molecules may be used to conjugate tetrazines to lysine and cysteine residues of proteins, for example a VEGF protein.
  • NHS-ester 13 can be prepared from 5-oxo-5-(6-(6-(pyridin-2-yl)-l,2,4,5-tetrazin-3-yl)pyridin-3- ylamino)pentanoic acid, in turn prepared by the method of R. Rossin, P. R.
  • the inventors have conjugated maleimides 26 and 27 to a protein (thioredoxin) using standard methods known in the art, and the conjugates have been shown to undergo rapid tetrazine ligation in high yield .
  • Other proteins may be similarly conjugated according to the invention.
  • Compound 33 the conjugate of 27 with thioredoxin, was rapidly adducted with 3,6-di(2-pyridyl)-s-tetrazine ( la) as shown below, where Trx represents a thioredoxin residue.
  • VEGF proteins may be labeled with 18 F via modification of the lysines of wild -type VEGF 12 i protein with amine reactive derivatives (e.g., 24 or 25).
  • the resulting conjugation products (29 and 30, respectively) may be combined with irsns-cyclooctene 18 F-17 and tetrazine 18 F-5 to provide radiolabeled conjugates 18 F-31 and 18 F-32, respectively.
  • Labeling of biomolecules with radionuclides using tetrazine -frans-cyclooctene ligation is not limited to the specific frans-cyclooctenes and tetrazines discussed above; any radiolabeled tetrazine or rrans-cyclooctene may be used.
  • the invention encompasses labeling of biomolecules with radionuclides other than F. Suitable further examples of compounds according to the invention, all of which may be used to label
  • I labeling may for example be suitable for producing PET probes, while 125 I and 131 I may be suitable for preparing SPECT probes.
  • IR liquid, CHCI 3 , cm “1 ) 3456, 3011, 2975, 2936, 2861, 1650, 1447, 1392, 1252, 1100, 1049, 988, 875.
  • the silica gel was filtered and the filtrate was placed into a separatory funnel.
  • the organic layer was separated, and the ammonium hydroxide layer was extracted three times with methylene chloride.
  • the organic layers were combined and twice washed with water.
  • the organic layers were dried with MgS0 4 , filtered, and purified by column chromatography with 5% ether to 30% ether in hexanes.
  • IR liquid, CHCI 3 , cm “1 ) 3449, 3012, 2935, 2861, 1647, 1445, 1353, 1198, 1096, 1050, 993, 968, 797.
  • IR liquid, CHCI 3 , cm “1 ) 3105, 3010, 2935, 1609, 1536, 1351, 1187, 1097, 932, 857, 776, 616.
  • the purification of the crude product was carried out on a analytical reversed - phase high performance liquid chromatography (HPLC) system equipped with a dual UV absorbance detector (Waters 2487) using a Phenomenex C18 RP ( 150 x 4.6 mm 5 micron).
  • the flow was 1 mL/min, with the mobile phase starting from 95% solvent A (0.1% TFA in water) and 5% solvent B (0.1% TFA in acetonitrile) (0-2 min), followed by a gradient mobile phase to 5% solvent A and 95% solvent B at 17 min, which was then kept at 95% B until 22 min.
  • the radioactivity was detected by a model of Ludlum 2200 single-channel radiation detector.
  • a semi-preparative C18 reverse, phase column (Phenomenex C18) was used in separations with a 4 mL/min flow rate under gradient conditions.
  • the 18 0 is in the form of water with an isotopic purity of greater than 95%.
  • the operation of the cyclotron and target functions was automatically controlled by the cyclotron computer system.
  • the target was loaded with the required amount of [ 18 0]-water, and bombarded for the appropriate time with a suitable beam current.
  • the target was then unloaded to a collection vial located in the dose calibrator; where the amount of fluoride was measured.
  • the fluoride solution was then transferred to the chemistry operation.
  • the fluorination ( ⁇ 200 mCi) was performed on an automated synthesis module as shown in Figure 2.
  • two-way valves V1-V6 were used to control the solvent and reagent containing reservoirs 1-6.
  • Reservoirs 3-6 are connected with a nitrogen or argon gas line.
  • Reservoir 1 is connected with reactor through several control valves.
  • the reactor is connected with vacuum pump, gas line, and the injection port of the HPLC system.
  • the solutions of potassium carbonate and Kryptofix K2.2.2 (or TBAB and MeCN) were loaded into Reservoirs 1 and 2, respectively.
  • Reservoirs 3, 4, 5, and 6 were filled with precursor solution and other chemicals/solutions as needed.
  • Reservoir 2 was added into the reactor and the whole mixture was dried at 95°C in combination with nitrogen flow and vacuum .
  • the precursor solution from Reservoir 3 was added to the dried 18 F ion and heated at the desired temperature.
  • the reaction mixture will be sampled out for analysis or loaded on HPLC for purification.
  • Tetrazine 4 was prepared by mesylation of (MsCI, Et 3 N, CH 2 CI 2 ) of 3-(4-hydroxymethyl)-phenyl-s-tetrazine, which had been prepared from 4-hydroxymethylbenzonitrile (formamidine acetate, S 8 , hydrazine hydrate, then NaN0 2 /HOAc), according to the method of S. A. Lang Jr., B. D. Johnson, E. J. Cohen, J. Heterocycl. Chem. 1975, 12, 1143. Compound 4 was dissolved in MeCN and then allowed to react with 18 F-TBAF at 85°C for 15 min. The reaction mixture was then analyzed by HPLC.
  • Fluoride was dried as described above.
  • Precursor 8 was dissolved in MeCN and added to the azeotropically dried fluoride from Reservoir 3.
  • the crude mixture was heated at desired temperature and then analyzed by HPLC.
  • the optimized labeling conditions are described in entry 3 in table 2.
  • the crude reaction mixture was loaded onto the semi-prep HPLC for separation.
  • the purified sample was injected to the analytical HPLC.
  • 18 F-9 was eluted off at 17.4 min on HPLC, which correlates with the retention time of the standard compound . Under the optimized conditions, the radiochemical purity of 18 F-9 was more than 98%.
  • HPLC-purified 18 F-9 was mixed with 3,6-Di(2-pyridyl)-s-tetrazine ( la) under the conditions described in Table 2. Immediately after mixing, the crude reaction mixture was analyzed by HPLC. The HPLC injection was made within 10 seconds of mixing. 18 F- 10 was eluted off at 11.8 min on HPLC, which correlates with the retention time of the standard compound . Small amounts of isomers 18 F-11 were also observed on the HPLC radio trace. The retention time of peaks attributable to the isomers of 18 F-11 were 12.7 and 13.3 min, which correlates with the retention times for the 19 F standards.
  • [ 18 F] Fluoride was prepared by the 18 0(p,n) 18 F nuclear reaction, and it was then adsorbed onto an anion exchange resin cartridge.
  • Kryptofix 222/K 2 C0 3 solution 1 mL 9 : 1 acetonitrile/water, 15 mg Kryptofix 222, 3 mg K 2 C0 3 ) was used to elute the cartridge, and the resulting mixture was dried in a glass reactor.
  • 18 F-9 was prepared according to reported procedures and purified via semipreparative HPLC. Labeling of tetrazine-RGD conjugate 14 with 18 F-9 was performed in DMSO/EtOH (1 : 3). The resulting mixture was diluted with water and purified by semi -preparative HPLC. The final product 18 F-15 was concentrated and formulated in saline (0.9%, 500 ⁇ ) for in vivo studies.
  • U87MG human glioblastoma cells were grown in Dulbecco's medium (Gibco) supplemented with 10% fetal bovine serum (FBS), 100 IU/mL penicillin, and 100 pg/mL streptomycin (Invitrogen Co.). Animal procedures were performed according to a protocol approved by the Stanford University Institutional Animal Care and Use
  • ROIs regions of interest
  • the radioactivity concentration (accumulation) within a tumor was obtained from the mean value within the multiple ROIs and then converted to %ID/g.
  • mice bearing U87MG tumors on the front left flank were scanned after co-injection with 18 F-15 and c(RGDyK) (10 mg/kg).
  • the vial was swept with n itrogen, and anhydrous DMF ( 1.5 mL) was added via syringe.
  • the reaction mixture was allowed to stir for 20 h at room temperature.
  • the mixture was then diluted by CH 2 CI 2 (5 mL), centrifuged, and the supernatant was decanted.
  • the purple solid was subjected to three further cycles of suspension in CH 2 CI 2 (5 mL), centrifugation, and decantation to provide the 130 mg (59%) of the title compound as a purple solid ( 130 mg, 59%) .
  • a dry 3 mL vial was charged with 13 (5.2 mg, 0.011 mmol) and 12 (3.4 mg, 0.0055 mmol).
  • the vial was capped by a screw cap with a Teflon septum, and the vial was swept with nitrogen.
  • a solution of diisopropylethylamine ( 1.4 mg, 0.011 mmol) in DMF (30 pL) was added via syringe, followed by the addition of DMF (0.57 mL).
  • the reaction mixture was allowed to stir for 18 h at room temperature.
  • the mixture was then diluted by methanol (1 mL), centrifuged, and the supernatant was decanted .
  • the purple solid was subjected to two further cycles of suspension in methanol ( 1 mL), centrifugation, and decantation to provide 14 as a purple solid (5.1 mg, 96%) .
  • the purity was judged to be 95% by HPLC analysis (Shimadzu C18 column, 4.6 mm x 50 mm, 5 micron). HPLC analysis was performed with eluents that contained 0.1% trifluoroacetic acid, eluting at 1 mL/min, initially with 10% acetonitrile/water for 5 min, followed by a gradient of 10%-20% acetonitrile/water for 5 min, and final elution with 20% acetonitrile/water.
  • HPLC analysis was performed with eluents that contained 0.1% trifluoroacetic acid, eluting at 1 mL/min, initially with 5% acetonitrile/water for 2 min, followed by a gradient of 5%-95% acetonitrile/water for 22 min, and final elution with 95%
  • Tetrazine-R conjugate 18 was prepared according to the following sequence.
  • a dry round-bottom flask was charged with 37 (0.804 g, 2.00 mmol), CH 3 CN (15 mL) and 64% hydrazine hydrate (0.097 mL, 2.00 mmol).
  • the flask was fitted with a reflux condenser, and the mixture was heated to 50 °C for 1 h behind a blast shield.
  • K 2 C0 3 (0.553 g, 4.00 mmol) was added, and the reaction mixture was allowed to reflux and stir for 24 h.
  • 64% hydrazine hydrate (0.291 mL, 6.00 mmol) was added to the mixture, which was allowed to reflux for an additional hour.
  • the reaction mixture was cooled to room temperature.
  • the resulting orange precipitate was isolated by filtration, washed with cold H 2 0, and dried under vacuum.
  • the crude residue was diluted with glacial acetic acid (4.0 mL) at 0°C, and a solution of NaN0 2 (0.690 g, 10.0 mmol) in H 2 0 (1.1 mL) was added dropwise to the solution.
  • the reaction mixture was diluted with CH 2 CI 2 (100 mL) and washed repeatedly with saturated NaHC0 3 .
  • the organic solution was dried over MgS0 4 , filtered, and concentrated in vacuo.
  • a resealable glass vial was flushed with N 2 and charged with 36 (0.010 g, 0.0320 mmol), glutaric anhydride (0.018 g, 0.158 mmol) and anhydrous THF (0.3 mL). The vial was capped, and the mixture was stirred at 80 °C for 1 h. The reaction mixture was cooled to room temperature, triturated with CH 2 CI 2 and hexanes and dried under vacuum to give 0.010 g (0.023 mmol, 72%) of 35 as a pink solid, mp 246-248°C.
  • a dry 3 mL vial was sequentially charged with 5-oxo-5-((3-(6-(4- (trifluoromethyl)phenyl)-l,2,4,5-tetrazin-3-yl)phenyl)amino)pentanoic acid (170 mg, 0.46 mmol), /V-hydroxysuccinimide (86 mg, 0.20 mmol) and l-ethyl-3-(3- dimethylaminopropyl)carbodiimide hydrochloride (77 mg, 0.40 mmol).
  • the vial was capped by a screw cap with a Teflon septum.
  • the vial was swept with nitrogen, and anhydrous DMF (1.5 mL) was added via syringe.
  • the reaction mixture was allowed to stir for 34 h at room temperature.
  • the mixture was then diluted by CH 2 CI 2 (10 mL), centrifuged, and the supernatant was decanted.
  • the purple solid was subjected to three further cycles of suspension in CH 2 CI 2 (10 mL), centrifugation, and decantation to provide the 79 mg (75%) of the title compound as a purple solid (79 mg, 75%). !
  • a tetrazine conjugate 41 was prepared from c(RGDyC) (39) according to the following sequence.
  • tetrazine-maleimide 200 pg, 0.41 pmol
  • 100 pL dimethyl sulfoxide (DMSO) and 39 (200 pg, 0.33 pmol) (Peptides International of Louisville, KY) in 500 pL phosphate buffer (50 mM, pH 6.5-7.0) were mixed together at room temperature. After the mixture was stirred at room temperature for 5 h, the conjugate was purified by semipreparative HPLC. The collected fractions were combined and lyophilized to afford the final product as a white powder. Compound 40 was obtained in 85% yield.
  • tetrazine-maleimide 24 200 pg, 0.41 pmol in 100 pL DMSO and VEGF (100 pg, 5.5 nmol) in 500 pL phosphate buffer (50 mM, pH 6.5-7.0) were mixed together at room temperature. After the mixture was stirred at room temperature for 5 h, the conjugate was purified by size exclusion PD-10 column and concentrated by Centricon filter (Millipore, Bedford, MA), and the final concentration was determined based on UV absorbance at 280 nm using unconjugated VEGF of known concentrations as standard. The final concentration was adjusted to 50 pg/mL for use.
  • 19 F-9 was synthesized according to Li Z, Cai H, Hassink M, Blackman ML, Brown RC, Conti PS, et al. Tetrazine-trans-cyclooctene ligation for the rapid construction of 18F labeled probes.
  • 19 F-41 was obtained in 92% yield with 15.5 min retention time on analytical HPLC.
  • MALDI-TOF-MS was m/z 1226.4 for [MH] + (C57H73FN15O13S, calculated molecular weight 122
  • 18 F-9 was synthesized according to Li Z, Cai H, Hassink M, Blackman ML, Brown RC, Conti PS, et al. Tetrazine-trans-cyclooctene ligation for the rapid construction of 18F labeled probes. Chem Commun (Camb). 2010;46:8043-8045. 18 F-9 (148 MBq, 4 mCi) in about 50 pL ethanol was added to 40 (10 pg) in 50 pL DMSO followed by shaking for 5 min. The conjugate was purified by semipreparative HPLC. The collected fractions were combined and the solvent was removed by rotary evaporation under reduced pressure. 18 F-41 was reconstituted in 1 mL PBS and passed through a 0.22 pm syringe filter for in vivo animal experiments.
  • Human glioblastoma cell line U87MG cells were obtained from the American Type Culture Collection (Manassas, VA) and were cultured in DMEM containing high glucose (GIBCO, Carlsbad, CA), which was supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin. The cells were expanded in tissue culture dishes and kept in a humidified atmosphere of 5% C0 2 at 37°C. The medium was changed every other day. A confluent monolayer was detached with 0.05% Trypsin-EDTA, 0.01M PBS (pH 7.4) and dissociated into a single-cell suspension for further cell culture.
  • DMEM high glucose
  • FBS fetal bovine serum
  • penicillin-streptomycin penicillin-streptomycin
  • mice were anesthetized with isoflurane (5% for induction and 2% for maintenance in 100% 0 2 ) using a knock-down box.
  • isoflurane 5% for induction and 2% for maintenance in 100% 0 2
  • the biodistribution of 18 F-43 was evaluated in normal Sprague Dawley nude mice. Representative coronal microPET images at 0.5 and 3 h post injection of about 3.7 MBq (100 Ci) 18 F-43 were obtained. The activity was mainly accumulated in the liver and kidney. MicroPET quantification by measuring the ROI is shown in Figure 3B. The 10 kidney uptakes of 18 F-43 were 23.20 ⁇ 2.15 and 16.51 ⁇ 1.52 %ID/g at 30 min and 180 min post injection respectively. Low muscle uptakes were observed (2.98 ⁇ 0.66 and 1.59 ⁇ 0.57 %ID/g at 0.5 and 3 h post injection).
  • Diaminotetrazine 21 was re ared as follows.
  • Diisopropylethyl amine (0.032 ml, 0.195 mmol, 3 eq.) was added dropwise to the mixture. The reaction mixture was stirred for 24 hours at room temperature. The reaction was condensed in vacuo and then loaded onto a silica gel column using the minimum amount of MeOH required along with CH 2 CI 2 .

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

A Diels-Alder adduct of a irans-cyclooctene with a tetrazine is provided, wherein the adduct bears a substituent labeled with a radionuclide. A method of producing a PET or other image of an organ in an animal or human includes forming the Diels-Alder adduct in the animal or human. Trans-cyclooctenes and tetrazines suitable for preparing the adducts are provided.

Description

TETRAZINE-TRA/VS-CYCLOOCTENE LIGATION FOR THE RAPID CONSTRUCTION OF RADIONUCLIDE LABELED PROBES
BACKGROUND OF THE INVENTION
Positron emission tomography (PET) is a non-invasive imaging modality that utilizes positron-emitting radionuclides (C-ll, N-13, 0-15 and F-18). For example, F-18 PET has a number of attributes that make it clinically attractive, including 100% positron efficiency, a very high specific radioactivity, and a short half-life of ~110 min. However, the short half-life of F-18 and the poor nucleophilicity of fluoride render it difficult to incorporate F-18 in complex molecules. Currently, radiochemistry is a major limiting factor for the field of PET. Despite recent advances, challenges exist for improving F-18 incorporation with respect to reaction rates, efficiency, and selectivity.
The inventors have previously described tetrazine-trans-cyclooctene ligation OTTCO ligation') as a method of bioconjugation that proceeds with fast reaction rates without need for catalysis (M. L. Blackman, M. Royzen and J. M. Fox, J. Am. Chem. Soc. 2008, 130, 13518-13519.) tra/is-Cyclooctene derivatives are readily prepared from c/'s- cyclooctenes using a photochemical flow-reaction that the inventors developed (M.
Royzen, G. P. A. Yap and J. M. Fox, J. Am. Chem. Soc. 2008, 130, 3760-3761). The inventors have found that 3,6-diaryl-s-tetrazines offer an excellent combination of fast reactivity and stability for both the conjugate and starting material. In particular, 3,6 - di(2-pyridyl)-s-tetrazines have been shown to display excellent characteristics. Thus, the reaction between frans-cyclooctene and la proceeds with a rapid rate (k2 ~ 2000 M 1 s"1 in 9: 1 MeOH :water), and is successful in cell media and cell lysate. 3,6 -Di(2-pyridyl)- s-tetrazines can easily be functionalized as their amido derivatives ( lb), which display excellent st
Figure imgf000002_0001
Because of the fast rate of reactivity, the TTCO-ligation offers opportunities for the rapid conjugation of radionuclides to biomolecules, both of which are often available only at low concentration. SUMMARY OF THE INVENTION
In one aspect, the invention provides a Diels-Alder adduct of a trans-cyclooctene with a tetrazine, wherein the adduct bears a substituent labeled with a radionuclide.
In another aspect, the invention provides a method of producing a PET or other image of an organ in an animal or human. The method includes forming in the animal or human a Diels-Alder adduct of a trans-cyclooctene with a tetrazine, wherein the adduct bears a substituent labeled with a radionuclide.
In another aspect, the invention provides the compound according to structure 5
Figure imgf000003_0001
5
In another aspect, the invention provides the compound according to structure 9
Figure imgf000003_0002
In another aspect, the invention provides the compound according to structure 17
Figure imgf000003_0003
In another aspect, the invention provides the compound according to structure 13
Figure imgf000004_0001
In another aspect, the invention provides the compound according to structure 24 vides the compound according to structure 25
Figure imgf000004_0002
In another aspect, the invention provides the compound according to structure 26 provides the compound according to structure 27
Figure imgf000004_0003
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows cell binding affinity studies of c(RGDyK) ( 12) and 19F-15.
Figure 2 is a schematic representation of an automated synthesis module for performing fluorination (~200 mCi).
Figure 3A shows tumor and major organ uptake of 18F-41.
Figure 3B shows the biodistribution of 18F-43 in regions of interest in normal Sprague Dawley nude mice by microPET quantification .
DETAILED DESCRIPTION OF THE INVENTION
The invention provides an extremely fast and reactive method for generating radionuclide labeled probes based on TTCO-ligation, employing a Diels-Alder reaction between 3,6-diaryl-s-tetrazines and frans-cyclooctenes, one of which is labeled with a radionuclide, for example 18F. The reaction proceeds at exceptionally fast rates, providing effective conjugation method within seconds at low micromolar concentrations. Using the compounds and methods of the invention, it is possible to effectively label an organ in an animal or human for PET or other imaging. In a typical use, the Diels-Alder adduct is covalently bound to a biomolecule. This may be achieved via a linking group derived from a substituent present on the trans-cyclooctene or the tetrazine prior to reaction with the biomolecule.
The invention provides a number of ways to use TTCO-ligation for labeling, as will now be described. Labeling with 18F will be described first, followed by a description of labeling with other radionuclides.
18F-Labeled tetrazines
The inventors focused on the development of direct methods for 18F-incorporation via reactions with fluoride ion, with an initial focus on the synthesis of 18F-labeled tetrazines (Scheme 1). Attempts to convert nitrotetrazine derivatives 2a and 2b into 18F-labeled substitution products 3 using 18F-fluoride/kryptofix or 18F-TBAF gave decomposition products and only traces of radiolabeled products. The inventors also combined mesylate 4 with fluoride, and in the most successful experiment ( 18F-TBAF at 85°C for 15 min) 18F-labeled product 5 was obtained in ~1% labeling yield.
Figure imgf000006_0001
Scheme 1. Synthesis of 18F-labeled tetrazines. Attempts to prepare 3 by nucleophilic substitution were low yielding. Fluorination of 4 (18F-fluoride, MeCN, 85°C, 15 min) gave 5 in only 1% radiochemical yield.
18F-Labeled trans-cyclooctenes
The above difficulties prompted the inventors to consider methods for preparing 18F-labeled trans-cyclooctenes. To this end, the inventors synthesized the nosylate 8 as shown in Scheme 2. The key step in the synthesis was photoisomerization of 6 to 7 using a flow reactor that continuously removes the trans- isomers through selective metal complexation. See M. Royzen, G. Yap, and J. Fox, J. Am. Chem. Soc. 2008, 130 (12), 3760. The major diastereomer of 7 was carried forward in the synthesis of 8.
Figure imgf000006_0002
7 8
Scheme 2. Synthesis of trans-cyclooctene nosylate 8. Reagents and conditions: (a) NaH, a-bromoacetic acid, (b) CH2N2, 69%, 2 steps (c) DIBAL, 78% (d) Methyl benzoate- sensitized photoisomerization with active removal of trans-isomers, AgN03, 55%. (e) NsCI, Et3N, 87%.
Nosylate 8 reacted efficiently with TBAF to provide 19F-9 in high yield. As used herein, the notation 19F- as part of a structure name indicates that the compound has not been radiolabeled, and 18F- indicates labeling .
Conditions for the preparation of 18F-9 were then optimized (Table 1). It was found that 18F-9 could be obtained in good yield by adding nosylate 8 to a mixture of [18F]-fluoride (100 mCi)/ tetrabutylammonium bicarbonate (TBAB) in acetonitrile (0.8 ml_). The inventors determined the effect of varying the concentration of 8 in reactions conducted for 15 min at 75°C (Table 1, entries 1-4).
Figure imgf000007_0001
Table 1. Optimization of synthesis of F-labeled rrans-cyclooctene
Entry Amount of 8 Temp. Reaction Radiochemical
Time Yield
1 100 g 75°C 15 min 18%
(0.35 mM)
2 500 g 75°C 15 min 25%
(1.8 mM)
3 2.0 mg 75°C 15 min 71%
(7.0 mM)
4 3.0 mg 75°C 15 min 71%
(11 mM)
5 2.0 mg 40°C 15 min 24%
(7.0 mM)
6 2.0 mg 55°C 15 min 34%
(7.0 mM)
7a 2.0 mg 90°C 15 min 71%
(7.0 mM)
8 2.0 mg 75°C 3 min 43%
(7.0 mM)
9 2.0 mg 75°C 7 min 68%
(7.0 mM)
10 2.0 mg 75°C 30 min 71%
(7.0 mM)
a Comparable results (70% radiochemical yield) were obtained under similar conditions using K2CO3/K222 instead of TBAB.
The highest labeling yield (71%) was achieved with 7.0 mM 8, but a useful radiochemical yield (18%) was still obtained with 0.35 mM 8. Different reaction temperatures were studied, and 75°C was found to be optimal (Table 1, entries 3, 5-7).
Finally, the inventors investigated the efficiency of the 18F labeling as a function of time using 2 mg of 8 at 75°C (Table 1, entries 8-10). While the labeling yield was optimal (71%) in an experiment conducted for 15 min, useful labeling yields were also obtained after 3 min (43%) and 7 min (68%). Overall, entry 3 appeared optimal for 18F labeling.
More generally, the invention provides analogs of 18F-9 in which the spacer group between 0 and 18F is an alkylene group. For example, the alkylene group may be a C2 alkylene group (as in 18F-9), or it may be any alkylene group from C3 to C20, more typically from C3 to CIO, and most typically from C3 to C5. In another exemplary embodiment, an analog of 18F-9 uses an 18F-CH2-phenyl moiety in place of the 18F-CH2- CH2 moiety of 18F-9. Such a compound may be prepared from the corresponding bromide, chloride, iodide, sulfonate, etc. by nucleophilic displacement with 18F fluoride.
The inventors also used a photochemical flow reactor (M. Royzen, G. Yap, and J. Fox, J. Am. Chem. Soc. 2008, 130 ( 12), 3760) to synthesize 16, an analog of 7 with a functionalizable group. The rate of the reaction of compound 16 with 3,6-di(2-pyridyl)- s-tetrazine (la) in methanol was k2 = 22000 M' 1 , 18 times faster than the reaction of la with frans-cyclooctene in the same solvent. In aqueous solvent systems, reaction between la and 16 was even faster, in fact too fast to measure by UV-vis spectroscopy. (Tetrazine ligations show significant acceleration due to the hydrophobic effect.) In addition to excellent reactivity, 16 also displayed excellent stability, showi ng no degradation after 24 hours in water or human serum or when exposed to 5 mM n- butylamine in MeOH or to 5 mM ethanethiol in MeOH for 24 hours. Compound 16 can be readily converted into carbamate derivatives, and compound 16 can be selectively conjugated to the protein thioredoxin using the conjugation of maleimide 26 (vide infra) to a cysteine residue, and that conjugate can in turn can take part in tetrazine ligation analogous to that sho
Figure imgf000008_0001
1a
Labeled compound 18F-17 may be prepared from compound 16 using the same general procedures used for making labeled compound 18F-9.
Figure imgf000008_0002
Reaction of 18F-labeled frans-cyclooctenes with tetrazines
Derivatives of 3,6-di(2-pyridyl)-s-tetrazine (e.g ., lb) are readily prepared by known methods, for example as described by M . L. Blackman, M. Royzen and J. M. Fox, J. Am. Chem. Soc. 2008, 130, 13518-13519 and R. Rossin, P. R. Verkerk, S. M. v. d . Bosch, R. C. M. Vulders, I. Verel, J . Lub and M . S. Robillard, Angew. Chem. Int. Ed.
2010, 49, 3375-3378. Therefore, 3,6-di(2-pyridyl)-s-tetrazine (la) was used to test the efficiency of 18F-labeled irans-cyclooctene 9 in the TTCO-ligation. A 'cold' study with 19F- 9 was initially conducted . The conjugate 19F-10 formed immediately, and then slowly isomerized to 1,4-dihydropyrazine 19F-11 as a mixture of isomers (Scheme 3, showing the corresponding 18F compounds). These isotopically stable conjugates served as co- inj
Figure imgf000009_0001
Scheme 3 Conjugation between 9 and la occurs rapidly to form conjugate 10, which slowly rearranges to 11 as a mixture of regio- and stereoisomers.
The conjugations between la and 18F-9 were carried out in 1 : 1
acetonitrile/water, and were analyzed within 10 seconds of mixing . Prior to the conjugation, 18F-9 was purified by HPLC and easily separated from unreacted precursor 8. When 18F-9 (1 mCi, 2 μΜ) was combined with la (concentrations of >21 μΜ), 18F-9 was completely consumed with 10 s, and 18F-10 had formed in 98% radiochemical yield, accompanied by 18F-11 ( 1%) (Table 2, entries 1 -2) . When the concentration of 18F-9 was decreased to 0.1 mCi (0.2 μΜ), the conjugate 18F-10/11 was still formed in excellent radiochemical yield (98%, entry 3). Useful radiochemical yields could also be obtained with even lower concentrations of la (entries 4-5) . The inventors also investigated the efficiency of the conjugation between la (21 μΜ) and 18F-9 ( 1 mCi, 2 μΜ) in PBS buffer and serum media, and found formation of 18F-10 in quantitative yield within 10 seconds (entries 6-7) . Table 2 The effect of concentration on the formation of 18F-10 through the conjugation between 18F-9 and 3,6-di(2-pyridyl)-s-tetrazine. All reactions were performed at room temperature.
Entry 18F-9a la Solvent Reaction Radiochemical Yield
time (18F-10 + 18F-11)
1 1 mCi 210 μΜ MeCN/H20 < 10 s >98 %
(2 μΜ)
2 1 mCi 21 M MeCN/H20 < 10 s >98 %
(2 μΜ)
3 0.1 mCi 21 MM MeCN/H20 <10 s 98 %
(0.2 μΜ)
4 0.1 mCi 2.1 MM MeCN/H20 < 10 s 56 %
(0.2 μΜ)
5 0.1 mCi 0.21 M MeCN/H20 < 10 s 15 %
(0.2 μΜ)
6 1 mCi 21 MM PBS buffer < 10 s >98 %
(2 μΜ)
7 1 mCi 21 MM Serum < 10 s >98 %
(2 μΜ)
aThe concentration of 18F-9 was estimated based on the specific activity of fluoride after bombardment (~4 Ci/Mmol), taking into account a correction for the rate of radioactive decay.
The Diels-Alder conjugate 10 was found to be stable in water, and the benign isomerization to 11 was the only side reaction. Thus, 19F-10 was the only product detected by :H NMR analysis immediately after the conjugation. In CD3CN/H20, the rearrangement of 19F-10 to 19F-11 proceeded to 11% conversion after 4 hours, and >95% conversion after 48 hours. The stability of the radiolabeled conjugation product was monitored in PBS buffer and serum media for 4 hours, and no degradation products of 18F-10 were observed.
18F-Labelinq of biomolecules
As will now be described, biomolecules may be readily radiolabeled using bioconjugation based on the above-described TTCO-ligation methods and compositions. This makes possible reliable methods for 18F-labeling of biomolecules in PET and other in vivo applications. In one embodiment, the invention provides a protein or peptide having covalently attached thereto a cyclooctane bearing an 18F-containing substituent.
Herein, the inventors report the construction of a 18F labeled PET probe for imaging (such as cancer imaging) based on the tetrazine-trans-cyclooctene ligation.
Figure imgf000011_0001
15
Scheme 4
The integ rin ανβ3 is upregulated on the endothelial surface of tumor blood vessels, and has been linked to tumor progression and metastasis. Radiolabeled synthetic RGD antagonists of the integ rin ανβ3 have been shown to be effective tools for cancer imaging . Although strateg ies for F- 18 incorporation into RGD mimics are known in the art, they typically involve lengthy synthetic procedures. Consequently, it is challenging to obtain high radiochemical yields and to develop synthetic protocols that can be auto mated. It has now been found that F- 18 labeled RGD peptides can be constructed using tetrazine- frans-cyclooctene ligation, thus allowing cancer detection, patient stratification, and treatment monitoring through PET imaging of integrin ανβ3 expression in vivo. One way of doing this involves the use of labeling with 18F as shown in Scheme 4.
As shown in Scheme 4, tetrazine-RGD conjugate 14 was readily prepared by coupling NHS-ester 13 and the peptide c(RGDyK) ( 12) . To provide an LC-standard, the tetrazine-RGD conjugate 14 was then combined with 19F-TCO (9) to provide 19F-15 as a mixtu re of isomers (due to isomerization of the initially formed 4, 5 -dihydropyridazine to the corresponding 1,4-dihydropyridazine) .
Cell bind ing affinity studies of c(RGDyK) ( 12) and 19F- 15 were conducted (Figure 1 ) . Both peptides inhibited the binding of 125I-echistatine to U87MG cells (integrin expositive human glioblastoma) in a dose dependent manner. The IC50 values for c(RGDyK) and 19F-tetrazine-RGD were ( 1. 16 ±0.35) x 10"7 and ( 1.93 ± 0.27) x 10"7 M, respectively, indicating that the fluorinated label had only a minimal effect on the integrin binding affinity of the RGD moiety of 15.
The reaction speed for labeling tetrazine-RGD peptide 14 with 18F-9 was then investigated . 18F-9 was synthesized from nosylate 8. The conjugation of 18F-9 and tetrazine-RGD was performed at room temperature for 5 min. The conjugation was efficient and high yielding : starting with only 30 pg (78 μΜ) of tetrazine-RGD conjugate 14 and 2 mCi (5 μΜ) of 18F-rrans-cyclooctene, the labeling yield was 90% after 5 min by HPLC analysis. The stability of 18F-tetrazine-RGD conjugate 15 was evaluated in PBS in the presence 5% EtOH. The stability was excellent: >95% of the tracer remained after incubation for 6 h as judged by radio-HPLC analysis.
Static microPET scans were performed for 18F-tetrazine-RGD conjugate 15, via injection into athymic female nude mice bearing an U87MG tumor. High tumor accumulation was observed as early as 30 min time point. The tumor uptake was 4.6 ± 0.2, 4.4 ± 0.6, 4.2 ± 0.6, and 2.7 ± 0.5 %ID/g at 0.5, 1, 2, 4 h post injection, respectively. 18F-15 was cleared through both liver and kidney. Up to 4 h post injection, there was still a fair amount of activity accumulated in the abdomen, which may be a result of the relative hydrophobicity of this system.
Blocking experiments were also performed by co-injecting 10 mg/kg of c(RGDyK) along with 18F-15. The tracer uptake in U87MG tumor dropped to 1.4 ± 0.2, 1.0 ± 0.3, 0.6 ± 0.2, and 0.4 ± 0.1 %ID/g at 0.5, 1, 2, 4 h post injection respectively. The successful blocking confirmed the integrin ανβ3 specific binding of the tracer 18F-15.
Figure imgf000012_0001
In another example, the conjugation of 18 with 9 to give conjugate 19 is extremely rapid and extremely efficient, and conjugate 19 oxidized spontaneously in solution to form the aromatized compound 20.
Figure imgf000013_0001
An in vivo metabolic study of 19 was carried out by injecting F-labeled 19 into an athymic nude mouse that was sacrificed 2 h post injection. Major tissues were collected and homogenized. The activity was extracted (CH 3CN), filtered (C18 Sep-Pak cartridge), evaporated and analyzed by HPLC. Fractions were collected each minute and radioactivity measured with the γ-counter and analyzed by HPLC. The average fraction of intact tracer was significantly improved compared with compound 15, which showed significant hydrophilic degradation product. For 18F-19, a hydrophilic byproduct was not observed.
No defluoridation of 18F-19 was observed as no visible bone uptake was observed in any of the microPET scans. 18F-19 exhibited good metabolic stability in vivo, and injection of 18F-19 into a U87MG mouse model resulted in an effective method for ανβ3 imaging . The integrin ανβ3 receptor specificity was confirmed by blocking experiments, in which unlabeled cRGD was administered prior to the injection of the 18F-19.
In a further example, the inventors have synthesized the diaminotetrazine 21 and derivative 22, which can be expected to show rapid conjugation with trans-cyclooctenes such as 9 while affording robust in vivo stability.
Figure imgf000013_0002
In another aspect, the invention provides efficient methods for conjugating tetrazine and irans-cyclooctenes to proteins. As shown below, the amine-reactive NHS- ester 13 can be readily converted into the thiol-reactive maleimide derivative 24. The reactive functionalities of these molecules may be used to conjugate tetrazines to lysine and cysteine residues of proteins, for example a VEGF protein. NHS-ester 13 can be prepared from 5-oxo-5-(6-(6-(pyridin-2-yl)-l,2,4,5-tetrazin-3-yl)pyridin-3- ylamino)pentanoic acid, in turn prepared by the method of R. Rossin, P. R. Verkerk, S. M . v. d . Bosch, R. C. M . Vulders, I. Verel, J . Lub and M . S. Robillard, Angew. Chem. Int. Ed. 2010, 49, 337
Figure imgf000014_0001
Similarly, the inventors have prepared the amine-reactive (25) and thiol-reactive (26, 27) bifunctional conjugates of frans-cyclooctenes shown below. Compound 25 was readily accessed in 62% yield by the reaction of 16 with 4-nitrophenyl chloroformate. Treatment of 25 with n-(2-aminoethyl)maleimide trifuoroacetate salt yielded 26 in 32% yield . Compound 27 was prepared according to the method of M. L. Blackman, M .
Royzen and J. . Fox, J. Am. Chem. Soc. 2008, 130, 13518-13519.
Figure imgf000015_0001
Figure imgf000015_0002
The inventors have conjugated maleimides 26 and 27 to a protein (thioredoxin) using standard methods known in the art, and the conjugates have been shown to undergo rapid tetrazine ligation in high yield . Other proteins may be similarly conjugated according to the invention. Compound 33, the conjugate of 27 with thioredoxin, was rapidly adducted with 3,6-di(2-pyridyl)-s-tetrazine ( la) as shown below, where Trx represents a thioredoxin residue.
Figure imgf000015_0003
VEGF proteins may be labeled with 18F via modification of the lysines of wild -type VEGF12i protein with amine reactive derivatives (e.g., 24 or 25). The resulting conjugation products (29 and 30, respectively) may be combined with irsns-cyclooctene 18F-17 and tetrazine 18F-5 to provide radiolabeled conjugates 18F-31 and 18F-32, respectively.
Figure imgf000016_0001
Further exemplary labeled compounds according to the invention include the following.
Figure imgf000016_0002
The foregoing results demonstrate an efficient labeling method for PET or other probe construction based on tetrazine-frans-cyclooctene ligation using the methods and compositions of the invention. A major advantage of these techniques lies in the ability to achieve fast and efficient bioconjugation at low concentration. Although the above examples employ specific exemplary biomolecules, the methods and compositions described herein are not limited to those embodiments and labeling of any biomolecule is within the scope of the invention. Other nonlimiting examples of suitable biomolecules that may be labeled include peptides, proteins, antibodies, antibody fragments, and modified oligonucleotides, among others.
Labeling of biomolecules with radionuclides using tetrazine -frans-cyclooctene ligation is not limited to the specific frans-cyclooctenes and tetrazines discussed above; any radiolabeled tetrazine or rrans-cyclooctene may be used. The invention encompasses labeling of biomolecules with radionuclides other than F. Suitable further examples of compounds according to the invention, all of which may be used to label
Figure imgf000017_0001
The use of I labeling may for example be suitable for producing PET probes, while 125I and 131I may be suitable for preparing SPECT probes.
EXAMPLES
General Considerations
All commercially available chemical reagents were used without further purification. Chromatography was performed using Silacycle P60 silica gel. All moisture sensitive reactions were carried out in glassware that was flame-dried under vacuum and cooled under nitrogen. Solid phase extraction cartridges (silica gel, 900 mg) were purchased from Waters. Ion exchange cartridges were purchased from ABX (Germany). Z)-2-(cyclooct-4-en-l-yloxy)acetic acid
Sodium hydride (1.14 g, 26.4 mmol) was added to a flame dried rou nd bottom flask. The INaH was washed with dry hexane (10 mL) and then decanted. Dry
tetrahydrofuran (15 mL) was added and the mixture was allowed to stir at room temperature. cis-Cyclooctene-4-ol (P. Lombardi, Chemistry and Industry 1990, 21, 708) (0.834 g, 6.61 mmol) in tetrahydrofuran (10 mL) was added to the flask. The mixture was stirred and heated to reflux for 1 hour. a-Bromoacetic acid (0.919 g, 6.61 mmol) in tetrahydrofuran (25 mL) was added and the mixture was allowed to reflux overnight. The mixture was cooled to room temperature, and then concentrated on a rotary evaporator. The residue was cooled in an ice bath and water was added followed by acidification with 3 M HCI . The aqueous layer was extracted with three portions of ether. The extracts were dried with MgS04 and concentrated in vacuo to provide an oil. The title compound was used directly for the next reaction without further purification.
*H NMR (400 MHz, C6D6) δ 5.83 (br s, 1H), 5.55-5.43 (m, 2H), 3.70 (d, JAB= 16.7 Hz 1H), 3.67 (d, JAB= 16.7 Hz, 1H), 3.19-3.14 (m, 1 H), 2.11-2.07 (m, 1H), 1.99- 1.84 (m, 2H), 1.80-1.69 (m, 2H), 1.67- 1.53 (m, 2H), 1.45-1.34 (m, 2H), 1.19-1.08 (m, 1H).
13C NMR ( 100 MHz, C6D6) δ 174.7 (C), 130.2 (CH), 129.5 (CH), 81.8 (CH), 65.6 (CH2), 34.0 (CH2), 33.1 (CH2), 25.8 (CH2), 25.5 (CH2), 22.7 (CH2) .
IR (liquid, CHCI3, cm"1) 3235, 2933, 2859, 1733, 1619, 1453, 1330, 1162, 1133, -ESI m/z: [M+Na] calcd for Ci0H16O3Na, 207.0997; found 207.0994.
Figure imgf000018_0001
(Z)-methyl 2-(cyclooct-4-en-l-yloxy)acetate
An Erlenmeyer flask was sequentially charged with (Z) -2-(cyclooct-4-en- l- yloxy)acetic acid (1.45 g, 7.90 mmol) and diethyl ether ( 150 mL). Diazomethane was introduced to this flask using the apparatus developed by Lombardi.1 Thus, Diazald (5.07 g, 23.7 mmol) and ethanol ( 150 mL) were added to a stoppered flask, equipped to bubble into the aforementioned Erlenmeyer flask. The ethanol mixture was sparged with nitrogen, and a blast shield was placed in front of the two flasks. Sodium hydroxide (7.30 g, 182 mmol) in water ( 10 mL) was slowly added via syringe to the ethanol - containing flask. Nitrogen was bubbled through this Lombardi flask until no yellow color persisted in either flask. The reaction was purified by column chromatography using 5% ether to 30% ether in hexanes as the eluent to give 1.08 g (69%, 5.47 mmol) of the title compound as a colorless oil.
*H NMR (400 MHz, C6D6) δ 5.61-5.45 (m, 2H), 3.84 (d, JAB= 16.6 Hz, 1H), 3.84 (d, JAB= 16.6 Hz, 1H), 3.37 (app dt, J = 4.1, 9.1 Hz, 1H), 3.29 (s, 3H), 2.22-2.13 (m, 1H), 2.05-1.78 (m, 5H), 1.75- 1.65 (m, 1H), 1.60-1.46 (m, 2H), 1.30- 1.20 (m, 1H).
13C NMR ( 100 MHz, C6D5) δ 171.0 (C), 130.4 (CH), 129.5 (CH), 81.5 (CH), 66.1
(CH2), 51.0 (CH3), 34.3 (CH2), 33.4 (CH2), 26.0 (CH2), 25.8 (CH2), 22.8 (CH2).
IR (liquid, CHCI3, cm- 1) 3155, 3019, 2978, 2934, 2860, 1753, 1440, 1383, 1291, 1216, 1129, 900, 722, 650.
HRMS-CI (NH3)m/z: [M+NH4] calcd for CuH22N03, 216.1599; found 216.1590.
Figure imgf000019_0001
(Z)-2-(cyclooct-4-en-l-yloxy)ethanol (6)
(Z)-methyl 2-(cyclooct-4-en-l-yloxy)acetate (2.34 g, 11.9 mmol) and anhydrous ether ( 150 mL) were sequentially added to a dry round bottom flask. The flask was cooled to -78 °C. DIBAL (9.03 mL, 47.6 mmol) in ether (30 mL) was added slowly via syringe to the flask. The reaction mixture was allowed to stir for another 3 hours at -78 °C, then warmed to 0 °C and stirred for a further 3 hours. The reaction was quenched at 0 °C with Na2SO4» 10 H20. The mixture was concentrated in vacuo. The reaction was purified by column chromatography using 5% ether to 30% ether in hexanes as the eluent to give 1.55 g (78%, 9.31 mmol) of the title compound as a colorless oil.
*H NMR (400 MHz, C6D6) δ 5.61-5.48 (m, 2H), 3.54-3.51 (m, 2H), 3.22-3.15 (m, 3H), 2.19-2.13 (m, 1 H), 2.03-1.92 (m, 3H), 1.85-1.74 (m, 3H), 1.68-1.62 (m, 1H), 1.51- 1.44 (m, 2H), 1.25-1.20 (m, 1H) .
13C NMR ( 100 MHz, C6D6) δ 130.4 (CH), 129.5 (CH), 80.9 (CH), 69.8 (CH2), 62.2 (CH2), 34.5 (CH2), 33.4 (CH2), 26.1 (CH2), 25.8 (CH2), 22.9 (CH2) .
IR (liquid, CHCI3, cm"1) 3456, 3011, 2975, 2936, 2861, 1650, 1447, 1392, 1252, 1100, 1049, 988, 875.
HRMS-ESI m/z: [M+Na] calcd for Ci0Hi8O2Na, 193.1204; found 193.1205.
Major diastereomer:
Figure imgf000019_0002
2-[re/-( lR-4E-pR)-cyclooct-4-en-l-yloxy]ethanol (7)
Minor diastereomer:
Figure imgf000019_0003
2-[re/-(lR-4E-pS)-cyclooct-4-en-l-yloxy]ethanol (7)
(Z)-2-(cyclooct-4-en- l-yloxy)ethanol ( 1.54 g, 9.31 mmol) and methyl benzoate (2.49 g, 18.6 mmol) were dissolved in 500 mL of 9 : 1 ethen hexane in a quartz flask. The photoisomerization was carried out using the flow apparatus described in M . Royzen, G. Yap, and J. Fox, J. Am. Chem. Soc. 2008, 130 ( 12), 3760. ] . The following minor modifications were made: a Biotage "SNAP cartridge" column (50 g, Biotage part no. FSKO-l 107-0050) was used, and the FMI pump was a model QG 400. The column was packed with 8.5 cm of silica gel, and then silver impregnated silica gel ( 16 g) on top. The column was flushed with 9 : 1 ether: hexane (250 mL) . The pump was turned on at a flow rate of 100 mL/min and irradiation begun. Photoisomerization of the mixture was carried out for 6 hours. The column was flushed with 9 : 1 ether: hexane (250 mL) and then dried with compressed air. The silica was placed into an Erlenmeyer flask and stirred with ammonium hydroxide (200 mL) and methylene chloride (200 mL) for 5 min. The silica gel was filtered and the filtrate was placed into a separatory funnel. The organic layer was separated, and the ammonium hydroxide layer was extracted three times with methylene chloride. The organic layers were combined and twice washed with water. The organic layers were dried with MgS04, filtered, and purified by column chromatography with 5% ether to 30% ether in hexanes. Two diastereomers were isolated 0.572 g (3.44 mmol, 37%) of 2-[re/-(lR-4E-pR)-cyclooct-4-en- l-yloxy]ethanol and 0.275 g ( 1.66 mmol, 18%) of 2-[re/-( lR-4E-pS)-cyclooct-4-en-l-yloxy]ethanol as colorless oils. The major diastereomer was contaminated by 7% of the cis-isomer: peaks attributable to cis-isomer 5.61-5.48 (m), 1.51- 1.44 (m). The structures were assigned on the basis of the chemical shift for the C- l methine, as described by M. Royzen, G. Yap, and J. Fox, J. Am. Chem. Soc. 2008, 130 ( 12), 3760.
Spectroscopic properties of the minor diastereomer:
*H NMR (400 MHz, C6D6) δ 5.75-5.68 (m, IH), 5.44-5.36 (m, IH), 3.56 (m, 2Η), 3.31-3.29 (m, IH), 3.28-3.21 (m, IH), 3.16-3.11 (m, IH), 2.44 (br s, IH), 2.41-2.31 (m, IH), 2.24-2.21 (m, IH), 2.12-2.08 (m, IH), 1.99- 1.91 (m, 2H), 1.89- 1.71 (m, 2H), 1.63-1.57 (m, IH), 1.25- 1.17 (m, IH), 0.98-0.91 (m, IH) .
13C NMR ( 100 MHz, C6D6) δ 136.0 (CH), 131.5 (CH), 75.2 (CH), 70.4 (CH2), 62.2 (CH2), 40.4 (CH2), 34.9 (CH2), 33.2 (CH2), 30.2 (CH2), 27.9 (CH)
IR (liquid, CHCI3, cm"1) 3428, 3021, 2923, 2859, 1655, 1442, 1352, 1215, 1135,
1099, 1050, 989, 907, 738
Spectroscopic properties of the major diastereomer:
H NMR (400 MHz, C6D6) δ 5.56-5.34 (m, IH), 5.22-5.14 (m, IH), 3.53-3.52 (m, 2H), 3.22-3.17 (m, IH), 3.12-3.07 (m, IH), 2.81-2.77 (m, I H), 2.20-2.16 (m, 2H), 2.05-2.00 (m, 2H), 1.95- 1.92 (m, IH), 1.81- 1.68 (m, 4H), 1.35-1.20 (m, 2H).
13C NMR (100 MHz, C6D6) δ 135.5 (CH), 132.2 (CH), 86.0 (CH), 69.6 (CH2), 62.2 (CH2), 41.1 (CH2), 38.0 (CH2), 34.8 (CH2), 33.2 (CH2), 31.9 (CH2) .
IR (liquid, CHCI3, cm"1) 3449, 3012, 2935, 2861, 1647, 1445, 1353, 1198, 1096, 1050, 993, 968, 797.
-CI (NH3) m/z: [M+H] calcd for C10Hi9O2, 171.1385; found 171.1384.
Figure imgf000020_0001
2-[re/-( lR-4E-pR)-cyclooct-4-en-l-yloxy]ethyl 4-nitrobenzenesulfonate (8)
Triethylamine (0.21 mL, 1.5 mmol) was added to a flame dried round botto m flask containing anhydrous ether (5 mL). p-Nitrosulfonyl chloride (0.073 g, 0.33 mmol) from a freshly opened bottle was added to the flask. The mixture was stirred at room temperature for 30 minutes. The mixture was cooled to 0 °C and 2-[re/-(lR-4E-pR)- cyclooct-4-en-l-yloxy]ethanol was added. The mixture was allowed to stir for 5 hours at 0 °C. The cold mixture was directly transferred to a column of silica gel. Flash
chromatography using a gradient of 5% ether/hexane to 20% ether/hexane as the eluent to afford 0.093 g (87%, 0.26 mmol) of the title compound as a white solid.
In experiments using an aged bottle of p-Nitrosulfonyl chloride, 20% of (R,Z)-2- (cyclooct-4-en-l-yloxy)ethyl 4-nitrobenzenesulfonate was formed. The E-isomer could be separated using preparative, reverse phase HPLC (C-18, 20 x 250 cm, 65%
methanol/H20). For 18F labeling experiments 2-[re/-(lR-4E-pR)-cyclooct-4-en-l- yloxy]ethyl 4-nitrobenzenesulfonate was purified away from the cis isomer.
H NMR peaks attributable to the cis isomer: 5.59-5.47 (m)
XH NMR (400 MHz, C6D6) δ 7.50 (m, 4H), 5.34-5.26 (m, IH), 5.17-5.10 (m, IH), 3.84-3.82 (m, 2H), 3.05-3.02 (m, IH), 2.99-2.94 (m, IH), 2.65-2.61 (m, IH), 2.17-2.10 (m, 2H), 2.02-1.92 (m, IH), 1.82-1.77 (m, IH), 1.74-1.63 (m, 2H), 1.59-1.50 (m, 2H), 1.24-1.12 (m, 2H).
13C NMR (100 MHz, C6D6) δ 142.0 (C, 2 peaks), 135.8 (CH), 131.2 (CH), 131.2 (CH), 124.1 (CH), 75.3 (CH), 70.4 (CH2), 65,9 (CH2), 39.8 (CH2), 34.7 (CH2), 33.0 (CH2), 30.0 (CH2), 27.6 (CH2).
IR (liquid, CHCI3, cm"1) 3105, 3010, 2935, 1609, 1536, 1351, 1187, 1097, 932, 857, 776, 616.
HRMS-LIFDI m/z: [M+] calcd for C16H21N06S, 355.1089; found 355.1083.
Figure imgf000021_0001
re/-(lR-4E-pR)-5-(2-fluoroethoxy)cyclooct-l-ene (9)
A dry round bottom flask was sequentially charged with anhydrous acetonitrile ( 1 mL), 2-[re/-(lR-4E-pR)-cyclooct-4-en-l-yloxy]ethyl 4-nitrobenzenesulfonate (0.0037g, 0.010 mmol) ,and TBAF (0.17 mmol, 0.17 mL of a 1M solution in THF). The reaction was heated to 80 °C and allowed to stir for 3 hours. The mixture was cooled and then transferred directly to a column of silica gel. Flash chromatography with a gradient of pentane to 5% ether/pentane as the eluent to afford the title compound. After chromatography, most of the solvents were removed on a rotary evaporator. However, the compound was not dried in vacuo due to volatility. The yield was estimated to be 63% by adding an H NMR standard- mesitylene (0.015 mL, 0.010 mmol). Minor peaks attributable to the cis isomer were detected in the *H NMR spectrum at: 5.61-5.47 (m), 1.55-1.47 (m). Minor peaks attributable to the cis isomer were detected by 13C NMR at: 130.4, 129.5, 80.9, 34.5, 26.0, 25.8, 22.9, 22.7. XH NMR (400 MHz, C6D6) δ 5.40-5.33 (m, IH), 5.22-5.14 (m, IH), 4.16 (dt, JHF= 48 Hz, JHH= 4.3 Hz, 2H), 3.26-3.21 (m, 2H), 3.19-3.13 (m, IH), 2.85-2.74 (m, IH), 2.21-2.14 (m, 2H), 2.09-1.99 (m, 2H), 1.83-1.67 (m, 4H), 1.36-1.21 (m, 2H).
13C NMR (100 MHz, C6D6) δ 135.6 (CH), 132.2 (CH), 86.2 (CH), 83.2 (d, JC-F= 169 Hz, CH2), 67.5 (d, JC-F=20 Hz, CH2), 67.4 (CH2), 41.1 (CH2), 38.0 (CH2), 34.8 (CH2), 33.2 (CH2), 31.9 (CH2).
19F NMR (376.5 MHz, C6D6) δ 222.3
IR (liquid, CHCI3, cm"1) 3095, 2934, 2860, 1610, 1445, 1191, 1104
-CI (NH3)m/z: [M+H] calcd for Ci0Hi8OF, 173.1342; found 173.1342.
Figure imgf000022_0001
7-(2-fluoroethoxy)-l,4-di(pyridin-2-yl)-
4a,5,6,7,8,9,10,10a-octahydrocycloocta[rf]pyridazine (10)
A dry round bottom flask was charged with 3,6-di(2-pyridyl)-s-tetrazine (0.005 g, 0.02 mmol) and anhydrous acetonitrile (1 ml_). The mixture was allowed to stir at room temperature and re/-(lR-4E-pR)-5-(2-fluoroethoxy)cyclooct-l-ene in acetonitrile (0.5 mL) was added dropwise to the flask until yellow color persisted. Flash chromatography using a gradient of 10% acetone/hexane to 60% acetone/hexane as the eluent yielded an 8 : 2 mixture of 7-(2-fluoroethoxy)-l,4-di(pyridin-2-yl)-4a,5,6,7,8,9,10,10a- octahydrocycloocta[d]pyridazine and 8-(2-fluoroethoxy)-l,4-di(pyridin-2-yl)- 2,4a, 5,6,7, 8,9,10-octahydrocycloocta[d]pyridazine in 90% yield, as judged by ^ MR. Rearrangement of 10 to 11 occurs at room temperature, so analytical data must be collected within 10 min and without chromatographic purification, to minimize peaks from the rearrangement product.
*H NMR (400 MHz, CD3CN) δ 8.73-8.72 (m, 2H), 8.30-8.25 (m, 2H), 7.97-7.86 (m, 2H), 7.48-7.45 (m, 2H), 4.68 (dm, JHF= 48 Hz, 2H), 4.08-4.03 (m, IH), 3.98-3.93 (m, IH), 3.84-3.77 (m, 2H), 3.73-3.70 (m, IH), 2.18-2.08 (m, 5H), 1.85-1.81 (m, 4H), 1.71-1.62 (m, IH)
19F NMR (376.5 MHz, C6D6) δ 223.3
HRMS-CI (NH3)m/z: [M+H] calcd for C22H26N4OF, 381.2090; found 381.2080.
Figure imgf000023_0001
8-(2-fluoroethoxy)-l,4-di(pyridin-2-yl)-
2,4a,5,6,7,8,9,10-octahydrocycloocta[ef]pyridazine ( 11)
D20 (0.1 mL) was added to 7-(2-fluoroethoxy)-l,4-di(pyridin-2-yl)- 4a,5,6,7,8,9, 10, 10a-octahydrocycloocta[c |pyridazine in acetonitrile d-3 ( 1 mL) . The mixture was allowed to stand at room temperature for 48 hours. Column
chromatography afforded the title compound. H NMR analysis indicated 97% yield of the title compound.
IH NMR peaks attributable to aliphatic impurity: 1.30, 0.89.
*H NMR (400 MHz, CD3CN) δ 8.99 (br s, IH), 8.68-8.67 (m, I H), 8.59-8.57 (m, IH), 8.09-8.07 (m, IH), 7.90-7.86 (m, IH), 7.79-7.74 (m, IH), 7.65-7.63 (m, IH),
7.40-7.36 (m, IH), 7.31-7.28 (m, IH), 4.51 (dt, JHF= 48 Hz, JHH= 4.1 Hz 2H), 4.38-4.34 (m, IH), 3.75-3.57 (m, 3H), 2.94-2.90 (m, I H), 2.31-2.27 (m, IH), 1.91- 1.75 (m, 4H), 1.68- 1.60 (m, 3H), 1.46- 1.37 (m, 2H)
13C NMR ( 100 MHz, CD3CN) δ 155.3 (C), 152.7 (C), 150.3 (CH), 149.6 (CH), 144.1 (C), 137.8 (CH), 137.1 (CH), 135.7 (C), 125.0 (CH), 124.1 (CH), 123.7 (CH), 121.3 (CH), 110.6 (C), 84.5 (d, JC-F= 165 Hz, CH2), 80.0 (C), 68.1 (d, JC-F= 19 HZ, CH2), 35.5 (CH), 33.4 (CH2), 31.4 (CH2), 27.0 (CH2), 25.4 (CH2), 22.0 (CH2)
19F NMR (376.5 MHz, C5D5) δ 223.3
IR (liquid, CHCI3, cm-1) 2934, 2861, 1708, 1599, 1571, 1462, 1361, 1225, 1117, 1047
HRMS-CI (NH3)m/z: [M + H] calcd for C22H26N4OF, 381.2090; found 381.2085. HPLC methods for analyzing radiolabeled materials and standards
The purification of the crude product was carried out on a analytical reversed - phase high performance liquid chromatography (HPLC) system equipped with a dual UV absorbance detector (Waters 2487) using a Phenomenex C18 RP ( 150 x 4.6 mm 5 micron). The flow was 1 mL/min, with the mobile phase starting from 95% solvent A (0.1% TFA in water) and 5% solvent B (0.1% TFA in acetonitrile) (0-2 min), followed by a gradient mobile phase to 5% solvent A and 95% solvent B at 17 min, which was then kept at 95% B until 22 min. The radioactivity was detected by a model of Ludlum 2200 single-channel radiation detector. A semi-preparative C18 reverse, phase column (Phenomenex C18) was used in separations with a 4 mL/min flow rate under gradient conditions.
Production of no carrier added (NCA) [18F]-fluoride ([18F]-F).
The radioisotope 18F (t 1/2 = 110 m) was prepared by the nuclear reaction 180 (p, n) to give 18F in a CTI/Siemens RDS112 l lMev cyclotron. The 180 is in the form of water with an isotopic purity of greater than 95%. The operation of the cyclotron and target functions was automatically controlled by the cyclotron computer system. The target was loaded with the required amount of [180]-water, and bombarded for the appropriate time with a suitable beam current. The target was then unloaded to a collection vial located in the dose calibrator; where the amount of fluoride was measured. The fluoride solution was then transferred to the chemistry operation.
General Fluorination Method
The fluorination (~200 mCi) was performed on an automated synthesis module as shown in Figure 2. As illustrated in the schematic diagram of the module, two-way valves V1-V6 were used to control the solvent and reagent containing reservoirs 1-6. Reservoirs 3-6 are connected with a nitrogen or argon gas line. Reservoir 1 is connected with reactor through several control valves. The reactor is connected with vacuum pump, gas line, and the injection port of the HPLC system. The solutions of potassium carbonate and Kryptofix K2.2.2 (or TBAB and MeCN) were loaded into Reservoirs 1 and 2, respectively. Reservoirs 3, 4, 5, and 6 were filled with precursor solution and other chemicals/solutions as needed. The target water containing 18F was passed through a preconditioned QMA cartridge where the 18F-F" was trapped. The 18F was released from the QMA cartridge by passing K2C03 or TBAB solution from Reservoir 1 through the cartridge and allowed to enter into the reactor. Kyrptofix solution or MeCN from
Reservoir 2 was added into the reactor and the whole mixture was dried at 95°C in combination with nitrogen flow and vacuum . The precursor solution from Reservoir 3 was added to the dried 18F ion and heated at the desired temperature. The reaction mixture will be sampled out for analysis or loaded on HPLC for purification.
3-(4-( -s-tetrazine (5)
Figure imgf000024_0001
F-18 fluoride was dried as described above. Tetrazine 4 was prepared by mesylation of (MsCI, Et3N, CH2CI2) of 3-(4-hydroxymethyl)-phenyl-s-tetrazine, which had been prepared from 4-hydroxymethylbenzonitrile (formamidine acetate, S8, hydrazine hydrate, then NaN02/HOAc), according to the method of S. A. Lang Jr., B. D. Johnson, E. J. Cohen, J. Heterocycl. Chem. 1975, 12, 1143. Compound 4 was dissolved in MeCN and then allowed to react with 18F-TBAF at 85°C for 15 min. The reaction mixture was then analyzed by HPLC. 18F-5 was eluted off at 15.6 min on HPLC, which correlates with the retention time of the standard compound. The labeling yield was estimated to be 1 % (non-decay corrected). Changing the reaction temperature to 110°C or the solvent to DMSO/DMF did not increase the reaction yield .
General procedure for reactions indicated in Table 1.
Figure imgf000025_0001
Fluoride was dried as described above. Precursor 8 was dissolved in MeCN and added to the azeotropically dried fluoride from Reservoir 3. The crude mixture was heated at desired temperature and then analyzed by HPLC. The optimized labeling conditions are described in entry 3 in table 2. In the automated synthesis, the crude reaction mixture was loaded onto the semi-prep HPLC for separation. The purified sample was injected to the analytical HPLC. 18F-9 was eluted off at 17.4 min on HPLC, which correlates with the retention time of the standard compound . Under the optimized conditions, the radiochemical purity of 18F-9 was more than 98%.
General procedure for reactions described in Table 2
HPLC-purified 18F-9 was mixed with 3,6-Di(2-pyridyl)-s-tetrazine ( la) under the conditions described in Table 2. Immediately after mixing, the crude reaction mixture was analyzed by HPLC. The HPLC injection was made within 10 seconds of mixing. 18F- 10 was eluted off at 11.8 min on HPLC, which correlates with the retention time of the standard compound . Small amounts of isomers 18F-11 were also observed on the HPLC radio trace. The retention time of peaks attributable to the isomers of 18F-11 were 12.7 and 13.3 min, which correlates with the retention times for the 19F standards.
Radiochemistry
[18F] Fluoride was prepared by the 180(p,n)18F nuclear reaction, and it was then adsorbed onto an anion exchange resin cartridge. Kryptofix 222/K2C03 solution ( 1 mL 9 : 1 acetonitrile/water, 15 mg Kryptofix 222, 3 mg K2C03) was used to elute the cartridge, and the resulting mixture was dried in a glass reactor. 18F-9 was prepared according to reported procedures and purified via semipreparative HPLC. Labeling of tetrazine-RGD conjugate 14 with 18F-9 was performed in DMSO/EtOH (1 : 3). The resulting mixture was diluted with water and purified by semi -preparative HPLC. The final product 18F-15 was concentrated and formulated in saline (0.9%, 500 μΐ) for in vivo studies.
Cell Line and Animal Models
U87MG human glioblastoma cells were grown in Dulbecco's medium (Gibco) supplemented with 10% fetal bovine serum (FBS), 100 IU/mL penicillin, and 100 pg/mL streptomycin (Invitrogen Co.). Animal procedures were performed according to a protocol approved by the Stanford University Institutional Animal Care and Use
Committee. A U87MG xenograft model was generated by subcutaneous (s.c.) injection of 1 x 107 U87MG cells (integrin av 3-positive) into the front flank of female athymic nude mice. Three to four weeks after inoculation (tumor volume: 100-400 mm3), the mice (about 9-10 weeks old with 20-25 g body weight) were used for microPET studies. Cell Integrin Receptor-Binding Assay
In vitro integrin-binding affinity and specificity of c(RGDyK) and 19F-15 were assessed via competitive cell binding assays using 125I-echistatin as the integrin ανβ3- specific radioligand. The best-fit 50% inhibitory concentration (IC50) values for U87MG cells were calculated by fitting the data with nonlinear regression using GraphPad Prism (GraphPad Software, Inc.). Experiments were performed with triplicate samples.
microPET Studies
PET scans and image analysis were performed using a microPET R4 rodent model scanner (Siemens Medical Solutions). About 2 MBq of 18F-15 was intravenously injected into each mouse {n = 3) under isoflurane anesthesia (1-3%) and then subjected to static scans at 0.5, 1, 2, and 4 h p.i. For each microPET scan, regions of interest (ROIs) were drawn over the tumor, normal tissue, and major organs on decay-corrected whole- body coronal images. The radioactivity concentration (accumulation) within a tumor was obtained from the mean value within the multiple ROIs and then converted to %ID/g. For a receptor-blocking experiment, mice bearing U87MG tumors on the front left flank were scanned after co-injection with 18F-15 and c(RGDyK) (10 mg/kg).
2,5-dioxopyrrolidin-l-yl 5-oxo-5-((6-(6-(pyridin-2-yl)-l,2,4,5-tetrazin-3- yl)pyridin-3-yl)amino)pentanoate (13)
Figure imgf000026_0001
DMF A dry 3 mL vial was sequentially charged with 5-oxo-5-((6-(6-(pyridin-2-yl)- l,2,4,5-tetrazin-3-yl)pyridin-3-yl)amino)pentanoic acid ( 170 mg, 0.46 rnmol), 1,2 N- hydroxysuccinimide (75 mg, 0.65 mmol) and l -ethyl-3-(3- dimethylaminopropyl)carbodiimide hydrochloride (115 mg, 0.60 mmol) . The vial was capped by a screw cap with a Teflon septum. The vial was swept with n itrogen, and anhydrous DMF ( 1.5 mL) was added via syringe. The reaction mixture was allowed to stir for 20 h at room temperature. The mixture was then diluted by CH2CI2 (5 mL), centrifuged, and the supernatant was decanted. The purple solid was subjected to three further cycles of suspension in CH2CI2 (5 mL), centrifugation, and decantation to provide the 130 mg (59%) of the title compound as a purple solid ( 130 mg, 59%) .
JH NMR (400 MHz, DMSO-d6, δ) : 10.66 (s, 1H), 9.06 (d, 2.4 Hz, 1H), 8.94 (m , 1H), 8.64 (d, J = 8.8 Hz, 1H), 8.60 (dt, J = 7.9, 1.0 Hz, 1H), 8.44 (dd, J = 8.8, 2.4 Hz, 1H), 8.17 (dt, J = 7.9, 1.7 Hz, 1H), 7.74 (ddd, J = 7.5, 4.6, 1.0 Hz, 1H), 2.87-2.80 (m, 6H), 2.59 (t, J = 7.6 Hz, 2H), 1.99 (q, J = 7.6 Hz, 2H) ;
13C NMR ( 100 MHz, DMSO-d6, δ) : 172.0(C), 170.8(C), 169.3 (C), 163.5 (C), 163.2
(C), 151.1 (CH), 150.7 (C), 144.4 (C), 141.7 (CH), 138.9 (C), 138.3 (CH), 127.1 (CH), 126.7 (CH), 125.4 (CH), 124.7 (CH), 35.0 (CH2), 30.0 (CH2), 25.9 (CH2), 20.2 (CH2) ;
HRMS-ESI (m/z) : [M-C4H4N03]+(acylium ion) : calcd for Ci7H14N702, 348.1209; found : 348.1202.
Tetrazine-RGD conjugate (14)
A dry 3 mL vial was charged with 13 (5.2 mg, 0.011 mmol) and 12 (3.4 mg, 0.0055 mmol). The vial was capped by a screw cap with a Teflon septum, and the vial was swept with nitrogen. A solution of diisopropylethylamine ( 1.4 mg, 0.011 mmol) in DMF (30 pL) was added via syringe, followed by the addition of DMF (0.57 mL). The reaction mixture was allowed to stir for 18 h at room temperature. The mixture was then diluted by methanol (1 mL), centrifuged, and the supernatant was decanted . The purple solid was subjected to two further cycles of suspension in methanol ( 1 mL), centrifugation, and decantation to provide 14 as a purple solid (5.1 mg, 96%) . The purity was judged to be 95% by HPLC analysis (Shimadzu C18 column, 4.6 mm x 50 mm, 5 micron). HPLC analysis was performed with eluents that contained 0.1% trifluoroacetic acid, eluting at 1 mL/min, initially with 10% acetonitrile/water for 5 min, followed by a gradient of 10%-20% acetonitrile/water for 5 min, and final elution with 20% acetonitrile/water.
HRMS-ESI (m/z) : [M-H]+ : calcd for C44H54 i6O10 +, 967.4282; found : 967.4305. Diels-Alder adduct (18F-15)
A 1.5 mL Eppendorf tube was sequentially charged with tetrazine-RGD conjugate 14 (0.25 mg) in DMSO (0.2 mL) and trans-Cyclooctene 18F-9 (0.1 mg) in acetonitrile (0.1 ml_). The mixture was allowed to stir at room-temperature for 1 min to provide 8F- 15. Purification of the crude product was carried out on an analytical reverse-phase high performance liquid chromatography (HPLC) system equipped with a dual UV absorbance detector (Waters 2487) using a Phenomenex C18 reverse phase column ( 150 x 4.6 mm, 5 micron). .
HPLC analysis was performed with eluents that contained 0.1% trifluoroacetic acid, eluting at 1 mL/min, initially with 5% acetonitrile/water for 2 min, followed by a gradient of 5%-95% acetonitrile/water for 22 min, and final elution with 95%
acetonitrile/water.
The same conditions were used for radio-HPLC analysis of 18F-15. The radioactivity was detected by a model of Ludlum 2200 single-channel radiation detector. Thus, 2 mCi of 18F-9 (Li, Z. ; Cai, H. ; Hassink, M . ; Blackman, M . L. ; Brown, R. C ; Conti, P. S. ; Fox, J. M . Chem Commun. 2010, 46, 8043) in 300 pL EtOH was added to the tetrazine-RGD conjugate 14 (25 +/- 5 pg, 100 pL DMSO) and the mixture was allowed to sit at room temperature for 1 min. The purification of 18F-15 was carried out on an analytical reversed-phase HPLC system using the conditions mentioned above.
HRMS-ESI (m/z) : [M-H] + : calcd for C54H71F 14OH : 1111.5484; found : 1111.5510. Tetrazine-RGD conjugate (18)
Tetrazine-R conjugate 18 was prepared according to the following sequence.
Figure imgf000028_0001
To a stirring solution of 3-nitrobenzhydrazide (1.0 g, 5.52 mmol) and pyridine (0.9 mL) in DMF (1.5 ml_) in a round-bottom flask under N2 was added 4- (trifluoromethyl)benzoyl chloride (1.3 g, 6.07 mmol) dropwise at 0 °C. The reaction mixture was stirred at room temperature for 12 h and then transferred into a beaker containing ice water. The white solid that precipitated out of the solution was isolated by filtration and washed several times with cold H20. The crude solid was purified by recrystallization with acetone to give 1.6 g (4.4 mmol, 80%) of 38 as a white solid, mp 223-225°C. *H NMR (DMSO-d6, 400 MHz, δ) : 11.0 (s, 1H), 10.9 (s, 1H), 8.76 (t, J = 2.2 Hz, 1H), 8.47 (dd, J = 8.3 Hz, 2.4 Hz, 1H), 8.37 (dd, J = 7.9 Hz, 2.4 Hz, 1H), 8.13 (d, J = 8.3 Hz, 2H), 7.94 (d, J = 8.3 Hz, 2H), 7.87 (t, J = 7.6 Hz, 1H). 13C NMR (DMSO-d6, 100 MHz, δ) : 164.7 (u), 163.8 (u), 147.9 (u), 136.1 (u), 133.8 (dn), 133.7 (u), 131.7 (u) [q, 2J(CF) = 35.2 Hz], 130.5 (dn), 128.4 (dn), 126.6 (dn), 125.7 (dn) [q, 3J(CF) = 4.0 Hz], 123.9 (u) [q, XJ(CF) = 272.6 Hz] 122.2 (dn). IR (neat, KBr, cm"1) 3191, 3022, 2847, 1575.1, 1328. HRMS (ESI-) [M-H] calcd. for C15H9F3N3O4 352.0545; found 352.0536.
N'-(chloro(4-(trifluoromethyl)phenyl)methylene)-3-nitrobenzohydrazonoyl chlori
Figure imgf000029_0001
A dry round-bottomed flask was charged with 38 (1.0 g, 2.72 mmol) and the flask was evacuated and filled with nitrogen. Anhydrous dichloroethane (27 mL) and PCI5 (1.7 g, 8.17 mmol) were added to the reaction mixture. The reaction mixture was allowed to reflux and stir for 16 h. The reaction mixture was cooled to room temperature and slowly poured into ice water. The organic and aqueous layers were separated, and the organic layer was washed with saturated IMaHC03, dried over MgS04, filtered and concentrated in vacuo. The crude residue was chromatographed using a gradient (2-8%, then 20%, 40%) of CH2CI2 in hexanes to give 0.804 g (2.00 mmol, 73%) of 37 as a yellow solid, mp 78-80°C. *H NMR (CDCI3, 400 MHz, δ) : 8.93 (t, J = 2.0 Hz, 1H), 8.44 (dd, J = 8.0, 1.9 Hz, 1H), 8.38 (dd, J = 8.3 Hz, 2.3 Hz, 1H), 8.23 (d, J = 8.3 Hz, 2H), 7.71 (d, J = 8.4 Hz, 2H), 7.66 (t, J = 8.1 Hz, 1H). 13C NMR (CDCI3, 100 MHz, δ) : 148.4 (u), 143.6 (u), 142.2 (u), 136.4 (u), 135.1 (u), 133.9 (dn), 133.6 (u) [q, 2J(C-F) = 33.4 Hz], 129.8 (dn), 128.9 (dn), 126.4 (dn) [q, 3J(CF) = 4.0 Hz], 125.6 (dn), 123.6 (u) [q, JJ(CF) = 274.0 Hz] 123.5 (dn). IR (CHCI3, cm"1) 3088, 3059, 1602, 1537, 1326.
Ci5H9CI2F3N302 391.1441 ; found 391.1445. 3-(3-Nitrophenyl)-6-[4-(trifluoromethyl)phenyl]-s-tetrazine (2b)
Figure imgf000030_0001
A dry round-bottom flask was charged with 37 (0.804 g, 2.00 mmol), CH3CN (15 mL) and 64% hydrazine hydrate (0.097 mL, 2.00 mmol). The flask was fitted with a reflux condenser, and the mixture was heated to 50 °C for 1 h behind a blast shield. K2C03 (0.553 g, 4.00 mmol) was added, and the reaction mixture was allowed to reflux and stir for 24 h. 64% hydrazine hydrate (0.291 mL, 6.00 mmol) was added to the mixture, which was allowed to reflux for an additional hour. The reaction mixture was cooled to room temperature. The resulting orange precipitate was isolated by filtration, washed with cold H20, and dried under vacuum. The crude residue was diluted with glacial acetic acid (4.0 mL) at 0°C, and a solution of NaN02 (0.690 g, 10.0 mmol) in H20 (1.1 mL) was added dropwise to the solution. The reaction mixture was diluted with CH2CI2 (100 mL) and washed repeatedly with saturated NaHC03. The organic solution was dried over MgS04, filtered, and concentrated in vacuo. The crude residue was concentrated onto silica gel and chromatographed using a gradient (0-30%) of CH2CI2 in hexanes to give 0.583 g (1.68 mmol, 84%) of 2b as a pink solid, mp 217-219°C. *H NMR (CDCI3, 400 MHz, δ) : 9.54 (t, J = 2.0 Hz, 1H), 9.01 (dd, J - 7.8 Hz, 1.6 Hz, 1H), 8.81 (d, J = 8.3 Hz, 2H), 8.51 (dd, J = 8.3 Hz, 2.3 Hz, 1H), 7.89 (d, J = 8.3 Hz, 2H), 7.84 (t, J = 8.1 Hz, 1H). 13C NMR (CDCI3, 100 MHz, δ) : 164.7 (u), 163.4 (u), 147.9 (u), 136.1 (u), 133.8 (dn), 133.7 (u), 131.5 [u (q, 2J(C-F) = 34.5 Hz)], 130.5 (dn), 126.6 (dn) [q, 3J(CF) = 4.0 Hz], 125.6 (dn), 123.6 (u) [q, XJ(CF) = 271.5 Hz], 122.2 (dn). IR (CHCI3, cm"1) 2932, 2856, 1531, 1324. HRMS (ESI) [M-H] calcd. for C15H8F3N5O2
347.0630; found 347.0622. 3-(3-Aminoph )
Figure imgf000031_0001
To a stirring solution of 2b (0.010 g, 0.029 mmol) in CH3CH2OH (0.3 mL) under N2 was added ammonium formate (0.006 g, 0.093 mmol) and 5% Pd/C (0.004 g, 0.002
5 mmol) . The reaction mixture was allowed to reflux for 20 h then cooled to room
temperature and filtered through celite. The filtrate was concentrated in vacuo, and the crude residue was concentrated onto silica gel and chromatographed using a gradient (0- 6%) of acetone in CH2CI2 to give 0.004 g (0.013 mmol, 44%) of 36 as a red solid, mp 214-216°C. *H NMR (DMSO-d6, 400 MHz, δ) : 8.71 (d, J = 8.3 Hz, 2H), 8.06 (d, J = 8.8 io Hz, 2H), 7.81 (t, J = 2.0 Hz, 1H), 7.71 (m, J = 8.3 Hz, 2.2 Hz, 1H), 7.32 (t, J = 7.4 Hz, 1H), 6.89 (dd, J = 8.5 Hz, 1.9 Hz, 1H), 5.5 (m, 2H). 13C NMR (DMSO-d6, 100 MHz, δ) : 163.7 (u), 162.4 (u), 149.6 (u), 135.9 (u), 132.0 (u), 131.9 [u (q, 2J(C-F) = 34.5 Hz)], 128.2 (dn), 130.0 (dn), 126.3 (dn) [q, 3J(CF) = 4.0 Hz], 121.0 (u) [q, XJ(CF) = 273.2 Hz], 118.2 (dn), 115.2 (dn), 112.4 (dn). IR (CHCI3, cm"1) 3443, 3156. HRMS (ESI) i s [M+H] calcd. for Ci5HuF3N5 318.0967; found 318.0976.
5-oxo-5-(3-(6-(4-(trifluoromethyl)phenyl)-l,2,4,5-tetrazin-3- yl)phenylamino)pentanoic acid (35)
Figure imgf000031_0002
A resealable glass vial was flushed with N 2 and charged with 36 (0.010 g, 0.0320 mmol), glutaric anhydride (0.018 g, 0.158 mmol) and anhydrous THF (0.3 mL). The vial was capped, and the mixture was stirred at 80 °C for 1 h. The reaction mixture was cooled to room temperature, triturated with CH2CI2 and hexanes and dried under vacuum to give 0.010 g (0.023 mmol, 72%) of 35 as a pink solid, mp 246-248°C. *H NMR (DMSO-de, 400 MHz, δ) : 10.3 (s, 1H), 8.92 (t, J = 1.8 Hz, 1H), 8.74 (d, J = 8.2 Hz, 2H), 8.23 (dd, J = 7.8 Hz, 1.8 Hz, 1H), 8.09 (d, J = 8.2 Hz, 2H), 7.92 (dd, J = 8.2 Hz, 2.3 Hz, 1H), 7.63 (t, 1H), 2.43 (t, J = 7.1 Hz, 2H), 2.31 (t, J = 7.4 Hz, 2H), 1.85 (quin., J = 7.0 Hz, 2H). 13C NMR (DMSO-d6, 100 MHz, 6) : 174.3 (u), 171.3 (u), 163.5 (u),
162.6 (u), 140.4 (u), 135.9 (u), 132.0 (u) [q, 2J(C-F) = 32.5 Hz], 132.0 (u), 130.1 (dn), 128.4 (dn), 126.4 (dn) [q, 3J(CF) = 4.0 Hz], 124.2 (u) [q, ^(CF) = 275.1 Hz], 123.1 (dn), 122.6 (dn), 118.0 (dn), 35.5 (u), 33.1 (u), 20.4 (u). IR (neat, KBr, cm"1) 3307, 2952, 1663, 1392. HRMS (ESI) [M + H] calcd. for CzoH^NsC^ 431.3679; found
431.3677.
2,5-dioxopyrrolidin-l-yl 5-oxo-5-((6-(6-(pyridin-2-yl)-l,2,4,5-tetrazin-3- yl
Figure imgf000032_0001
A dry 3 mL vial was sequentially charged with 5-oxo-5-((3-(6-(4- (trifluoromethyl)phenyl)-l,2,4,5-tetrazin-3-yl)phenyl)amino)pentanoic acid (170 mg, 0.46 mmol), /V-hydroxysuccinimide (86 mg, 0.20 mmol) and l-ethyl-3-(3- dimethylaminopropyl)carbodiimide hydrochloride (77 mg, 0.40 mmol). The vial was capped by a screw cap with a Teflon septum. The vial was swept with nitrogen, and anhydrous DMF (1.5 mL) was added via syringe. The reaction mixture was allowed to stir for 34 h at room temperature. The mixture was then diluted by CH2CI2 (10 mL), centrifuged, and the supernatant was decanted. The purple solid was subjected to three further cycles of suspension in CH2CI2 (10 mL), centrifugation, and decantation to provide the 79 mg (75%) of the title compound as a purple solid (79 mg, 75%). !H NMR (400 MHz, DMSO-d6, δ) : 10.35 (s, 1H), 8.92 (t, 1.8 Hz, 1H), 8.74 (d, 8.4Hz ,2H), 8.25 (dt, J = 8.4, 1.7 Hz, 1H), 8.09 (d, J = 8.4, 2H), 7.94 (dt, J = 8.4, 1.7 Hz, 1H), 7.64 (t, J = 8.4 Hz, 1H), 2.83 (s, 4H), 2.81 (t, J = 7.4 Hz, 2H) 2.53 (t, J = 7.4 Hz, 2H), 1.97 (q, J = 7.4 Hz, 2H). Tetrazine-RGD conjugate (18)
Figure imgf000033_0001
A dry 3 mL vial was charged with 34 (10.6 mg, 0.02 mmol) and 12 (6.2 mg, 0.01 mmol). The vial was capped by a screw cap with a Teflon septum, and the vial was swept with nitrogen. A solution of diisopropylethylamine (5.2 mg, 0.04 mmol) in DMF (110 μΙ_) was added via syringe, followed by the addition of DMF (390 μΙ_). The reaction mixture was allowed to stir for 18 h at room temperature. The mixture was centrifuged, and the residue was dissolved in 800 pL of DMSO and purified by reverse phase HPLC to provide 18 as a purple solid (2.2 mg, 11%). LCMS (m/z) [M-H]+ : calcd for
C47H55F3IMi40io+, 1032; found : 1033.
Tetrazine-RGD conjugate (41)
Cyclic peptide c(RGDyC) (39) peptide which targets av 3-integrin on cell surfaces. A tetrazine conjugate 41 was prepared from c(RGDyC) (39) according to the following sequence.
Figure imgf000034_0001
Conjugation of c(RGDyC) (39) and tetrazine-maleimide 24 to form 40
The conjugation followed the method of Cai W, Zhang X, Wu Y, Chen X, employing a thiol-reactive 18F-labeling agent, l\l-[2-(4-18F- fluorobenzamido)ethyl]maleimide, and synthesis of RGD peptide-based tracer for PET imaging of alpha v beta 3 integrin expression. J Nucl Med. 2006;47 : 1172-1180. Briefly, tetrazine-maleimide (200 pg, 0.41 pmol) in 100 pL dimethyl sulfoxide (DMSO) and 39 (200 pg, 0.33 pmol) (Peptides International of Louisville, KY) in 500 pL phosphate buffer (50 mM, pH 6.5-7.0) were mixed together at room temperature. After the mixture was stirred at room temperature for 5 h, the conjugate was purified by semipreparative HPLC. The collected fractions were combined and lyophilized to afford the final product as a white powder. Compound 40 was obtained in 85% yield.
Conjugation of VEGF and tetrazine-maleimide to form 42
Briefly, tetrazine-maleimide 24 (200 pg, 0.41 pmol) in 100 pL DMSO and VEGF (100 pg, 5.5 nmol) in 500 pL phosphate buffer (50 mM, pH 6.5-7.0) were mixed together at room temperature. After the mixture was stirred at room temperature for 5 h, the conjugate was purified by size exclusion PD-10 column and concentrated by Centricon filter (Millipore, Bedford, MA), and the final concentration was determined based on UV absorbance at 280 nm using unconjugated VEGF of known concentrations as standard. The final concentration was adjusted to 50 pg/mL for use.
Synthesis of 19F-41
19F-9 was synthesized according to Li Z, Cai H, Hassink M, Blackman ML, Brown RC, Conti PS, et al. Tetrazine-trans-cyclooctene ligation for the rapid construction of 18F labeled probes. Chem Commun (Camb). 2010;46 : 8043-8045. 40 (100 pg, 92 nmol) in 100 pL DMSO and 19F-9 (200 pg, 0.33 μητιοΙ) in 100 pL DMSO were mixed together at room temperature. After the mixture was stirred at room temperature for 5 min, the conjugate was purified by semipreparative HPLC. The collected fractions were combined and lyophilized to afford the final product as a white powder. 19F-41 was obtained in 92% yield with 15.5 min retention time on analytical HPLC. MALDI-TOF-MS was m/z 1226.4 for [MH]+ (C57H73FN15O13S, calculated molecular weight 1226.5).
Synthesis of 18F-41
18F-9 was synthesized according to Li Z, Cai H, Hassink M, Blackman ML, Brown RC, Conti PS, et al. Tetrazine-trans-cyclooctene ligation for the rapid construction of 18F labeled probes. Chem Commun (Camb). 2010;46:8043-8045. 18F-9 (148 MBq, 4 mCi) in about 50 pL ethanol was added to 40 (10 pg) in 50 pL DMSO followed by shaking for 5 min. The conjugate was purified by semipreparative HPLC. The collected fractions were combined and the solvent was removed by rotary evaporation under reduced pressure. 18F-41 was reconstituted in 1 mL PBS and passed through a 0.22 pm syringe filter for in vivo animal experiments.
Synthesis of 18F-43
18F-9 (148 MBq, 4 mCi) in about 50 pL ethanol was added to the 42 (10 pg) in water followed by shaking for 5 min. The conjugate was purified by PD-10 column using l BS as eluent. The collected fractions and passed through a 0.22 pm syringe filter for in vivo animal experiments.
Cell Culture
Human glioblastoma cell line U87MG cells were obtained from the American Type Culture Collection (Manassas, VA) and were cultured in DMEM containing high glucose (GIBCO, Carlsbad, CA), which was supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin. The cells were expanded in tissue culture dishes and kept in a humidified atmosphere of 5% C02 at 37°C. The medium was changed every other day. A confluent monolayer was detached with 0.05% Trypsin-EDTA, 0.01M PBS (pH 7.4) and dissociated into a single-cell suspension for further cell culture.
MicroPET Imaging
Animal procedures were performed according to a protocol approved by the University of Southern California Institutional Animal Care and Use Committee. For static microPET scans, mice bearing U87MG xenografts were injected with 3.7 MBq (100 pCi) of 18F-41 via the tail vein (n = 3 for each group). At 0.5, 1, and 2 h post injection (p.i.), the mice were anesthetized with isoflurane (5% for induction and 2% for maintenance in 100% 02) using a knock-down box. With the help of a laser beam 5 attached to the scanner, the mice were placed in the prone position and near the center of the field of view of the scanner. The 3-min static scans were then obtained. Images were reconstructed using a 2-dimensional ordered -subsets expectation maximization (OSEM) algorithm. No background correction was performed. Regions of interest (ROIs; 5 pixels for coronal and transaxial slices) were drawn over the tumor on decay-corrected l o whole-body coronal images. The maximum counts per pixel per minute were obtained from the ROI and converted to counts per milliliter per minute using a calibration constant. With the assumption of a tissue density of 1 g/mL, the ROIs were converted to counts per gram per min. Image ROI-derived %ID/g values were determined by dividing counts per gram per minute with injected dose. No attenuation correction was
is performed. Similarly, 18F-43 was injected into healthy nude mice for microPET study.
Chemistry and Radiochemistry
The conjugation of tetrazine-maleimide with 39 and VEGF proceeded smoothly in the phosphate buffer with pH 6.5-7.0. The nonradioactive cyclization reaction of 40 and 19F-9 was performed and the product was used for characterization and as a standard for0 18F labeled product. Right after addition of 19F-9 to the solution of 40, the pink color of
40 disappeared, indicating completion of the reaction. The identity of the 19F-41 was confirmed by MALDI-TOF-MS. 18F-9 was produced using the protocol described in Li Z, Cai H, Hassink M, Blackman ML, Brown RC, Conti PS, et al. Tetrazine-trans-cyclooctene ligation for the rapid construction of 18F labeled probes. Chem Commun (Camb). 5 2010;46:8043-8045. The radiolabeling yields for 18F-41 and 18F-43 showed almost quantitative capture of 18F-9. The radiopurity of 18F-41 was more than 95%, determined by radio HPLC. The specific activity of 18F-41 and 18F-43 were estimated to be about 3-6 Ci/μιτιοΙ.
MicroPET Imaging
0 Representative coronal microPET images of U87MG tumor-bearing mice (n =
3/group) were obtained at different times after intravenous injection of about 3.7 MBq (100 μθ) of 18F-41. The tumors after injection of the radiotracers were all clearly visible with good contrast to contralateral background at all time points measured. The mice showed high abdominal activity accumulation. Prominent uptake of 18F-41 was
5 observed in the kidneys and urinary bladder at early time points, indicating that this radiotracer is mainly excreted through the renal system. Quantification of tumor and major organ activity accumulation in the microPET scans was achieved by measuring regions of interest (ROIs) encompassing the entire organ on the coronal images. Tumor and major organ uptake of 18F-41 is depicted in Figure 3A. The tumor uptake of 18F-41 in U87MG tumor was 1.98 ± 0.33, 1.80 ± 0.15, and 1.27 ± 0.33 % ID/g at 0.5, 1, and 2 h p.i., respectively. The uptake of the 18F-41 decreased rapidly with time in the muscle, 5 affording better image contrast at later time points after injection (2 h).
The biodistribution of 18F-43 was evaluated in normal Sprague Dawley nude mice. Representative coronal microPET images at 0.5 and 3 h post injection of about 3.7 MBq (100 Ci) 18F-43 were obtained. The activity was mainly accumulated in the liver and kidney. MicroPET quantification by measuring the ROI is shown in Figure 3B. The 10 kidney uptakes of 18F-43 were 23.20 ±2.15 and 16.51 ± 1.52 %ID/g at 30 min and 180 min post injection respectively. Low muscle uptakes were observed (2.98 ± 0.66 and 1.59 ± 0.57 %ID/g at 0.5 and 3 h post injection).
Diaminotetrazine (21)
Diaminotetrazine 21 was re ared as follows.
Figure imgf000037_0001
21 i s
XH NMR (400 MHz, DMSO-c?6, δ) 21 : 8.27 (d, J = 8.8 Hz, 2H), 8.20 (d, J = 2.3 Hz ,2H), 7.10 (dd, J = 8.8, 2.3 Hz, 2H), 6.24 (s, 4H).
N1-(2-(2 5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)ethyl)-N5-(6-(6-(pyridin-2-yl)- l,2,4,5-tetrazin-3-yl)pyridin-3-yl)glutaramide (24)
0 Compound 24 was prepared according to the following sequence.
Figure imgf000037_0002
To a flame dried flask 1.78 g (8.52 mmol) of 4-(2-hydroxyethyl)-10-oxa-4-aza- tricyclo[5.2.1.02,6]-dec-8-ene-3,5-dione (Willson, C. G. Macromolecules 2008, 41, 719) was added. The flask was equipped with a condenser. The flask was then evacuated5 and refilled with nitrogen. Toluene (36 ml_) was added. The reaction flask was heated to 110°C and stirred and sparged with nitrogen for 5 hr. The flask was then cooled to 0°C The mixture was filtered in a Buchner funnel and washed with diethyl ether (20 mL). The collected solid was then sublimed and collected to yield 28% (0.339 g, 2.40 mmol) of l-(2-hydroxyethyl)-l V-pyrrole-2,5-dione.
:H NMR (400 MHz, CD3CI) 6.74 (s, 2H), 3.82-3.77 (m, 2H), 3.74-3.70 (m, 2H), 2.03 (brs, 1H)
Figure imgf000038_0001
To a flame dried round bottom flask 0.5 g (3.6 mmol) of l-(2-hydroxyethyl)-lW- pyrrole-2,5-dione and 0.7 g (4.0 mmol) of methanesulfonic anhydride was added. The flask was evacuated and refilled with nitrogen. The flask was heated to 105°C and stirred for 3.5 hr. The reaction was cooled to room temperature and dissolved in ethyl acetate (5 mL). The solution was taken up into a separatory funnel and washed four times with a saturated solution of sodium carbonate (40 mL). The aqueous layers were collected and washed with ethyl acetate (50 mL). The organic layers were combined and dried with MgS04 and concentrated in vacuo. The product was recrystallized from t-butyl methyl ether to yield 43 % (0.347 g, 1.58 mmol) of 2-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)ethyl methanesulfonate.
XH NMR (400 MHz, CD3CI) 6.78 (s, 2H), 4.41 (t, J = 5.23, 2H), 3.90 (t, J = 5.23, 2H), 3.04 (s, 3H)
N1-(2-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)ethyl)-N5-(6-(6-(pyridin-2-yl)-
Figure imgf000038_0002
To a flame dried flask was added 0.03 g (0.065 mmol, 1 eq.) of 2,5- dioxopyrrolidin-l-yl 5-oxo-5-((6-(6-(pyridin-2-yl)-l,2,4,5-tetrazin-3-yl)pyridin-3- yl)amino)pentanoate (13), followed by 0.02 g (0.078 mmol, 1.2 eq.) of 2-(2,5-dioxo- 2,5-dihydro-lH-pyrrol-l-yl)ethanaminium trifluoroacetate. The flask was evacuated and then refilled with nitrogen. DMF (1 ml) was added and the mixture was stirred.
Diisopropylethyl amine (0.032 ml, 0.195 mmol, 3 eq.) was added dropwise to the mixture. The reaction mixture was stirred for 24 hours at room temperature. The reaction was condensed in vacuo and then loaded onto a silica gel column using the minimum amount of MeOH required along with CH2CI2. The column was then run using a gradient of 0-10% MeOH in CH2CI2 to yield 48% (0.015 g, 0.031 mmol) of N1-(2-(2,5- dioxo-2,5-dihydro-lH-pyrrol- l-yl)ethyl)-N5-(6-(6-(pyridin-2-yl)- l,2,4,5-tetrazin-3- yl)pyridin-3-yl)glutaramide.
H NMR (400 MHz, CD3OD) δ 9.06 (d, J = 2.3 Hz, 1H), 8.92-8.88 (m, 1H), 8.82- 8.75 (m, 2H), 8.51 (dd, J = 8.8, 2.6 Hz, 1H), 8.19 (dt, J = 1.7, 7.8 Hz, 1H), 7.80 - 7.69 (m, 1H), 6.84 (s, 2H), 5.51 (s, 1H), 4.60 (s, 2H), 3.72 - 3.56 (m, 2H), 3.42-3.36 (m, 2H), 2.68 (s, 1H), 2.53 (t, J = 7.3 Hz, 2H), 2.27 (t, J - 7.4 Hz, 2H), 2.08 - 1.90 (m, 2H).
Although the invention is illustrated and described herein with reference to specific embodiments, the invention is not intended to be limited to the details shown. Rather, various modifications may be made in the details within the scope and range of equivalents of the claims without departing from the invention.

Claims

What is Claimed :
1. A Diels-Alder adduct of a trans-cyclooctene with a tetrazine, wherein the adduct bears a substituent labeled with a radionuclide .
2. The Diels-Alder adduct of claim 1, wherein the substituent is a substituent on the trans-cyclooctene residue of the Diels-Alder adduct.
3. The Diels-Alder adduct of claim 1, wherein the substituent is a substituent on the tetrazine residue of the Diels-Alder adduct.
4. The Diels-Alder adduct of any preceding claim, wherein the Diels-Alder adduct is covalently bound to a biomolecule.
5. The Diels-Alder adduct of claim 4, wherein the biomolecule is in an animal or human.
6. The Diels-Alder adduct of claim 4 or 5, wherein the frans-cyclooctene residue of the Diels-Alder adduct is covalently bound to the biomolecule.
7. The Diels-Alder adduct of claim 6, wherein the frans-cyclooctene residue is a residue formed by reaction of the biomolecule with a compound according to any one of structures 25, 26 or 27
Figure imgf000040_0001
8. The Diels-Alder adduct of claim 6 or 7, wherein the substituent is on the tetrazine residue of the Diels-Alder adduct and wherein the radionuclide is 18F.
9. The Diels-Alder adduct of claim 8, wherein the tetrazine residue is a residue of the compou nd accord i ng to structure 5
Figure imgf000041_0001
5
10. The Diels-Alder adduct of claim 4, wherein the tetrazine residue of the Diels-Alder adduct is covalently bou nd to the biomolecu le.
11. The Diels-Alder adduct of claim 10, wherein the tetrazine residue is a residue formed by reaction of the biomolecule with a compou nd accord i ng to structu re 13
Figure imgf000041_0002
12. The Diels-Alder adduct of claim 10, wherein the tetrazine residue is a residue formed by reaction of the biomolecu le with a compound according to structure 24
Figure imgf000041_0003
13. The Diels-Alder adduct of any one of claims 10- 12, wherein the substituent is on the trans-cyclooctene residue of the Diels-Alder adduct and wherein the
rad ionuclide is 18F.
14. The Diels-Alder adduct of claim 13, wherein the frans-cyclooctene residue is a res und according to structure 9
Figure imgf000042_0001
15. The Diels-Alder adduct of claim 13, wherein the frans-cyclooctene residue is a re d according to structure 17
Figure imgf000042_0002
16. A method of producing a PET or other image of an organ in an animal or human, comprising forming in the animal or human a Diels-Alder adduct of a trans- cyclooctene with a tetrazine, wherein the adduct bears a substituent labeled with a radionuclide.
17. The method of claim 16, wherein the Diels-Alder adduct is covalently bound to a biomolecule.
18. The method of claim 16 or 17, wherein the radionuclide is 18F.
The compound according to structure 5
Figure imgf000042_0003
5
20. The compound according to structure 9
Figure imgf000042_0004
The compound according to structure 17
Figure imgf000043_0001
The compound according to structure 13
Figure imgf000043_0002
23. The compound according to structur
structure 25 structure 26
Figure imgf000043_0003
Figure imgf000044_0001
PCT/US2011/044814 2010-07-23 2011-07-21 Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes WO2012012612A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP11810394.4A EP2595967B1 (en) 2010-07-23 2011-07-21 Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes
US13/811,568 US10434197B2 (en) 2010-07-23 2011-07-21 Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes
CN201180041210.XA CN103221398B (en) 2010-07-23 2011-07-21 For the tetrazine-trans cyclooctene ligation of rapid build radioisotope labeling probe

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US36717410P 2010-07-23 2010-07-23
US61/367,174 2010-07-23

Publications (2)

Publication Number Publication Date
WO2012012612A2 true WO2012012612A2 (en) 2012-01-26
WO2012012612A3 WO2012012612A3 (en) 2012-05-24

Family

ID=45497458

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/044814 WO2012012612A2 (en) 2010-07-23 2011-07-21 Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes

Country Status (4)

Country Link
US (1) US10434197B2 (en)
EP (1) EP2595967B1 (en)
CN (1) CN103221398B (en)
WO (1) WO2012012612A2 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014081301A1 (en) * 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Bio-orthogonal drug activation
WO2014081303A1 (en) * 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Chemically cleavable group
WO2014117001A1 (en) * 2013-01-25 2014-07-31 Memorial Sloan-Kettering Cancer Center Tetrazines/trans-cyclooctenes in solid phase synthesis of labeled peptides
WO2014205126A1 (en) 2013-06-19 2014-12-24 The Regents Of The University Of California Chemical structures for localized delivery of therapeutic agents
WO2014182704A3 (en) * 2013-05-06 2015-01-15 The General Hospital Corporation Bioorthogonal turn-on probes
EP3116486A4 (en) * 2014-03-14 2017-11-08 Daniel J. Capon Hybrid immunoglobulin containing non-peptidyl linkage
US10130723B2 (en) 2014-03-14 2018-11-20 The Regents Of The University Of California TCO conjugates and methods for delivery of therapeutic agents
WO2018234488A1 (en) 2017-06-23 2018-12-27 Basf Se Substituted cyclopropyl derivatives
EP3389729A4 (en) * 2015-12-15 2019-12-11 United Kingdom Research and Innovation Conformationally strained trans-cycloalkenes for radiolabeling
WO2020108720A1 (en) 2018-11-30 2020-06-04 Københavns Universitet Novel tetrazine compounds for in vivo imaging
US10828373B2 (en) 2015-09-10 2020-11-10 Tambo, Inc. Bioorthogonal compositions
WO2020239687A1 (en) 2019-05-24 2020-12-03 Rigshospitalet 18f-labeled novel tetrazines imaging probes for pretargeting in pet imaging
US11220556B2 (en) 2013-03-15 2022-01-11 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US11253600B2 (en) 2017-04-07 2022-02-22 Tambo, Inc. Bioorthogonal compositions
EP4056546A1 (en) 2021-03-11 2022-09-14 University of Copenhagen Aliphatic 18f-radiolabeling of a tetrazine precursor
WO2022189304A2 (en) 2021-03-11 2022-09-15 University Of Copenhagen Aliphatic 18f-radiolabeling of a tetrazine precursor
CN115974892A (en) * 2022-12-27 2023-04-18 四川大学华西医院 Triazole tetrazine compound and preparation method and application thereof
US11857636B2 (en) 2011-05-16 2024-01-02 Tagworks Pharmaceuticals B.V. Bio-orthogonal drug activation
WO2024130105A1 (en) * 2022-12-16 2024-06-20 Biogen Ma Inc. Cyclooctene compositions and uses thereof

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2522369A1 (en) * 2011-05-09 2012-11-14 Koninklijke Philips Electronics N.V. Pretargeting kit, method and agents used therein
US10556024B2 (en) 2013-11-13 2020-02-11 Whitehead Institute For Biomedical Research 18F labeling of proteins using sortases
CN106715453B (en) 2014-03-24 2021-04-30 哥伦比亚大学董事会 Chemical process for producing tagged nucleotides
WO2016025480A1 (en) 2014-08-11 2016-02-18 The General Hospital Corporation Cyclooctenes for bioorthogonol reactions
WO2017059397A1 (en) 2015-10-01 2017-04-06 Whitehead Institute For Biomedical Research Labeling of antibodies
WO2018172543A1 (en) * 2017-03-24 2018-09-27 Centre National De La Recherche Scientifique Biocompatible modular tetrazine platform
AU2018388467A1 (en) 2017-12-18 2020-06-11 Janssen Biotech, Inc. Radiolabeling of polypeptides
MA52363A (en) 2018-04-26 2021-03-03 Agenus Inc THERMAL SHOCK PROTEIN (HSP) PEPTIDIC COMPOSITIONS AND THEIR METHODS OF USE
AU2020273654A1 (en) 2019-05-10 2021-11-25 Janssen Biotech, Inc. Macrocyclic chelators and methods of use thereof
CN110954514B (en) * 2019-12-02 2022-06-21 上海交通大学 Fluorescent tracing system and method suitable for researching in-vivo and in-vitro distribution of liposome
CN111138337B (en) * 2019-12-26 2021-06-22 清华大学 Bicyclo [4.1.0] heptane nitrosourea derivatives for bioorthogonal reaction and preparation method and application thereof
JP2023526102A (en) 2020-05-20 2023-06-20 ヤンセン バイオテツク,インコーポレーテツド Site-specific conjugation of glycosylated monoclonal antibodies with transglutaminase
US20220143231A1 (en) 2020-11-10 2022-05-12 Janssen Biotech, Inc. Macrocyclic compounds and methods of use thereof
CN113248408B (en) * 2021-04-30 2022-03-04 南京大学 Multi-modal molecular imaging probe P-FFGd-TCO and preparation method and application thereof
CA3218089A1 (en) 2021-05-07 2022-11-10 Xiao Wang Spatiotemporally resolved transcriptomics at subcellular resolution
IL311070A (en) 2021-08-27 2024-04-01 Janssen Biotech Inc Anti-psma antibodies and uses thereof
CA3236851A1 (en) 2021-11-09 2023-05-19 Janssen Biotech, Inc. Macrocyclic compounds and diagnostic uses thereof
CA3240194A1 (en) 2021-11-09 2023-05-19 Edward Cleator Macrocyclic compounds and methods of making the same
CN114380813B (en) * 2022-01-17 2023-06-06 厦门大学 VISTA targeted probe and preparation method and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4053528A (en) * 1973-12-28 1977-10-11 Hooker Chemicals & Plastics Corporation Process for the preparation of diels-alder adducts of halogenated cyclopentadienes
GB0428012D0 (en) * 2004-12-22 2005-01-26 Hammersmith Imanet Ltd Radiolabelling methods
US8236949B2 (en) 2007-07-17 2012-08-07 University Of Delaware Tetrazine-based bio-orthogonal coupling reagents and methods
CN101723849B (en) * 2008-10-10 2012-11-21 北京师范大学 Novel 18F labeled amino acid derivatives, preparation method and application thereof in tumor imaging
EP2361098B1 (en) * 2008-10-31 2019-08-28 The General Hospital Corporation Compositions and methods for delivering a substance to a biological target

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
M. L. BLACKMAN; M. ROYZEN; J. M. FOX, J. AM. CHEM. SOC., vol. 130, 2008, pages 13518 - 13519
M. ROYZEN; G. P. A. YAP; J. M. FOX, J. AM. CHEM. SOC., vol. 130, 2008, pages 3760 - 3761
R. ROSSIN ET AL., ANGEW. CHEM. INT. ED., vol. 49, 2010, pages 3375 - 3378
S. GUHLKE ET AL., NUCL. MED. BIOL., vol. 21, no. 6, 1994, pages 819 - 825
See also references of EP2595967A4

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11857636B2 (en) 2011-05-16 2024-01-02 Tagworks Pharmaceuticals B.V. Bio-orthogonal drug activation
EP4245744A3 (en) * 2012-11-22 2024-05-01 Tagworks Pharmaceuticals B.V. Chemically cleavable group
WO2014081303A1 (en) * 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Chemically cleavable group
US10927139B2 (en) 2012-11-22 2021-02-23 Tagworks Pharmaceuticals B.V. Chemically cleavable group
WO2014081301A1 (en) * 2012-11-22 2014-05-30 Tagworks Pharmaceuticals B.V. Bio-orthogonal drug activation
WO2014117001A1 (en) * 2013-01-25 2014-07-31 Memorial Sloan-Kettering Cancer Center Tetrazines/trans-cyclooctenes in solid phase synthesis of labeled peptides
US11220556B2 (en) 2013-03-15 2022-01-11 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
WO2014182704A3 (en) * 2013-05-06 2015-01-15 The General Hospital Corporation Bioorthogonal turn-on probes
US10517965B2 (en) 2013-05-06 2019-12-31 The General Hospital Corporation Bioorthogonal turn-on probes
US10130711B2 (en) 2013-06-19 2018-11-20 The Regents Of The University Of California Chemical structures for localized delivery of therapeutic agents
WO2014205126A1 (en) 2013-06-19 2014-12-24 The Regents Of The University Of California Chemical structures for localized delivery of therapeutic agents
US10953098B2 (en) 2013-06-19 2021-03-23 The Regents Of The University Of California Chemical structures for localized delivery of therapeutic agents
US10130723B2 (en) 2014-03-14 2018-11-20 The Regents Of The University Of California TCO conjugates and methods for delivery of therapeutic agents
US10342882B2 (en) 2014-03-14 2019-07-09 The Regents Of The University Of California TCO conjugates and methods for delivery of therapeutic agents
EP4169944A1 (en) * 2014-03-14 2023-04-26 Biomolecular Holdings LLC Process for preparing hybrid immunoglobulin containing non-peptidyl linkage
US10806807B2 (en) 2014-03-14 2020-10-20 The Regents Of The University Of California TCO conjugates and methods for delivery of therapeutic agents
EP3116486A4 (en) * 2014-03-14 2017-11-08 Daniel J. Capon Hybrid immunoglobulin containing non-peptidyl linkage
US11066459B2 (en) 2014-03-14 2021-07-20 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
US10828373B2 (en) 2015-09-10 2020-11-10 Tambo, Inc. Bioorthogonal compositions
EP3389729A4 (en) * 2015-12-15 2019-12-11 United Kingdom Research and Innovation Conformationally strained trans-cycloalkenes for radiolabeling
US11253600B2 (en) 2017-04-07 2022-02-22 Tambo, Inc. Bioorthogonal compositions
WO2018234488A1 (en) 2017-06-23 2018-12-27 Basf Se Substituted cyclopropyl derivatives
WO2020108720A1 (en) 2018-11-30 2020-06-04 Københavns Universitet Novel tetrazine compounds for in vivo imaging
WO2020239687A1 (en) 2019-05-24 2020-12-03 Rigshospitalet 18f-labeled novel tetrazines imaging probes for pretargeting in pet imaging
EP4056546A1 (en) 2021-03-11 2022-09-14 University of Copenhagen Aliphatic 18f-radiolabeling of a tetrazine precursor
WO2022189304A2 (en) 2021-03-11 2022-09-15 University Of Copenhagen Aliphatic 18f-radiolabeling of a tetrazine precursor
WO2024130105A1 (en) * 2022-12-16 2024-06-20 Biogen Ma Inc. Cyclooctene compositions and uses thereof
CN115974892A (en) * 2022-12-27 2023-04-18 四川大学华西医院 Triazole tetrazine compound and preparation method and application thereof
CN115974892B (en) * 2022-12-27 2023-09-08 四川大学华西医院 Triazole tetrazole compound, and preparation method and application thereof

Also Published As

Publication number Publication date
EP2595967B1 (en) 2016-03-23
EP2595967A4 (en) 2013-12-04
CN103221398A (en) 2013-07-24
US10434197B2 (en) 2019-10-08
WO2012012612A3 (en) 2012-05-24
US20130266512A1 (en) 2013-10-10
CN103221398B (en) 2016-03-23
EP2595967A2 (en) 2013-05-29

Similar Documents

Publication Publication Date Title
US10434197B2 (en) Tetrazine-trans-cyclooctene ligation for the rapid construction of radionuclide labeled probes
Boros et al. RGD conjugates of the H2dedpa scaffold: synthesis, labeling and imaging with 68Ga
Decristoforo et al. 68 Ga-and 111 In-labelled DOTA-RGD peptides for imaging of αvβ3 integrin expression
Price et al. H 6 phospa-trastuzumab: bifunctional methylenephosphonate-based chelator with 89 Zr, 111 In and 177 Lu
ES2547712T3 (en) Methods and compositions for protein labeling
Läppchen et al. DOTA-tetrazine probes with modified linkers for tumor pretargeting
Gasser et al. Synthesis, copper (II) complexation, 64Cu-labeling, and bioconjugation of a new bis (2-pyridylmethyl) derivative of 1, 4, 7-triazacyclononane
Zhou et al. An efficient method for labeling single domain antibody fragments with 18F using tetrazine-trans-cyclooctene ligation and a renal brush border enzyme-cleavable linker
US20200268913A1 (en) Radioactive drug
AU2021202665A1 (en) Radio-pharmaceutical complexes
Liu et al. A new 18 F-heteroaryltrifluoroborate radio-prosthetic with greatly enhanced stability that is labelled by 18 F–19 F-isotope exchange in good yield at high specific activity
US20130343990A1 (en) Radiolabelled octreotate analogues as pet tracers
Gotzmann et al. Synthesis, 64 Cu-labeling and PET imaging of 1, 4, 7-triazacyclononane derived chelators with pendant azaheterocyclic arms
Al Jammaz et al. Novel synthesis and preclinical evaluation of folic acid derivatives labeled with 18F-[FDG] for PET imaging of folate receptor-positive tumors
Fernández et al. Synthesis, in vitro and in vivo characterization of two novel 68Ga-labelled 5-nitroimidazole derivatives as potential agents for imaging hypoxia
Ruivo et al. Improved stability of a novel fluorine-18 labeled TCO analogue for pretargeted PET imaging
Choi et al. N-Succinimidyl guanidinomethyl iodobenzoate protein radiohalogenation agents: influence of isomeric substitution on radiolabeling and target cell residualization
EP4043041A1 (en) Cxcr4-ligands for diagnostic and therapeutic use and precursors thereof
Vaidyanathan et al. SIB-DOTA: A trifunctional prosthetic group potentially amenable for multi-modal labeling that enhances tumor uptake of internalizing monoclonal antibodies
Singh et al. A homodimeric bivalent radioligand derived from 1-(2-methoxyphenyl) piperazine with high affinity for in vivo 5-HT1A receptor imaging
Nakashima et al. Development of novel trifunctional chelating agents that enhance tumor retention of radioimmunoconjugates
EP3554559B1 (en) Method for labeling of sensitive and thermosensitive targeting biomolecules with technetium based compounds
US20200188540A1 (en) Conformationally strained trans-cycloalkenes for radiolabeling
US20240051976A1 (en) A Tracer Compound and a Preparation Method Thereof
Leier Chemoselective bioconjugation reactions of tyrosine residues for application in PET radiochemistry

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180041210.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11810394

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011810394

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13811568

Country of ref document: US