Skip to main content
  • Research article
  • Open access
  • Published:

Improved automated one-pot two-step radiosynthesis of (S)-[18F]FETrp, a radiotracer for PET imaging of indoleamine 2,3-dioxygenase 1 (IDO1)

Abstract

Background

(S)-[18F]FETrp is a promising PET radiotracer for imaging IDO1 activity, one of the main enzymes involved in the tryptophan metabolism that plays a key role in several diseases including cancers. To date, the radiosynthesis of this tryptophan analogue remains highly challenging due to partial racemization occurring during the nucleophilic radiofluorination step. This work aims to develop a short, epimerization-free and efficient automated procedure of (S)-[18F]FETrp from a corresponding enantiopure tosylate precursor.

Results

Enantiomerically pure (S)- and (R)-FETrp references as well as tosylate precursors (S)- and (R)-3 were obtained from corresponding Na-Boc-(L and D)-tryptophan in 2 and 4 steps, respectively. Manual optimisation of the radiolabelling conditions resulted in > 90% radiochemical conversion with more than 99% enantiomeric purity. Based on these results, the (S)-[18F]FETrp radiosynthesis was fully automated on a SynChrom R&D EVOI module to produce the radiotracer in 55.2 ± 7.5% radiochemical yield, 99.9% radiochemical purity, 99.1 ± 0.5% enantiomeric excess, and molar activity of 53.2 ± 9.3 GBq/µmol (n = 3).

Conclusions

To avoid racemisation and complicated purification processes, currently encountered for the radiosynthesis of (S)-[18F]FETrp, we report herein significant improvements, including a versatile synthesis of enantiomerically pure tosylate precursor and reference compound and a convenient one-pot two-step automated procedure for the radiosynthesis of (S)-[18F]FETrp. This optimised and robust production method could facilitate further investigations of this relevant PET radiotracer for imaging IDO1 activity.

Background

Cancer immunotherapy based on immune checkpoint (IC) blockade has made impressive progress in recent years thanks to a better understanding of tumour immune escape mechanisms (Waldman et al. 2020; Guo et al. 2023). In particular, IC inhibitors (ICIs) targeting the programmed cell death protein-1/ligand-1 (PD-1/PDL-1) axis or cytotoxic T lymphocyte antigen 4 (CTLA-4), alone or in combination with further targeted- or chemo-therapies, have become the first-line treatments of numerous cancers such as advanced melanoma (Michielin et al. 2019), metastatic colorectal cancer (Morris et al. 2023), head and neck squamous cell carcinoma (Daste et al. 2024) and non-small cell lung cancer (Cortiula et al. 2022). Based on these promising therapeutic results, a broad range of ICIs targeting several immunosuppressive pathways are being developed or undergo clinical trials (Liu et al. 2022).

Indoleamine 2,3-dioxygenase 1 (IDO1) is an intracellular heme protein that catalyses along with two other enzymes, IDO2 and tryptophan 2,3-dioxygenase 2 (TDO2), the first and rate-limiting step of the tryptophan catabolism known as the kynurenine pathway (Badawy 2017). IDO1 is considered as an immune regulator that plays a pivotal role in numerous diseases such as central nervous system disorders, infections, autoimmune and inflammatory diseases, and cancers (Platten et al. 2019). Aberrant activation of IDO1 by cells located in tumours or found in the tumour microenvironment (TME) (e.g. dendritic cells, cancer-associated fibroblasts, and macrophages) is known to contribute to cancer immune escape through the combined effect of tryptophan depletion and accumulation of kynurenine and its metabolites in the tumour region (Cheong and Sun 2018). Moreover, high-level expression of IDO1 was found in a large variety of cancer types and correlated with tumour progression and poor prognosis of patients (Godin-Ethier et al. 2011). Based on these findings, IDO1 inhibitors have been designed and extensively studied alone or in combination with others IC drugs for solid tumours treatment (Le Naour et al. 2020).

However, such immunotherapies often induce adverse effects (Ibis et al. 2023), emergence of resistance (Guo et al. 2023; Pang et al. 2023) and/or low response rates (Haslam and Prasad 2019). Therefore, there is an urgent need to decipher the compensatory mechanisms that contribute to cancer resistance after IC immunotherapy (Charehjoo et al. 2023) and to identify relevant tools to improve the selection of eligible patients and/or predict the treatment response (Pilard et al. 2021). In addition and complementary to tissue and blood biomarkers under investigation (Pilard et al. 2021; Sankar et al. 2022), positron emission tomography (PET) imaging of IC and other key components of the TME are being studied intensively (Wierstra et al. 2019; van de Donk et al. 2020; Bouleau et al. 2021; Ge et al. 2022). Indeed, this non-invasive and quantitative molecular imaging methodology, allows the whole-body and repetitive assessment of the tumour and TME uptakes of specific and selective radiotracers that may provide valuable information regarding the spatiotemporal expression of IC before and during treatment, which in turn may guide the stratification of patients, predict the treatment response, thereby helping developing efficient combination therapies.

Regarding PET imaging of IDO1 activity, the main radiotracers investigated so far are 11C- or 18F-radiolabelled analogues of L- or D-tryptophan (Wierstra et al. 2019; John et al. 2020). Among them, 18F-labelled (S)-1-(2-fluoroethyl)tryptophan ((S)-[18F]FETrp), a substrate of the IDO1 enzyme (Henrottin et al. 2015), and currently under clinical investigation (NCT05556473), has demonstrated very promising preclinical results in terms of IDO1 positive-tumour cell uptake (Henrottin et al. 2016; Xin and Cai 2017; Xin et al. 2019) and pharmacokinetic profile in (patient-derived) tumour xenografts and transgenic tumour mouse models (Xin and Cai 2017; Xin et al. 2019; Xin et al. 2020; Michelhaugh et al. 2017). However, the widespread use of this radiotracer might be limited due to low radiochemical yields (RCYs) and long total radiosynthesis times of the procedures described so far (Henrottin et al. 2016; Xin and Cai 2017; Xin et al. 2019; Yue et al. 2020; Jiang et al. 2023). The latter are mainly caused by partial racemization occurring during the radiolabelling step and thus requiring lengthy and tedious chiral/reversedphase HPLC purification procedures.

Herein we describe simple and convenient syntheses of the standard compounds (S)- and (R)-FETrp and the corresponding tosylate precursors (S)- and (R)-3 with high enantiomeric excesses and the fully automated production of (S)-[18F]FETrp through a racemization-free one-pot two-step procedure.

Methods

General

All commercially available reagents and solvents were purchased at the following commercial suppliers: Sigma Aldrich, Alpha Aesar, ABX, Acros Organics, Fisher Scientific or Carlo Erba Reagents. Nα-(tert-butoxycarbonyl)-L-tryptophan and Nα-(tert-butoxycarbonyl)-D-tryptophan were purchased respectively from abcr GmbH (Karlsruhe, Germany) and Merck KGaA (Darmstadt, Germany). Whenever necessary, solvents were dried using common techniques (Armarego and Chai 2009). Unless otherwise noted, moisture-sensitive reactions were conducted under dry argon atmosphere. Temperatures indicated in the protocols correspond to the temperature of the oil bath. Analytical thin-layer chromatography (TLC) were performed on precoated silica gel 60 F254 plates (Merck or Macherey-Nagel) and visualised with UV light (254 nm) and/or developed with phosphomolybdic acid (8 wt%) in EtOH. Flash column chromatography was performed on silica gel 60 A normal phase, 35–70 Î¼m (Merck or SDS). Uncorrected melting points (mp) were recorded on an electrothermal capillary digital melting point apparatus IA9100 (Bibby Scientific). Nuclear magnetic resonance (NMR) spectra (500.13 MHz for 1H and 125.76 MHz for 13C) were recorded on a Bruker Avance 500 instrument with chemical shift values (δ) expressed in parts per million (ppm) relative to residual solvent as standard and coupling constants (J) given in Hz. 19F NMR spectra (470.3 MHz) were recorded on a Bruker Avance 500 apparatus using trifluorotoluene as internal reference (δ -63.72 ppm). Infrared (IR) spectra were recorded in the range 4000 – 440 cm− 1 on a Nicolet IS10 (Fisher Scientific) with attenuated total reflectance accessory. Organic compounds were analysed by High-Resolution Mass Spectrometry (HRMS) in positive or negative mode (Waters Micromass Q-Tof micro Mass Spectrometer). Optical rotations were measured with a Jasco-P-2000 digital polarimeter at 20 Â°C and for concentrations (c) given in g/100 mL. Analytical chiral chromatographic measurements of the compounds were performed on a JASCO LC-4000 system incorporating a PU-4180-LPG pump, an AS-4050 autosampler (100 µL sample loop), and a UV diode array detector MD-4015 coupled with a circular dichroism (CD) chiral detector CD-4095 (monitoring at 230 nm). Data analysis was performed with the ChromNAV 2.3 C software (JASCO Deutschland GmbH, Pfungstadt, Germany). The CD spectra were recorded with the CD-4095 detector during the high-performance liquid chromatography (HPLC) run in stopped-flow mode, during which the flow through the detector cell was bypassed using a software-controlled switching valve. Each spectrum was obtained with a scanning speed of 10 nm/sec. The single enantiomers were dissolved in appropriate solvents and subjected to chiral separations, which were performed in isocratic mode at a flow rate of 1 mL/min on a CHIRALPAK IA column (250 × 4.6 mm, 5 Î¼m, Daicel-Chiral Technologies Europe, France) for all intermediate compounds, or on a Reprosil Chiral-AA column (250 × 4.6 mm, 8 Î¼m, Dr. Maisch, Germany) for the final compounds (S)- and (R)-FETrp. Mobile phase compositions and wavelength UV monitoring used for chiral HPLC analysis: method A: n-hexane/i-PrOH (90/10, v/v) at 280 nm, method B: n-hexane/i-PrOH/TFA (90/10/0.05, v/v/v) at 280 nm; method C: MeCN/H2O (82/18, v/v) at 280 nm; method D: n-hexane/EtOH (90/10, v/v) at 225 nm. The enantiomeric excess (e.e.) was determined according to a previously published procedure (Eliel 1997).

Chemistry

General procedure for the synthesis of (S)- and (R)-5

These compounds were obtained with slight modifications of the experimental protocol described by Kim et al. (Kim et al. 2019). To a solution of N-(tert-butoxycarbonyl)-(L or D)-tryptophan (1.50 g, 4.93 mmol) in anhydrous N,N-dimethylacetamide (DMA) (6 mL) were successively added under argon atmosphere at room temperature benzyltriethylammonium chloride (BTEAC) (1.13 g, 4.96 mmol), K2CO3 (3.41 g, 24.7 mmol) and t-BuBr (5.54 mL, 49.3 mmol). The reaction mixture was heated at 55 Â°C for 5.5 h. After cooling to room temperature, deionised H2O (50 mL) was added and the resulting solution was extracted with EtOAc (3 × 50 mL). The combined organic layers were successively washed with deionised H2O (2 × 50 mL) and brine (50 mL), dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, EtOAc/cyclohexane, 2/8, v/v) to give compound (S)- or (R)-5.

(S)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1H-indol-3-yl)propanoate ((S)-5) was obtained according to the general method mentioned above as a white solid (1.10 g, 3.05 mmol). Yield: 62%. mp 143–144 Â°C (Lit.: 180–184 Â°C (Fujiwara et al. 2008); [α]20D +18.8 (c 0.39, CHCl3); Rf = 0.26 (SiO2, EtOAc/cyclohexane, 2/8, v/v); IR (ATR accessory) ν 3324, 2977, 1736, 1697, 1502, 1364, 1226, 1162, 1141 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.33 and 1.42 (s, 9H), 1.37 (s, 9H), 3.08 and 3.22 (dd, 1H, J = 14.7, 5.6 Hz), 3.08 and 3.28 (dd, 1H, J = 14.7, 5.7 Hz), 4.35 and 4.55 (m, 1H), 4.76 and 5.07 (d, 1H, J = 7.7 Hz), 7.02 (s, 1H), 7.12 (t, 1H, J = 7.6 Hz), 7.19 (t, 1H, J = 7.2 Hz), 7.35 (d, 1H, J = 8.1 Hz), 7.62 (d, 1H, J = 7.9 Hz), 8.11 (brs, 1H); 13C NMR (125.76 MHz, CDCl3) δ 28.0 (3C), 28.1, 28.4 (3C), 54.9, 79.7, 81.9, 110.5, 111.2, 119.1, 119.5, 122.1, 122.8, 128.0, 136.2, 155.4, 171.6; e.e.: >99% (chiral HPLC conditions: method A).

(R)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1H-indol-3-yl)propanoate ((R)-5) was obtained according to the general method mentioned above as a white solid (1.00 g, 2.77 mmol). Yield: 56%. mp 140–142 Â°C; [α]20D -21.4 (c 0.72, CHCl3); Rf = 0.26 (SiO2, EtOAc/cyclohexane, 2/8, v/v); IR (ATR accessory) ν 3324, 2977, 1736, 1697, 1502, 1365, 1226, 1162, 1141 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.34 and 1.43 (s, 9H), 1.38 (s, 9H), 3.08 and 3.22 (dd, 1H, J = 14.7, 5.5 Hz), 3.08 and 3.28 (dd, 1H, J = 14.7, 5.5 Hz), 4.36 and 4.56 (m, 1H), 4.86 and 5.09 (d, 1H, J = 7.1 Hz), 7.00 (s, 1H), 7.12 (t, 1H, J = 7.6 Hz), 7.18 (t, 1H, J = 7.2 Hz), 7.34 (d, 1H, J = 8.1 Hz), 7.62 (d, 1H, J = 7.9 Hz), 8.27 (brs, 1H); 13C NMR (125.76 MHz, CDCl3) δ 28.0 (3C), 28.1, 28.4 (3C), 54.9, 79.7, 81.9, 110.4, 111.2, 119.1, 119.4, 122.0, 122.9, 128.0, 136.2, 155.4, 171.6; e.e.: >99% (chiral HPLC conditions: method A).

General procedure for the synthesis of (S)- and (R)-4

To a solution of compound (S)- or (R)-5 (200 mg, 555 µmol) in anhydrous N,N-dimethylformamide (DMF) (2 mL) was added, under argon atmosphere at 0 Â°C, NaH (60 wt%, 27 mg, 675 µmol). After 5 min, 2-fluoroethyl tosylate (114 µL, 674 µmol), obtained according to the experimental protocol described by Wadsworth et al. (Wadsworth et al. 2010) (Additional file 1), was added and the reaction mixture was stirred at 0 Â°C for 50 min. The reaction was quenched with an aqueous saturated NH4Cl solution (15 mL) and extracted with EtOAc (3 × 15 mL). The combined organic layers were dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, EtOAc/cyclohexane, 2/8, v/v) to give compound (S)- or (R)-4.

(S)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1-(2-fluoroethyl)-1H-indol-3-yl)propanoate ((S)-4) was obtained according to the general method mentioned above as a white solid (132 mg, 325 µmol). Yield: 59%. mp 111–113 Â°C; Rf = 0.30 (SiO2, EtOAc/cyclohexane, 2/8, v/v); IR (ATR accessory) ν 3500 − 3100, 2973, 2950 − 2800, 1723, 1701, 1507, 1466, 1391, 1232, 1146, 1087, 1046 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.39 (s, 9H), 1.39 and 1.45 (s, 9H), 3.10 and 3.22 (dd, 1H, J = 14.8, 5.3 Hz), 3.10 and 3.28 (dd, 1H, J = 14.8, 5.5 Hz), 4.32 (dt, 2H, 3JH−F = 25.8 Hz, J = 5.0 Hz), 4.33 and 4.56 (d, 1H, J = 7.5 Hz), 4.68 (dt, 2H, 2JH−F = 47.0 Hz, J = 5.0 Hz), 4.87 and 5.09 (d, 1H, J = 7.5 Hz), 6.99 (s, 1H), 7.13 (t, 1H, J = 7.6 Hz), 7.23 (t, 1H, J = 7.3 Hz), 7.28 (d, 1H, J = 8.2 Hz), 7.63 (d, 1H, J = 7.9 Hz); 13C NMR (125.76 MHz, CDCl3) δ 28.0 (3C), 28.1, 28.4 (3C), 46.4 (d, 1C, 2JC−F = 21.8 Hz), 54.8, 79.6, 81.9, 82.3 (d, 1C, 1JC−F = 172.3 Hz), 109.0, 110.3, 119.5, 119.6, 122.0, 126.7, 128.8, 136.4, 155.4, 171.4; 19F NMR (470.59 MHz, CDCl3) δ -220.3 ppm; HRMS m/z calculated for C22H32FN2O4 [M + H]+: 407.2341, found: 407.2336; e.e.: 53.5% (chiral HPLC conditions: method A).

(R)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1-(2-fluoroethyl)-1H-indol-3-yl)propanoate ((R)-4) was obtained according to the general method mentioned above as a white solid (175 mg, 431 µmol). Yield: 78%. mp 111–113 Â°C; Rf = 0.30 (SiO2, EtOAc/cyclohexane, 2/8, v/v); IR (ATR accessory) ν 3357, 2978, 2931, 1723, 1699, 1507, 1468, 1392, 1250, 1233, 1147 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.39 (s, 9H), 1.39 and 1.45 (s, 9H), 3.11 and 3.22 (dd, 1H, J = 14.8, 5.5 Hz), 3.10 and 3.28 (dd, 1H, J = 14.8, 5.6 Hz), 4.35 (dt, 2H, 3JH−F = 25.9 Hz, J = 5.0 Hz), 4.37 and 4.54 (m, 1H), 4.68 (dt, 2H, 2JH−F = 47.0 Hz, J = 5.0 Hz), 4.80 and 5.09 (d, 1H, J = 8.0 Hz), 6.99 (s, 1H), 7.13 (t, 1H, J = 7.6 Hz), 7.23 (t, 1H, J = 8.1 Hz), 7.28 (d, 1H, J = 8.2 Hz), 7.63 (d, 1H, J = 7.9 Hz); 13C NMR (125.76 MHz, CDCl3) δ 28.0 (3C), 28.2, 28.4 (3C), 46.4 (d, 1C, 2JC−F = 21.8 Hz), 54.8, 79.6, 81.9, 82.4 (d, 1C, 1JC−F = 172.2 Hz), 109.0, 110.2, 119.4, 119.6, 122.0, 126.7, 128.8, 136.3, 155.4, 171.4; 19F NMR (470.59 MHz, CDCl3) δ -220.3 ppm; HRMS m/z calculated for C22H32FN2O4 [M + H]+: 407.2341, found: 407.2336; e.e.: 63.9% (chiral HPLC conditions: method A).

General procedure for the synthesis of (S)- and (R)-6

To a solution of N-(tert-butoxycarbonyl)-(L or D)-tryptophan (1.00 g, 3.29 mmol) in anhydrous DMF (15 mL) was added under argon atmosphere at room temperature a solution of 1 M KOtBu in anhydrous tetrahydrofuran (THF) (6.9 mL, 6.9 mmol). The resulting solution was cooled to 0 Â°C and 2-fluoroethyl tosylate (Wadsworth et al. 2010; additional file 1) (778 µL, 1.00 g, 4.6 mmol) was added. After stirring at 0 Â°C for 1.5 h, the reaction was quenched by successive addition of EtOAc (30 mL) and 1 M aqueous HCl solution (30 mL). The resulting mixture was decanted and the aqueous layer was extracted with EtOAc (3 × 30 mL). The combined organic layers were dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, EtOAc/cyclohexane/AcOH, 10/90/0.1 → 40/60/0.1, v/v/v), then (SiO2, CH2Cl2/EtOH/AcOH, 96/4/0.2, v/v/v) to give compound (S)- or (R)-6.

(S)-2-((tert-Butoxycarbonyl)amino)-3-(1-(2-fluoroethyl)-1H-indol-3-yl)propanoic acid ((S)-6) was obtained according to the general method mentioned above as a white solid (536 mg, 1.52 mmol). Yield: 47%. mp 72–74 Â°C; [α]20D +4.3 (c 0.48, CHCl3); Rf = 0.15 (SiO2, EtOAc/cyclohexane/AcOH, 40/60/0.1, v/v/v); Rf = 0.27 (SiO2, CH2Cl2/EtOH/AcOH, 96/4/0.2, v/v/v); IR (ATR accessory) ν 3700 − 3200, 3200 − 2700, 1709, 1505, 1469, 1394, 1367, 1332, 1249, 1158, 1052, 1015 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.25 and 1.43 (s, 9H), 3.08 and 3.33 (m, 2H), 4.30 (m, 2H), 4.46 and 4.70 (m, 1H), 4.64 (m, 2H), 5.06 and 6.12 (d, 1H, J = 7.2 Hz), 7.00 (s, 1H), 7.13 (t, 1H, J = 7.4 Hz), 7.23 (t, 1H, J = 7.1 Hz), 7.29 (d, 1H, J = 8.2 Hz), 7.61 (d, 1H, J = 7.8 Hz), 8.79 (brs, 1H); 13C NMR (125.76 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 27.7, 28.0 and 28.4 (3C), 46.5 (d, 1C, 2JC−F = 21.7 Hz), 54.3 and 55.2 (1C), 80.3, 82.3 (d, 1C, 1JC−F = 172.1 Hz), 109.3, 109.6, 119.3, 119.7, 122.2, 127.1, 128.6, 136.4, 155.7, 177.0; 19F NMR (470.59 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ -220.2 and -220.4 ppm; HRMS m/z calculated for C18H22FN2O4 [M-H]−: 349.1569, found: 349.1569; e.e.: >99% (chiral HPLC conditions: method B).

(R)-2-((tert-Butoxycarbonyl)amino)-3-(1-(2-fluoroethyl)-1H-indol-3-yl)propanoic acid ((R)-6) was obtained according to the general method mentioned above as a white solid (0.87 g, 2.48 mmol). Yield: 75%. mp 72–74 Â°C; [α]20D -6.0 (c 0.49, CHCl3); Rf = 0.15 (SiO2, EtOAc/cyclohexane/AcOH, 40/60/0.1, v/v/v); Rf = 0.27 (SiO2, CH2Cl2/EtOH/AcOH, 96/4/0.2, v/v/v); IR (ATR accessory) ν 3700 − 3200, 3200 − 2700, 1693, 1505, 1469, 1393, 1367, 1331, 1249, 1160, 1046 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.25 and 1.43 (s, 9H), 3.08 and 3.33 (m, 2H), 4.31 (m, 2H), 4.46 and 4.68 (m, 1H), 4.64 (m, 2H), 5.06 and 6.13 (d, 1H, J = 7.4 Hz), 7.00 (s, 1H), 7.13 (t, 1H, J = 7.5 Hz), 7.23 (t, 1H, J = 7.2 Hz), 7.29 (d, 1H, J = 8.2 Hz), 7.61 (d, 1H, J = 7.9 Hz), 8.26 (brs, 1H); 13C NMR (125.76 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 27.7 and 28.7 (1C), 28.0 and 28.4 (3C), 46.5 (d, 1C, 2JC−F = 21.5 Hz), 54.3 and 55.2 (1C), 80.3, 82.4 (d, 1C, 1JC−F = 172.1 Hz), 109.3, 109.6, 119.3, 119.7, 122.2, 127.1, 128.6, 136.4, 155.7, 176.9; 19F NMR (470.59 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ -220.2 and -220.4 ppm; HRMS m/z calculated for C18H22FN2O4 [M-H]−: 349.1569, found: 349.1567; e.e.: >99% (chiral HPLC conditions: method B).

General procedure for the synthesis of hydrochloride salts of (S)- and (R)-FETrp

To a solution of (S)- or (R)-6 (200 mg, 571 µmol) in anhydrous CH2Cl2 (5 mL) was added under argon atmosphere at room temperature a solution of 2.5 M HCl in anhydrous Et2O (10 mL). After stirring at room temperature for 3 h, the reaction mixture was evaporated under reduced pressure. The residue was triturated with anhydrous Et2O (10 mL), filtered and the solid was dried overnight at 25 Â°C in a vacuum desiccator to give compound (S)- or (R)-FETrp.

(S)-2-Amino-3-(1-(2-fluoroethyl)-1H-indol-3-yl)propanoic acid, hydrochloride salt ((S)-FETrp) was obtained according to the general method mentioned above as a light grey solid (138 mg, 481 mmol). Yield: 84%. mp 222–224 Â°C; [α]20D +1.8 (c 1.0, H2O/MeCN, 1/1, v/v); IR (ATR accessory) ν 3200 − 2700, 1758, 1590, 1500, 1470, 1346, 1192, 1182, 1112, 1089, 1037 cm− 1; 1H NMR (500.13 MHz, D2O) δ 3.35 (dd, 1H, J = 15.4, 7.1 Hz), 3.43 (dd, 1H, J = 15.4, 5.5 Hz), 4.29 (dd, 1H, J = 7.1, 5.5 Hz), 4.43 (dt, 2H, 3JH−F = 28.8 Hz, J = 4.7 Hz), 4.73 (dt, 2H, 2JH−F = 47.2 Hz, J = 4.7 Hz), 7.17 (t, 1H, J = 7.4 Hz), 7.26 (s, 1H), 7.28 (t, 1H, J = 7.4 Hz), 7.47 (d, 1H, J = 8.3 Hz), 7.63 (d, 1H, J = 8.0 Hz); 13C NMR (125.76 MHz, D2O) δ 25.6, 46.0 (d, 1C, 2JC−F = 19.6 Hz), 53.4, 83.6 (d, 1C, 1JC−F = 165.1 Hz), 106.4, 110.2, 118.6, 119.8, 122.3, 127.2, 128.7, 136.4, 172.0; 19F NMR (470.59 MHz, D2O) δ -221.0 ppm; HRMS m/z calculated for C13H16FN2O2 [M + H]+: 251.1190, found: 251.1187; e.e.: >99% (chiral HPLC conditions: method C).

(R)-2-Amino-3-(1-(2-fluoroethyl)-1H-indol-3-yl)propanoic acid, hydrochloride salt ((R)- FETrp) was obtained from (R)-6 (169 mg, 482 µmol) according to the general method mentioned above as a light grey solid (119 mg, 415 μmol). Yield: 86%. mp 222–224 Â°C; [α]20D -2.9 (c 1.0, H2O/MeCN, 1/1, v/v); IR (ATR accessory) ν 3200 − 2700, 1758, 1587, 1500, 1469, 1346, 1244, 1192, 1182, 1112, 1089, 1037 cm− 1; 1H NMR (500.13 MHz, D2O) δ 3.35 (dd, 1H, J = 15.3, 7.1 Hz), 3.43 (dd, 1H, J = 15.3, 5.2 Hz), 4.30 (m, 1H), 4.43 (dt, 2H, 3JH−F = 28.8 Hz, J = 4.2 Hz), 4.74 (m, 2H), 7.18 (t, 1H, J = 7.4 Hz), 7.26 (s, 1H), 7.29 (t, 1H, J = 7.3 Hz), 7.47 (d, 1H, J = 8.2 Hz), 7.65 (d, 1H, J = 7.8 Hz); 13C NMR (125.76 MHz, D2O) δ 25.6, 46.0 (d, 1C, 2JC−F = 19.7 Hz), 53.4, 83.6 (d, 1C, 1JC−F = 165.2 Hz), 106.4, 110.2, 118.6, 119.8, 122.3, 127.2, 128.7, 136.5, 172.0; 19F NMR (470.59 MHz, D2O) δ -221.1 ppm; HRMS m/z calculated for C13H16FN2O2 [M + H]+: 251.1190, found: 251.1188; e.e.: >99% (chiral HPLC conditions: method C).

General procedure for the synthesis of (S)- and (R)-7

To a solution of N-(tert-butoxycarbonyl)-(L or D)-tryptophan (4.50 g, 14.8 mmol) in anhydrous DMF (75 mL) was added, under argon atmosphere and at room temperature, a solution of 1 M KOtBu in anhydrous THF (31 mL, 31 mmol). The solution was cooled to 0 Â°C and benzyl 2-bromoethyl ether (3.27 mL, 20.7 mmol) was added. After stirring at 0 Â°C for 1 h, the reaction was quenched by successive additions of EtOAc (80 mL), an aqueous 1 M HCl solution (80 mL) and deionised H2O (80 mL). The mixture was decanted and the aqueous layer was extracted with EtOAc (3 × 100 mL). The combined organic layers were dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, EtOAc/cyclohexane/AcOH, 10/90/0.1 → 30/70/0.1, v/v/v) to give compound (S)- or (R)-7.

(S)-3-(1-(2-(Benzyloxy)ethyl)-1H-indol-3-yl)-2-((tert-butoxycarbonyl)amino)propanoic acid ((S)-7) was obtained according to the general method mentioned above as a white solid (4.50 g, 10.3 mmol). Yield: 69%. mp 48–50 Â°C; [α]20D +3.4 (c 0.49, CHCl3); Rf = 0.16 (SiO2, EtOAc/cyclohexane/AcOH, 30/70/0.1, v/v/v); IR (ATR accessory) ν 3100 − 2800, 1709, 1496, 1468, 1453, 1393, 1367, 1333, 1248, 1159, 1104, 1057, 1027 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.28 and 1.42 (s, 9H), 3.10 and 3.34 (m, 2H), 3.75 (t, 2H, J = 5.6 Hz), 4.26 (m, 2H), 4.44 (s, 2H), 4.44 and 4.66 (m, 1H), 5.05 and 5.80 (m, 1H), 7.02 (s, 1H), 7.12 (dt, 1H, J = 7.1, 0.8 Hz), 7.18 (m, 3H), 7.26 (m, 4H), 7.60 (d, 1H, J = 7.9 Hz), 8.06 (brs, 1H); 13C NMR (125.76 MHz, CDCl3) (mixture of cis-trans carbamate rotamers, 1 C missing) δ 27.7 and 28.9, 28.0 and 28.4 (3C), 46.3, 54.3 and 55.3 (1C), 69.0, 73.3, 80.2, 108.9, 109.5, 119.1, 119.4, 121.9, 127.5, 127.7 (2C), 127.8, 128.5 (2C), 136.5, 137.8, 155.7, 176.9; HRMS m/z calculated for C25H29N2O5 [M - H]−: 437.2082, found: 437.2085.

(R)-3-(1-(2-(Benzyloxy)ethyl)-1H-indol-3-yl)-2-((tert-butoxycarbonyl)amino)propanoic acid ((R)-7) was obtained from N-(tert-butoxycarbonyl)-D-tryptophan (2.00 g, 6.56 mmol) according to the general method mentioned above as a white solid (1.91 g, 4.36 mmol). Yield: 66%. mp 48–50 Â°C; [α]20D -6.4 (c 0.55, CHCl3); Rf = 0.16 (SiO2, EtOAc/cyclohexane/AcOH, 30/70/0.1, v/v/v); IR (ATR accessory) ν 3100 − 2700, 1711, 1684, 1528, 1469, 1366, 1335, 1268, 1249, 1231, 1167 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.25 and 1.43 (s, 9H), 3.09 and 3.34 (m, 2H), 3.75 (t, 2H, J = 5.5 Hz), 4.26 (m, 2H), 4.44 (s, 2H), 4.44 and 4.68 (m, 1H), 5.08 and 6.10 (d, 1H, J = 7.3 Hz), 7.04 (s, 1H), 7.12 (t, 1H, J = 7.5 Hz), 7.20 (m, 3H), 7.29 (m, 4H), 7.62 (d, 1H, J = 7.8 Hz), 9.54 (brs, 1H); 13C NMR (125.76 MHz, CDCl3) (mixture of cis-trans carbamate rotamers, 1 C missing) δ 27.7, 28.0 and 28.4 (3C), 46.3, 54.3 and 55.3 (1C), 69.0, 73.3, 80.2, 108.9, 109.5, 119.1, 119.4, 121.8, 127.5, 127.7 (2C), 127.8, 128.5 (2C), 136.5, 137.8, 155.7, 176.8; HRMS m/z calculated for C25H29N2O5 [M - H]−: 437.2082, found: 437.2083.

General procedure for the synthesis of (S)- and (R)-8

To a solution of carboxylic acid (S)- or (R)-7 (4.50 g, 10.3 mmol) in anhydrous DMA (75 mL) were successively added, under argon atmosphere and at room temperature, BTEAC (2.34 g, 10.3 mmol) and anhydrous K2CO3 (35.46 g, 257 mmol). t-BuBr (55.5 mL, 492 mmol) was then slowly added dropwise over 15 min and the reaction was stirred at 55 Â°C for 5 h. After cooling to room temperature, the reaction mixture was poured into a mixture of ice-cold water (270 mL) and EtOAc (150 mL). The mixture was decanted and the aqueous layer was extracted with EtOAc (3 × 100 mL). The combined organic layers were washed with brine (3 × 100 mL), dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, EtOAc/cyclohexane, 1/9 → 2/8, v/v) to give compound (S)- or (R)-8.

(S)-tert-Butyl 3-(1-(2-(benzyloxy)ethyl)-1H-indol-3-yl)-2-((tert-butoxycarbonyl)amino)propanoate ((S)-8) was obtained according to the general method mentioned above as a colourless oil (4.61 g, 9.36 mmol). Yield: 91%. [α]20D +14.2 (c 0.57, CHCl3); Rf = 0.41 (SiO2, EtOAc/cyclohexane, 2/8, v/v); IR (ATR accessory) ν 2976, 2931, 2849, 1708, 1495, 1468, 1454, 1392, 1365, 1249, 1145, 1059, 1027, 1014 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.40 (s, 9H), 1.40 and 1.46 (s, 9H), 3.13 and 3.25 (dd, 1H, J = 14.8, 5.4 Hz), 3.13 and 3.31 (dd, 1H, J = 14.8, 5.4 Hz), 3.78 (t, 2H, J = 5.7 Hz), 4.29 (t, 2H, J = 5.7 Hz), 4.48 (s, 2H), 4.37 and 4.57 (m, 1H), 4.82 and 5.10 (d, 1H, J = 7.6 Hz), 7.04 (s, 1H), 7.14 (t, 1H, J = 7.4 Hz), 7.23 (m, 3H), 7.31 (m, 4H), 7.64 (d, 1H, J = 7.9 Hz); 13C NMR (125.76 MHz, CDCl3) δ 28.0, 28.1 (3C), 28.5 (3C), 46.3, 54.8, 69.1, 73.4, 79.6, 81.8, 109.3, 109.5, 119.1, 119.4, 121.7, 127.1, 127.7 (2C), 127.8, 128.5 (2C), 128.7, 136.4, 137.9, 155.4, 171.5; HRMS m/z calculated for C29H39N2O5 [M + H]+: 495.2854, found: 495.2845.

(R)-tert-Butyl 3-(1-(2-(benzyloxy)ethyl)-1H-indol-3-yl)-2-((tert-butoxycarbonyl)amino)propanoate ((R)-8) was obtained from (R)-7 (1.32 g, 3.01 mmol) according to the general method mentioned above as a colourless oil (1.38 g, 2.80 mmol). Yield: 93%. [α]20D -18.7 (c 0.65, CHCl3); Rf = 0.41 (SiO2, EtOAc/cyclohexane, 2/8, v/v); IR (ATR accessory) ν 2976, 2931, 2849, 1708, 1495, 1468, 1454, 1392, 1365, 1249, 1223, 1145, 1059, 1027, 1014 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.42 (s, 9H), 1.42 and 1.48 (s, 9H), 3.14 and 3.27 (dd, 1H, J = 14.7, 5.5 Hz), 3.14 and 3.33 (dd, 1H, J = 14.7, 5.5 Hz), 3.79 (t, 2H, J = 5.7 Hz), 4.30 (t, 2H, J = 5.7 Hz), 4.49 (s, 2H), 4.39 and 4.59 (m, 1H), 4.85 and 5.13 (d, 1H, J = 7.8 Hz), 7.05 (s, 1H), 7.16 (t, 1H, J = 7.4 Hz), 7.25 (m, 3H), 7.33 (m, 4H), 7.66 (d, 1H, J = 7.9 Hz); 13C NMR (125.76 MHz, CDCl3) δ 27.9, 28.0 (3C), 28.4 (3C), 46.3, 54.8, 69.0, 73.3, 79.5, 81.7, 109.3, 109.5, 119.1, 119.4, 121.7, 127.1, 127.6 (2C), 127.7, 128.4 (2C), 128.7, 136.4, 137.9, 155.3, 171.4; HRMS m/z calculated for C29H39N2O5 [M + H]+: 495.2854, found: 495.2849.

General procedure for the synthesis of (S)- and (R)-9

To a degassed solution of benzyl derivative (S)- or (R)-8 (870 mg, 1.77 mmol) in MeOH (60 mL) was added Pd/C 10 wt% (564 mg). After stirring at room temperature for 2 h under hydrogen atmosphere, the suspension was filtered through a pad of celite® 545 and the pad was washed with MeOH (2 × 10 mL). The filtrate was filtered over a 0.45 Î¼m PTFE membrane filter and evaporated under reduced pressure to provide compound (S)- or (R)-9.

(S)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1-(2-hydroxyethyl)-1H-indol-3-yl)propanoate ((S)-9) was obtained according to the general method mentioned above as a colourless oil (465 mg, 1.15 mmol), which was used in the next step without further purification. Yield: 65%. [α]20D +14.9 (c 1.05, CHCl3); Rf = 0.47 (SiO2, EtOAc/cyclohexane, 5/5, v/v); IR (ATR accessory) ν 3600 − 3100, 2976, 2927, 1697, 1499, 1468, 1392, 1366, 1249, 1151, 1053, 1026 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.27 and 1.31 (s, 9H), 1.42 (s, 9H), 1.88 (brs, 1H), 3.04 and 3.12 (dd, 1H, J = 14.6, 6.7 Hz), 3.28 (dd, 1H, J = 14.6, 5.0 Hz), 3.88 (q, 2H, J = 4.9 Hz), 4.23 (m, 2H), 4.33 and 4.49 (m, 1H), 4.72 and 5.08 (d, 1H, J = 8.1 Hz), 7.03 (s, 1H), 7.11 (t, 1H, J = 7.4 Hz), 7.19 (t, 1H, J = 7.5 Hz), 7.31 (d, 1H, J = 8.2 Hz), 7.59 (d, 1H, J = 7.9 Hz); 13C NMR (125.76 MHz, CDCl3) δ 28.1 (3C), 28.3 (3C), 28.4, 48.9, 55.4, 61.9, 79.6, 82.1, 109.4, 109.9, 119.3, 119.5, 121.8, 127.3, 129.1, 136.2, 155.3, 171.5; HRMS m/z calculated for C22H33N2O5 [M + H]+: 405.2384, found: 405.2378.

(R)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1-(2-hydroxyethyl)-1H-indol-3-yl)propanoate ((R)-9) was obtained from (R)-8 (300 mg, 609 µmol) with a reaction time of 24 h according to the general method mentioned above as a colourless oil (100 mg, 247 µmol), which was used in the next step without further purification. Yield: 41%. [α]20D -16.4 (c 1.1, CHCl3); Rf = 0.47 (SiO2, EtOAc/cyclohexane, 5/5, v/v); IR (ATR accessory) ν 3600 − 3100, 2977, 2932, 1698, 1499, 1469, 1392, 1366, 1249, 1145, 1053, 1027 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.28 and 1.32 (s, 9H), 1.42 (s, 9H), 3.03 and 3.12 (dd, 1H, J = 14.6, 6.6 Hz), 3.27 (dd, 1H, J = 14.6, 5.1 Hz), 3.82 (t, 2H, J = 5.2 Hz), 4.18 (m, 2H), 4.33 and 4.49 (m, 1H), 4.80 and 5.11 (m, 1H), 7.01 (s, 1H), 7.10 (t, 1H, J = 7.4 Hz), 7.19 (t, 1H, J = 7.5 Hz), 7.30 (d, 1H, J = 8.2 Hz), 7.59 (d, 1H, J = 7.9 Hz); 13C NMR (125.76 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 28.1 (3C), 28.3 (3C), 28.7 and 29.8 (1C), 48.8, 55.3 and 55.7 (1C), 61.7, 79.6, 82.0, 109.4, 109.7, 119.3, 119.4, 121.8, 127.3, 129.0, 136.2, 155.3, 171.5; HRMS m/z calculated for C22H33N2O5 [M + H]+: 405.2384, found: 405.2378.

General procedure for the synthesis of (S)- and (R)-3

To a solution of alcohol (S)- or (R)-9 (465 mg, 1.15 mmol) in anhydrous CH2Cl2 (30 mL) were successively added, under argon atmosphere and at 0 Â°C, triethylamine (TEA) (224 µL, 1.61 mmol), N,N-dimethylaminopyridine (DMAP) (16 mg, 131 µmol) and TsCl (377 mg, 1.96 mmol). After stirring at 0 Â°C for 30 min, the reaction mixture was warmed to room temperature, stirred for 20 h and evaporated under reduced pressure. The residue was purified by column chromatography (SiO2, EtOAc/cyclohexane 1/9→3/7, v/v) to give compound (S)- or (R)-3.

(S)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1-(2-(tosyloxy)ethyl)-1H-indol-3-yl)propanoate ((S)-3) was obtained according to the general method mentioned above as a white solid (545 mg, 975 µmol). Yield: 85%. mp 51–53 Â°C; [α]20D +19.7 (c 1.0, CHCl3); Rf = 0.37 (SiO2, EtOAc/cyclohexane, 3/7, v/v); IR (ATR accessory) ν 2977, 2928, 1709, 1495, 1468, 1364, 1250, 1191, 1171, 1151, 1000 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.34 and 1.39 (s, 9H), 1.42 (s, 9H), 2.33 (s, 3H), 3.02 and 3.13 (dd, 1H, J = 14.8, 5.6 Hz), 3.02 and 3.22 (dd, 1H, J = 14.8, 5.4 Hz), 4.27 (m, 2H), 4.29 (m, 2H), 4.29 and 4.49 (m, 1H), 4.75 and 5.06 (d, 1H, J = 7.8 Hz), 6.83 (s, 1H), 7.10 (m, 5H), 7.46 (d, 2H, J = 8.0 Hz), 7.55 (d, 1H, J = 7.4 Hz); 13C NMR (125.76 MHz, CDCl3) δ 21.6, 27.8, 28.0 (3C), 28.4 (3C), 45.0, 54.7, 67.9, 79.6, 81.9, 108.8, 110.3, 119.4, 119.5, 122.0, 126.6, 127.6 (2C), 128.8, 129.7 (2C), 132.0, 135.9, 144.9, 155.3, 171.3; HRMS m/z calculated for C29H39N2O7S [M + H]+: 559.2473, found: 559.2468. e.e.: >99% (chiral HPLC conditions: method D).

(R)-tert-Butyl 2-((tert-butoxycarbonyl)amino)-3-(1-(2-(tosyloxy)ethyl)-1H-indol-3-yl)propanoate ((R)-3) was obtained from (R)-9 (167 mg, 413 µmol) according to the general method mentioned above as a white solid (126 mg, 226 µmol). Yield: 55%; mp 51–53 Â°C; [α]20D -21.8 (c 1.0, CHCl3); Rf = 0.37 (SiO2, EtOAc/cyclohexane, 3/7, v/v); IR (ATR accessory) ν 2977, 2928, 1708, 1495, 1468, 1363, 1251, 1184, 1164, 1151, 999 cm− 1; 1H NMR (500.13 MHz, CDCl3) (mixture of cis-trans carbamate rotamers) δ 1.34 and 1.43 (s, 9H), 1.39 (s, 9H), 2.34 (s, 3H), 3.02 and 3.13 (dd, 1H, J = 14.8, 5.6 Hz), 3.02 and 3.22 (dd, 1H, J = 14.8, 5.5 Hz), 4.26 (m, 2H), 4.30 (m, 2H), 4.30 and 4.50 (d, 1H, J = 7.9 Hz), 4.73 and 5.05 (m, 1H), 6.83 (s, 1H), 7.10 (m, 5H), 7.47 (d, 1H, J = 8.1 Hz), 7.54 (d, 1H, J = 7.5 Hz); 13C NMR (125.76 MHz, CDCl3) δ 21.7, 27.9, 28.1 (3C), 28.4 (3C), 45.1, 54.8, 67.9, 79.7, 82.0, 108.9, 110.4, 119.4, 119.5, 122.1, 126.6, 127.7 (2C), 128.9, 129.8 (2C), 132.1, 135.9, 144.9, 155.4, 171.4; HRMS m/z calculated for C29H39N2O7S [M + H]+: 559.2473, found: 559.2469. e.e.: >99% (chiral HPLC conditions: method D).

Radiochemistry

General

No-carrier-added fluorine-18 was produced by Curium Pharma via the [18O(p, n)18F] nuclear reaction by irradiation of a 2.8 mL > 97%-enriched [18O]H2O target (Bruce Technology) on a PETtrace cyclotron (16 MeV proton beam, GE healthcare). The radio TLC plates (Macherey-Nagel, Precoated TLC sheets ALUGRAM® Xtra SIL G/UV254) were developed with EtOAc/cyclohexane (3/7, v/v) and measured on a miniGITA Dual radio-TLC instrument (Elysia-Raytest). Analytical radio/UV-HPLC measurements were performed on a system consisting of an Agilent HP series 1100 (Hewlett Packard, Les Ulis, France) combined with a Flo-one A500 Radiomatic detector (Packard, Canberra, Australia). The analyses were carried out on a C-18 column (Agilent Zorbax Extend C18, 5 Î¼m, 4.6 × 150 mm, equipped with a guard column) using the following solvent conditions: H2O containing 0.1% of trifluoroacetic acid (TFA) (solvent A) and MeCN containing 0.1% of TFA (solvent B); 0 to 15 min: gradient elution 90% → 70% A with a flow rate of 1 mL/min, λ = 290 nm. Analytical radio/chiral HPLC measurements were performed on a JASCO LC-4000 system incorporating a PU-4180-LPG pump, an AS-4050 autosampler (100 µL sample loop), an UV-4070 UV/Vis detector combined with a GabiNova Radio Flow monitor (mid energy probe 60–600 keV, Elysia-Raytest). Analyses were performed on a Reprosil Chiral-AA column (250 × 4.6 mm, 8 Î¼m, Dr. Maisch, Germany) eluted with a mixture of H2O/MeCN in isocratic mode (55/45, v/v) at λ = 290 nm and with a flow rate of 1 mL/min.

Radiochemical syntheses and semi-preparative HPLC purifications were performed using a SynChrom R&D EVOI radiosynthesiser (Raytest). Sep-Pak® Accell Plus QMA carbonate Plus Light cartridges (46 mg, 40 Î¼m) and Oasis MCX plus short cartridge (225 mg, 60 Î¼m) were purchased from Waters. Prior to use, Oasis MCX cartridges were washed with deionised H2O (5 mL), EtOH (5 mL) and deionised H2O (5 mL). Semi-preparative HPLC purifications were performed on a Symmetryprep C18 column (300 × 7.8 mm; 7 Î¼m; Waters) eluted with a mixture of H2O/MeCN/TFA (90/10/0.01, v/v/v) in isocratic mode at l = 254 nm and with a flow rate of 1.5 mL/min for one min and then 2.5 mL/min. All radiolabelled compounds were analysed by radio-TLC and/or analytical radio/UV-HPLC and compared to the authentic non-radioactive materials. Radiochemical yield (RCY) refers to the measure of the proportion of a given radiotracer, isolated after purification, with respect to the starting radioactivity engaged in the radiosynthesis. The RCYs have been decay corrected. Radiochemical conversions (RCCs), used to describe the reaction efficiency, were obtained after radio-TLC analysis of an aliquot of the crude reaction and were given as the percent of activity of the integrated region of the desired product versus the activity of total strip.

Preparation of [18F]fluoride solution for manual syntheses

The aqueous solution of [18F]fluoride in [18O]H2O was loaded on the anion exchange Sep-Pak® QMA cartridge (46 mg). The trapped [18F]fluoride was then eluted to a glass reactor using a solution of tetra-n-butylammonium hydrogen carbonate (TBA-HCO3) in a mixture of H2O/MeCN (1 mL, 1/1, v/v). After elution, solvents were evaporated at 90 Â°C for 5 min under reduced pressure and a gentle stream of helium. A second azeotropic drying was repeated by addition of anhydrous MeCN (2.0 mL). After cooling to 25–30 Â°C, the dried tetra-n-butylammonium [18F]fluoride (TBA[18F]F) was diluted in the appropriate anhydrous solvent (MeCN or DMF) and divided into portions for the manual radiosynthesis optimisation tests described below. To note, these solutions have been used immediately after preparation to ensure high and reproducible RCC.

Optimisation of radiolabelling: manual procedure

In a glass vial containing the precursor (S)-3 (1–8 mg) was added an aliquot of the TBA[18F]F stock solution prepared as described above (70–310 MBq). Whenever required, the solution was diluted with the appropriate solvent (i.e. anhydrous MeCN, DMF or t-BuOH) to reach a final volume of 500 µL. The vial was sealed and placed in a heating block preheated at 90–130 Â°C. After 5–10 min of heating, the vial was transferred in an ice bath and aliquots of the crude radiosynthesis mixture were analysed by radio TLC to determine RCC (Table 1). All experiments were performed in triplicate.

Automated radiolabelling procedure

The aqueous solution of [18F]fluoride in [18O]H2O, placed in the target vial (Scheme 1), was trapped on the anion exchange Sep-Pak® QMA cartridge (46 mg, Trap 1). The radioactivity was eluted into the 12 mL reactor using a solution of TBA-HCO3 (4.4 mg, 14.5 µmol) in a mixture of H2O/MeCN (1 mL, SC1 loop). After 30 s of gentle helium bubbling (through needle valve), the solvents were evaporated at 90 Â°C under reduced pressure and helium flow (slow increase from 25 Â°C to 50 Â°C by steps of 10 Â°C over a 2 min period, increase to 90 Â°C in 1 min, heating at 90 Â°C for 3 min). After cooling to 25–30 Â°C within 2 min, anhydrous MeCN (2 mL, SC2) was added to the reactor. After 30 s of gentle helium bubbling (through needle valve), the azeotropic drying process under helium flow and reduced pressure was repeated (heating for 2 min at 35 Â°C, increase to 90 Â°C in 1 min, heating at 90 Â°C for 4 min). After cooling to 25–30 Â°C within 2–3 min, a freshly prepared solution of the precursor (S)-3 (2 mg) in a mixture of anhydrous DMF and t-BuOH, (1 mL, 9/1, v/v, SC3) was added to the dried TBA[18F]F. After 30 s of gentle helium bubbling (through needle valve), the reaction mixture was heated at 110 Â°C for 7 min (2 min of increase of temperature to reach 110 Â°C and further 5 min heating at 110 Â°C). The reactor was cooled to 30 Â°C before addition of a 6 M aqueous HBr solution (500 µL, SC5) under stirring. After 30 s of gentle helium bubbling (through needle valve), the reaction mixture was heated at 110 Â°C for 5 min. After cooling to 25–30 Â°C in 2–3 min, the crude reaction mixture was diluted with a 1.1 M NaOAc buffer solution pH 5.5 (3.5 mL, SC8). After 30 s of gentle helium bubbling (through needle valve), the solution containing the crude (S)-[18F]FETrp was transferred to the HPLC loop (5 mL) and purified via semi-preparative HPLC (conditions in general radiochemistry section). The collected fraction (tR = 26–27 min) was diluted with deionised H2O (20 mL, loaded in SC11) and trapped on an Oasis MCX plus short cartridge (pre-conditioned as described in the general radiochemistry section, Trap 3). The cartridge was washed with deionised H2O (10 mL, SC9) and slowly eluted with 5% NH4OH (28–30% in H2O)/EtOH solution (3 mL, SC7). The final product was diluted with saline immediately after elution (30 mL, product vial) and the pH was adjusted to 5–7 using acetic acid (135 µL).

Results

Organic chemistry

The syntheses of enantiopure standard compounds (S)- and (R)-FETrp and precursors (S)- and (R)-FETrp are shown in Fig. 1.

Fig. 1
figure 1

Synthesis of references (S)- and (R)-FETrp and corresponding precursors (S)- and (R)-3. (a) BTEAC, anhyd. K2CO3, t-BuBr, anhyd. DMA, 55 Â°C, 5.5 h; (b) (i) NaH 60 wt%, anhyd. DMF, 0 Â°C, 5 min; (ii) 2-fluoroethyl tosylate (1), 0 Â°C, 50 min; (c) (i) KOtBu 1 M in anhyd. THF, anhyd. DMF, rt; (ii) 2-fluoroethyl tosylate (1), 0 Â°C, 1.5 h; (d) HCl 2.5 M in anhyd. Et2O, anhyd. CH2Cl2, rt, 3 h; (e) (i) KOtBu 1 M in anhyd. THF, anhyd. DMF, rt; (ii) benzyl 2-bromoethyl ether, 0 Â°C, 1 h; (f) BTEAC, anhyd. K2CO3, t-BuBr, anhyd. DMA, 55 Â°C, 5 h; (g) H2, Pd/C 10 wt%, MeOH, rt, 2–24 h; (h) TsCl, TEA, DMAP, anhyd. CH2Cl2, 0 Â°C, 30 min then, rt, 20 h

Briefly, N1-alkylation of commercially available Na-Boc-L- and D-tryptophan derivatives with 2-fluoroethyltosylate (Fig. 1, Way B) or benzyl 2-bromoethyl ether (Fig. 1, Way C) in the presence of potassium tert-butylate gave the corresponding intermediates (S)- and (R)-6 or (S)- and (R)-7, respectively. Subsequent acidic cleavage of the tert-butyl ester function of (S)- and (R)-6 afforded standard references (S)- and (R)-FETrp, respectively, while the protection of the carboxylic acid function of (S)- and (R)-7 as a tert-butyl ester, followed by hydrogenolytic debenzylation and final tosylation gave the corresponding precursors (S)- and (R)-3.

Radiochemistry

A broad set of different experimental conditions (i.e. amounts of base and precursor, solvent, reaction time and temperature) were manually screened with the aim to identify the key parameters that influence the RCC of (S)-[18F]4 from tosylate precursor (S)-3. The results are summarized in Table 1. The best RCCs (> 90% as estimated by radio-TLC of the crude reaction mixture) could be reached using low amounts of precursor (1–2 mg, 1.8–3.6 µmol), moderate reaction time (5 min) and temperature (110 Â°C) in MeCN or mixtures of MeCN or DMF with t-BuOH (Table 1, entries 11–15).

Table 1 Optimization of the experimental conditions for manual radiofluorination from (S)-3 leading to (S)-[18F]4 intermediate

Based on these findings, a fully automated radiosynthesis of (S)-[18F]FETrp on a Raytest Synchrom R&D Evo I radiosynthesizer was developed. The detailed synthesizer setup is described in Scheme 1.

Scheme 1
scheme 1

Raytest Synchrom R&D Evo I layout for the automated production of (S)-[18F]FETrp from precursor (S)-3. 0: aqueous solution of [18F]fluoride in [18O]H2O; 1: anion exchange Sep-Pak® QMA cartridge (46 mg); 2: solution of TBA-HCO3 (4.4 mg, 14.5 μmol) in H2O/MeCN (1 mL, 1/1, v/v); 3: anhyd. MeCN (2 mL); 4: precursor (S)-3 (2 mg) in anhyd. DMF/t-BuOH, (1 mL, 9/1, v/v); 5: 6 M aqueous HBr solution (500 µL); 6: 1.1 M NaOAc buffer solution pH 5.5 (3.5 mL); 7: Symmetryprep C18 column (300 × 7.8 mm; 7 Î¼m; Waters); 8: H2O/MeCN/TFA (90/10/0.01, v/v/v), isocratic mode, λ = 254 nm, flow rate = 1.5 mL/min for one min and then 2.5 mL/min; 9: collection vial pre-filled with deionised H2O (20 mL); 10: pre-conditioned Oasis MCX plus short cartridge; 11: deionised H2O (10 mL); 12: 5% NH4OH/EtOH solution (3 mL); 13: final product vial for formulation with saline (30 mL) and AcOH (135 µL)

The radiosynthesis of (S)-[18F]FETrp was completed in about 80–85 min in 55.2 ± 7.5% RCY (n = 3). Accordingly, (S)-[18F]FETrp could be obtained in excellent radiochemical purities (RCP, > 99%) and e.e. (> 99%).

Discussion

The first radiosynthesis of [18F]FETrp was reported by Sun et al. (Sun et al. 2012) according to a two-pot three-step procedure, including: (i) classical radiosynthesis of 2-[18F]fluoroethyl tosylate ([18F]1); (ii) N-alkylation of the precursor Nα-Boc-L-tryptophan ethyl ester with [18F]1; and (iii) cleavage of the amino acid protecting groups of intermediate [18F]2 before final RP-HPLC purification (Fig. 2, strategy A).

Fig. 2
figure 2

Main radiosynthetic routes available for the production of racemic (R/S)- or enantiopure (S)-[18F]FETrp

Accordingly, [18F]FETrp was obtained through a 65 min radiosynthesis, in low RCY (0.9 ± 0.2%, non-decay corrected) and without final control of the radiotracer stereochemistry. Subsequently and taking into account that (S)-[18F]FETrp has demonstrated both in vitro and in vivo greater specificity for IDO1 than its enantiomer (R)-[18F]FETrp (Henrottin et al. 2016; Xin and Cai 2017), efforts have been done to develop a more efficient and asymmetric radiolabelling procedure for the production of (S)-[18F]FETrp. As shown in Fig. 2 (strategy B), all the radiosyntheses published so far have in common the nucleophilic substitution of the tosylate group of precursor 3 with the K[18F]F/K222/K2CO3 complex, followed by removal of the tert-butoxycarbonyl (Boc) and tert-butyl ester groups of intermediate [18F]4 in acidic conditions, giving access to the (S)-[18F]FETrp enantiomer after chiral semi-preparative HPLC purification. To fulfill good manufacturing practices requirements, additional semi-preparative RP-HPLC purification was required to remove chemical impurities co-eluting with (S)-[18F]FETrp during the chiral HPLC separation step and thus increasing procedure times (Yue et al. 2020; Jiang et al. 2023). Overall, these radiosynthesis strategies resulted in the production of (S)-[18F]FETrp with RCYs of 6–30%, excellent RCP and e.e. (both > 95%), and moderate to high molar activities (Am) (15 to 495 GBq/µmol). The process durations range between 50 and 155 min. The main drawback of these strategies remains the partial racemization that occurred during the nucleophilic [18F]fluoride substitution reaction, leading to [18F]4 in a 2:1 ratio of the (S):(R) enantiomers even when the optically pure tosylate precursor (S)-3 was used. As postulated by Xin et al. (Xin and Cai 2017), we assumed that this phenomenon was most likely due to the basic conditions of the radiofluorination step using the K[18F]F/K222/K2CO3 complex that are sufficient enough to partially deprotonate the α-C-H of the amino ester group. To confirm this hypothesis and find optimal radiolabelling conditions for a racemization-free radiofluorination of (S)-[18F]FETrp, it was first necessary to synthesise both enantiomers of FETrp and precursor 3 with high optical purities.

Organic chemistry

Apart of chemical supplier sources, there is only one published asymmetric route to the tosylate precursor (S)-3 with assumed 78% e.e. (Xin and Cai 2017), while the synthesis of the pure enantiomer (S)-FETrp was reported in a single literature reference on a milligram scale using enzymatic kinetic resolution (Henrottin et al. 2016) (see Fig. S1, additional file 1).

Based on the previously published procedures (Henrottin et al. 2016; Xin and Cai 2017), different conditions were screened to introduce the fluoroethyl group by N1-alkylation of the Nα-Boc-L- or D-tryptophan t-Bu ester (S)- or (R)-5, obtained from corresponding Nα-Boc-L- or D-tryptophan (Fig. 1, Way A) with > 99% e.e. (see Fig. S2, additional file 1). The reaction was conducted at 0 Â°C with reduced amount of NaH (1.2 eq) and shorter reaction time (50 min) than previously described. Unfortunately, these milder conditions led to partial racemization of the esters (S)-4 or (R)-4 revealed by chiral UV-CD-HPLC (53.5 or 63.9% e.e., respectively) (Fig. 3A).

Fig. 3
figure 3

Representative chromatograms of the chiral UV-CD-HPLC analyses of compounds 4, 6, FETrp and 3. Conditions: CD detection at 230 nm, (A) CHIRALPAK IA, n-hexane/i-PrOH (90/10, v/v), UV detection at 280 nm; (B) CHIRALPAK IA, n-hexane/i-PrOH/TFA (80/20/0.05, v/v/v), UV detection at 280 nm; (C) Reprosil Chiral-AA, MeCN/H2O (82/18, v/v), UV detection at 280 nm; (D) CHIRALPAK IA, n-hexane/EtOH (90/10), UV detection at 225 nm

The same trend was observed when lithium bis(trimethylsilyl)amide was employed as a base in THF at -78 Â°C according to a previously reported procedure for the synthesis of N1-prenylated Nα-Boc-L-tryptophan methyl ester (Yu et al. 2020) (data not shown). To work around this problem, a recent method for the N1-alkylation of tryptophan derivatives was investigated (Junk et al. 2021). This interesting strategy requires the use of unprotected carboxylic acid tryptophan derivatives in the presence of 2.1 equivalents of KOtBu, a weaker base compared to NaH. The first base equivalent leads to the formation of the corresponding carboxylate, which in turn decreases the acidity of the α-C-H, thus avoiding the enolate formation and epimerisation of the chiral centre. The esterification side reaction could also be reduced when the reaction was conducted in DMF at 0 Â°C. Accordingly, both enantiomers of the N1-fluoroethyl derivative 6 were obtained from Nα-Boc-tryptophan by N1-alkylation with 2-fluoroethyl tosylate in good chemical yields (47–75%) (Fig. 1, Way B) and excellent e.e. (> 99%) (Fig. 3B). Removal of the Boc protecting group of (S)- and (R)-6 was then achieved by using HCl 2.5 M in anhydrous Et2O to yield standard references (S)- and (R)-FETrp, as hydrochloride salts, with high optical purities (> 99% e.e.) (Fig. 3C).

Based on these results, we then applied this efficient procedure to obtain the precursor (S)-3 and its enantiomer (R)-3 (Fig. 1, Way C). Briefly, L and D isomers of Nα-Boc-tryptophan were alkylated with benzyl 2-bromoethyl ether to yield the N1-benzyloxyethyl derivatives (S)- and (R)-7. Then, tert-butyl esterification according to the literature procedure of Chevallet et al. (Chevallet et al. 1993), followed by Pd/C-catalysed hydrogenolysis of the benzyl protecting group of (S)- and (R)-8 gave the alcohols (S)- and (R)-9, which were converted into their corresponding tosylates (S)- and (R)-3, respectively. Following this procedure, high enantiomeric purities (up to 99% e.e. determined by chiral-HPLC analyses) were attained for both enantiomers of precursor 3 (Fig. 3D).

Radiochemistry

With optically pure standard references and precursors in hands, aliphatic nucleophilic substitution of precursor (S)-3 using [18F]fluoride was then investigated. To achieve less basic radiofluorination conditions, we turned our attention to TBA[18F]F as an alternative to the K[18F]F/K222/K2CO3 complex (Haveman et al. 2023). First of all, we tested the elution efficiency of [18F]fluoride trapped on the QMA cartridge with increasing concentrations of TBA-HCO3 in a mixture of H2O/MeCN (1 mL, 1/1, v/v). We determined that a concentration ≥ 7.5 mM of TBA-HCO3 (i.e. amount of TBA-HCO3 ≥ 7.5 µmol) was required to recover at least 85% of [18F]fluoride in the reactor. We then manually explored a set of experimental conditions (molar amounts of base and precursor, solvent, reaction time and temperature) for the radiofluorination of the tosylate precursor (S)-3 (Table 1).In order to minimise the base content, the initial experiments were carried out with a fixed amount of 7.5 µmol of TBA-HCO3 (Table 1, entries 1–7). When using 1 mg (1.8 µmol) of precursor in anhydrous DMF at 110 Â°C, 58 ± 6% of the protected [18F]FETrp intermediate [18F]4 was obtained after 5 min. Lowering the temperature from 110 to 90 Â°C decreased the RCC (Table 1, entries 3 and 4), while prolonged reaction time had no significant impact on the radiolabelling efficiency (Table 1, entries 2 and 4). Increasing the amount of precursor from 1 to 8 mg led to a noticeable decrease of the RCC by 40%. The radiolabelling efficiency was also dependent on the amount of base used. Indeed, lowering the amount of TBA-HCO3 to 3.75 µmol reduced significantly the RCC, while doubling the amount of base to 15 µmol led to a slight increase of the RCC (Table 1, entries 8 and 9). At this stage and in order to verify that such large amounts of base did not lead to partial racemization, crude [18F]4, obtained from conditions of entry 9, was fully deprotected using 6 M aqueous HBr solution at 110 Â°C for 5 min according to the procedure of Henrottin et al. (Henrottin et al. 2016). After purification by semipreparative HPLC, high RCP as well as e.e. of (S)-[18F]FETrp (> 98% determined by radio-(chiral)-HPLC analyses) were obtained thus validating the choice of TBA-HCO3 as base for this radiosynthesis. Raising the reaction temperature to 130 Â°C did not give any further benefits (Table 1, entry 10). Next the effect of the solvent was investigated, while keeping constant the amount of base (i.e. 15 µmol). Running the reaction in anhydrous DMF in the presence of 10% and 20% of t-BuOH, known sometimes to greatly enhance the nucleophilic radiofluorination rates (Kim et al. 2006), significantly improved the RCC (91% for both) (Table 1, entries 11 and 12). When MeCN was used, the protected intermediate [18F]4, from 1 to 2 mg (1.8 or 3.6 µmol) of precursor (S)-3, was produced in near-quantitative RCC (Table 1, entries 13 and 14). In contrast to DMF, the addition of t-BuOH as co-solvent with MeCN was found to have a negative impact on the radiolabelling efficiency (Table 1, entry 15). Interestingly, in all conditions tested, apart from unreacted [18F]fluoride, only the protected derivative [18F]4 was detected using radio-HPLC and radio-TLC analyses of the crude reaction mixture.

Based on these findings, the optimised reaction conditions obtained (Table 1, entry 13, RCC: 98 ± 1%) were then applied on a Raytest Synchrom R&D Evo I radiosynthesizer to develop a novel automated radiosynthesis of (S)-[18F]FETrp (Scheme 1). Briefly, after trapping [18F]fluoride (2.0-3.8 GBq) on the anion exchange cartridge, elution of the activity to the reaction vessel using a solution of TBA-HCO3 (15 µmol, elution efficiency: 97.8 ± 0.9%) and two successive azeotropic drying steps under helium stream and reduced pressure, the resulting anhydrous TBA[18F]F was allowed to react with the tosylate precursor (S)-3 in anhydrous MeCN at 110 Â°C for 5 min. The RCC was controlled by radio-TLC analysis of the crude reaction mixture to ensure that the automated process gave similar results than the manual radiosynthesis. Surprisingly, the RCC dropped to 64% and after the acidic deprotection step, the e.e. of the final radiotracer was also significantly impaired (87% e.e.). Having noticed that the pressure in the closed reaction vessel increased significantly during the heating at 110 Â°C, we hypothesized that the geometry and the total volume of the reaction vessel (12 mL in the automate vs. 7 mL for the manual process) induced the partial vaporization of the solvent (i.e. MeCN), increasing the concentrations of the precursor (S)-3 and the TBA-HCO3 base in the reaction media thus contributing to the significant decrease in RCC and partial racemization of (S)-[18F]FETrp. In consequence, lower amounts of TBA-HCO3 (7.5, 11.2, and 14.5 µmol) and/or larger volumes of MeCN (750, and 1000 µL) were then investigated. Under these conditions, only RCC of 15-47% could be achieved while the e.e. was still lower than 93%. Being unsuccessful in reproducing the manual conditions optimised with MeCN, we decided to explore the use of the DMF/t-BuOH mixture (9/1, v/v, 500 µL) depicted in Table 1 (entry 11, RCC: 91 ± 2%). Under these conditions, RCC of 50% could be achieved with significantly lower overpressure in the reaction vessel (1340 mbar vs. >2000 mbar with MeCN). However, the e.e. of the final (S)-[18F]FETrp obtained after acidic cleavage of the protective groups was still sub-optimal (87%). To address this challenge, the TBA-HCO3 concentration in the reaction media was reduced by half by adjusting the volume of the reaction mixture to 1 mL while maintaining the concentration of precursor (i.e. 2 mg/mL). Applying these conditions, higher RCC of 89.8 ± 5.8% (n = 4) could be achieved. Then, the deprotection of the intermediate [18F]4 using a 6 M aqueous HBr solution was performed and followed by purification of the crude product by semi-preparative RP-HPLC (see Fig. S3, additional file 1). The collected fraction was diluted with water and the final formulation of the radiotracer was performed by solid-phase extraction using an Oasis MCX plus cartridge. Elution under basic conditions, as described by Jiang et al. (Jiang et al. 2023) successfully afforded (S)-[18F]FETrp in 55.2 ± 7.5% RCY, 99.9% RCP, 99.1 ± 0.5% e.e. and Am of 53.2 ± 9.3 GBq/µmol (n = 3) with a total process duration of 80–85 min (Figs. 4 and 5).

Fig. 4
figure 4

In-house developed enantiospecific one-pot two-step radiosynthesis of (S)-[18F]FETrp

Fig. 5
figure 5

Representative HPLC chromatograms of formulated (S)-[18F]FETrp. Analytical HPLC chromatograms of (S)-[18F]FETrp (B) (radioactivity) co-injected with (S)-FETrp (A) (UV, λ at 290 nm) on Agilent Zorbax Extend C18 column. Chiral HPLC chromatograms of (S)-[18F]FETrp (D) (radioactivity) co-injected with (S/R)-FETrp (C) (UV, λ at 290 nm) on Reprosil Chiral-AA column. The radio-HPLC detectors were connected in series after the UV detectors accounting for the slight differences in retention times (∼0.3 min) observed between UV and radioactive signals

Conclusion

In summary, we have completed the synthesis of enantiomerically pure (S)- and (R)-FETrp and tosylate precursor (S)-3. Manual radiolabelling screening was performed to find the optimal reaction conditions for the radiosynthesis of (S)-[18F]FETrp. The use of low amounts of precursor, short reaction times and TBA-HCO3 as the base was found to be important to achieve high RCC and e.e.

The transfer of the optimal manual labelling parameters to an automated radiosynthesizer (SynChrom R&D EVOI) required a few additional modifications regarding the total reaction volume in order to achieve high RCC in the crude reaction mixture (89.8 ± 5.8%, n = 4) while maintaining a very high e.e. (> 99%). A fully automated one-pot two-step radiosynthesis of (S)-[18F]FETrp could be finally achieved after acidic deprotection, semi-preparative RP-HPLC purification and final formulation via solid-phase extraction in 80–85 min. This simple and convenient procedure represents a significant improvement compared to the previously published protocols in terms of the purification process, the total radiosynthesis time and overall RCY (Fig. 4). We assume that this process could be easily implemented to other existing radiosynthesis modules and will broaden the use of (S)-[18F]FETrp for PET imaging of IDO1 activity with potential benefits in a wide range of physiological and pathophysiological applications.

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

Am :

Molar activity

Anhyd.:

Anhydrous

Boc:

tert-Butoxycarbonyl

BTEAC:

Benzyltriethylammonium chloride

CD:

Circular dichroism

DMF:

N,N-dimethylformamide

DMA:

N,N-dimethylacetamide

DMAP:

N,N-dimethylaminopyridine

e.e.:

Enantiomeric excess

FETrp:

1-(2-Fluoroethyl)tryptophan

HPLC:

High-performance liquid chromatography

HRMS:

High resolution mass spectrometry

IC:

Immune checkpoint

ICI:

Immune checkpoint inhibitor

IDO:

Indolamine 2,3-dioxygenase

IR:

Infrared

NMR:

Nuclear magnetic resonance

PET:

Positron emission tomography

PTFE:

Polytetrafluoroethylene

ppm:

Parts per million

QMA:

Quaternary methyl ammonium

RCC:

Radiochemical conversion

RCY:

Radiochemical yield

RCP:

Radiochemical purity

RP:

Reverse phase

rt:

Room temperature

TBA[18F]F:

tetra-n-butylammonium [18F]fluoride

TEA:

Triethylamine

TFA:

Trifluoroacetic acid

THF:

Tetrahydrofuran

TLC:

Thin-layer chromatography

TME:

Tumour microenvironment

t R :

retention time

UV:

Ultraviolet

References

  • Armarego WLF, Chai CLL. Purification of Laboratory Chemicals. 6th ed. Amsterdam; Butterworth-Heinemann: Oxford, UK: Elsevier; 2009.

    Google Scholar 

  • Badawy AAB. Kynurenine pathway of tryptophan metabolism: regulatory and functional aspects. Int J Tryptophan Res. 2017;10:1178646917691938.

    Article  PubMed  PubMed Central  Google Scholar 

  • Bouleau A, Lebon V, Truillet C. PET imaging of immune checkpoint protein in oncology. Pharmacol ther. 2021;222:107786.

    Article  CAS  PubMed  Google Scholar 

  • Charehjoo A, Majidpoor J, Mortezaee K. Indoleamine 21,3-dioxygenase 1 in circumventing checkpoint inhibitor responses: updated. Int Immunolpharmacol. 2023;118:110032.

    Article  CAS  Google Scholar 

  • Cheong JE, Sun L. Targeting the IDO1/TDO2-KYN-AhR pathway for cancer immunotherapy – challenges and opportunities. Trend Pharmacol Sci. 2018;39(3):307–25.

    Article  CAS  Google Scholar 

  • Chevallet P, Garrouste P, Malawska B, Martinez J. Facile synthesis of tert-butyl ester of N-protected amino acids with tert-butyl bromide. Tetrahedron Lett. 1993;34(46):7409–12.

    Article  CAS  Google Scholar 

  • Cortiula F, Reymen B, Peters S, Van Mol P, Wauters E, Vansteenkiste J, et al. Immunotherapy in unresectable stage III non-small-cell lung cancer: state of the art and novel therapeutic approaches. Anal Oncol. 2022;33(3):893–98.

    Article  CAS  Google Scholar 

  • Daste A, Larroquette M, Gibson N, Lasserre M, Domblides C. Immunotherapy for head and neck squamous cell carcinoma: current status and perspectives. Immunotherapy. 2024;16(3):187–97.

    Article  CAS  PubMed  Google Scholar 

  • Eliel EL. Infelicitous Stereochemical nomenclature. Chirality. 1997;9(5–6):428–30.

    Article  CAS  Google Scholar 

  • Fujiwara T, Yin B, Jin M, Kirk KL, Takeuchi Y. Synthetic studies of 3-(3-fluorooxindol-3-yl)-L-alanine. J Fluor Chem. 2008;129(9):829–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ge S, Jia T, Li J, Zhang B, Deng S, Sang S. Molecular imaging of immune checkpoints in oncology: current and future applications. Cancer Lett. 2022;548:215896.

    Article  CAS  PubMed  Google Scholar 

  • Godin-Ethier J, Hanafi LA, Piccirillo CA, Lapointe R. Indoleamine 2,3-dioxygenase expression in human cancers: clinical and immunologic perspectives. Clin Cancer res. 2011;17(22):6985–91.

    Article  CAS  PubMed  Google Scholar 

  • Guo Z, Zhang R, Yang AG, Zheng G. Diversity of immune checkpoints in cancer immunotherapy. Front Immunol. 2023;14:1121285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Haslam A, Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019;2(5):e192535.

    Article  PubMed  PubMed Central  Google Scholar 

  • Haveman LYF, Vugts DJ, Windhorst AD. State of the art procedures towards reactive [18F]fluoride in PET tracer synthesis. EJNMMI Radiopharm Chem. 2023;8(28):1–31.

    Google Scholar 

  • Henrottin J, Zervosen A, Lemaire C, Sapunaric F, Laurent S, Van den Eynde B, et al. N1-Fluoroalkyltryptophan analogues: synthesis and in vitro study as potential substrates for indoleamine 2,3-dioxygenase. ACS Med Chem Lett. 2015;6(3):260–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Henrottin J, Lemaire C, Egrise D, Zervosen A, Van den Eynde B, Plenevaux A, et al. Fully automated radiosynthesis of N1-[18F]fluoroethyl-tryptophan and study of its biological activity as a new potential substrate for indoleamine 2,3-dioxygenase PET imaging. Nucl Med Biol. 2016;43(6):379–89.

    Article  CAS  PubMed  Google Scholar 

  • Ibis B, Aliazis K, Cao C, Yenyuwadee S, Boussiotis VA. Immune-related adverse effects of checkpoint immunotherapy and implications for the treatment of patients with cancer and autoimmune diseases. Front Immunol. 2023:141197364.

  • Jiang H, Guo Y, Cai H, Viola N, Shields AF, Muzik O, Juhasz C. Automated radiosynthesis of 1-(2-[18F]fluoroethyl)-L-tryptophan ([18F]FETrp) for PET imaging of cancer in humans. J Label Comp Radiopharm. 2023;66(7–8):180–8.

    Article  CAS  Google Scholar 

  • John F, Muzik O, Mittal S, JuhĂ¡sz C. Fluorine-18-labeled PET radiotracers for imaging tryptophan uptake and metabolism: a systematic review. Mol Imaging Biol. 2020;22(4):805–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Junk L, Papadopoulos E, Kazmaier U. Tryptophan N1-alkylation: quick and simple access to diversely substituted tryptophans. Synthesis. 2021;53(14):2503–11.

    Article  CAS  Google Scholar 

  • Kim DW, Ahn DS, Oh YH, Lee S, Kil HS, Oh SJ, et al. A new class of Sn2 reactions catalyzed by protic solvents: facile fluorination for isotopic labeling of diagnostic molecules. J Am Chem Soc. 2006;128(50):16394–7.

    Article  CAS  PubMed  Google Scholar 

  • Kim HY, Lee JY, Lee YS, Jeong JM. Design and synthesis of enantiopure 18F-labelled [18F]trifluoromethyltryptophan from 2-halotryptophan derivatives via copper(I)-mediated [18F]trifluoromethylation and evaluation of its in vitro characterization for the serotonergic system imaging. J Label Comp Radiopharm. 2019;62(9):566–79.

    Article  CAS  Google Scholar 

  • Le Naour J, Galluzzi L, Zitvogel L, Kroemer G, Vacchelli E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology. 2020;9(1):1777625.

    Article  PubMed  PubMed Central  Google Scholar 

  • Liu C, Yang M, Zhang D, Chen M, Zhu D. Clinical cancer immunotherapy: current progress and prospects. Front Immunol. 2022;13:961805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Michelhaugh SK, Muzik O, Guastella AR, Klinger NV, Polin LA, Cai H, et al. Assessment of tryptophan uptake and kinetics using 1-(2-18F-fluoroethyl)-L-tryptophan and α-11C-methyl-L-tryptophan PET imaging in mice implanted with patient-derived brain tumor xenografts. J Nucl Med. 2017;58(2):208–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Michielin O, van Akkooi ACJ, Ascierto PA, Dummer R, Keilholz U. Cutaneous melanoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2019;30(12):1884–901.

    Article  CAS  PubMed  Google Scholar 

  • Morris VK, Kennedy EB, Baxter NN, Benson AB 3rd, Cercek A, Cho M, et al. Treatment of metasttic colorectal cancer: ASCO guideline. J Clin Oncol. 2023;41(3):678–700.

    Article  PubMed  Google Scholar 

  • Pang K, Shi ZD, Wei LY, Dong Y, Ma YY, Wang W et al. Research progress of therapeutic effects and drug resistance of immunotherapy based on PD-1/PD-L1 blockade. Drug Resist Updat. 2023:66100907.

  • Pilard C, Ancion M, Delvenne P, Jerusalem G, Hubert P, Herfs M. Cancer immunotherapy: it’s time to better predict patients’ response. Br J Cancer. 2021;125(7):927–38.

    Article  PubMed  PubMed Central  Google Scholar 

  • Platten M, Nollen EAA, Röhrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer neurodegeneration and beyond. Nat Rev drug Discov. 2019;18(5):379–401.

    Article  CAS  PubMed  Google Scholar 

  • Sankar K, Ye JC, Li Z, Zheng L, Song W, Hu-Lieskovan S. The role of biomarkers in personalized immunotherapy. Biomark Res. 2022;10:32.

    Article  PubMed  PubMed Central  Google Scholar 

  • Sun T, Tang G, Tian H, Wang X, Chen X, Chen Z, et al. Radiosynthesis of 1-[18F]fluoroethyl-L-tryptophan as a novel potential amino acid PET tracer. Appl Radiat Isot. 2012;70(4):676–80.

    Article  CAS  PubMed  Google Scholar 

  • Van de Donk PP, Kist de Ruijter L, Lub-de Hooge MN, Brouwers AH, van der Wekken AJ, Oosting SF, et al. Molecular imaging biomarkers for immune checkpoint inhibitor therapy. Theranostics. 2020;10(4):1708–18.

    Article  PubMed  PubMed Central  Google Scholar 

  • Wadsworth HJ, O’Shea D, Passmore J, Trigg W, Ewan A, Shan B. Tricyclic indole derivates as PBR ligands. 2010, Patent WO2010109007.

  • Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Wierstra P, Sandker G, Aarntzen E, Gotthardt M, Adema G, Bussink J, RaavĂ© R, Heskamp S. Tracers for non-invasive radionuclide imaging of immune checkpoint expression in cancer. EJNMMI Radiopharm Chem. 2019;4:29.

    Article  PubMed  PubMed Central  Google Scholar 

  • Xin Y, Cai H. Improved radiosynthesis and biological evaluations of L- and D-1-[18F]fluoroethyl-tryptophan for PET imaging of IDO-mediated kynurenine pathway of tryptophan metabolism. Mol Imaging Biol. 2017;19(4):589–98.

    Article  CAS  PubMed  Google Scholar 

  • Xin Y, Gao X, Liu L, Ge WP, Jain MK, Cai H. Evaluation of L-1-[18F]fluoroethyl-tryptophan for PET imaging of cancer. Mol Imaging Biol. 2019;21(6):1138–46.

    Article  CAS  PubMed  Google Scholar 

  • Xin Y, Yue X, Li H, Li Z, Cai H, Choudhary AK, et al. PET imaging of medulloblastoma with an 18F-labeled tryptophan analogue in a transgenic mouse model. Sci Rep. 2020;10(1):3800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Yu H, Zong Y, Xu T. Total synthesis of (-)penicimutanin a and related congeners. Chem Sci. 2020;11(3):656–60.

    Article  CAS  Google Scholar 

  • Yue X, Xin Y, Zhang S, Nikam R, Kandula V, Choudhary AK, Chugani HT, et al. Automated production of 1-(2-[18F]fluoroethyl)-L-tryptophan for imaging of tryptophan metabolism. Appl Radiat Isot. 2020;156:109022.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Prof. P. Chalard for optical rotation analyses and M. Leremboure (Mass Spectroscopy platform, UMR 6296 CNRS/UCA Partner, Clermont-Ferrand, France) for HRMS analyses.

Funding

This research received no external funding.

Author information

Authors and Affiliations

Authors

Contributions

AMB, JMC: conception and design of the study and major contributors in writing and editing the manuscript. JMC, BW: synthesis and characterisation of organic compounds. AMB: design and programming of the automated synthesis sequence. AMB, DK, JMC: investigation of radiosynthesis. DK, BW, EM, SL, KK: contribution to the writing, reviewing and editing of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jean-Michel Chezal.

Ethics declarations

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maisonial-Besset, A., Kryza, D., Kopka, K. et al. Improved automated one-pot two-step radiosynthesis of (S)-[18F]FETrp, a radiotracer for PET imaging of indoleamine 2,3-dioxygenase 1 (IDO1). EJNMMI radiopharm. chem. 9, 28 (2024). https://doi.org/10.1186/s41181-024-00256-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41181-024-00256-0

Keywords