Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Electricity-driven site-selective deuteration of pharmaceuticals

Minling Zhong a, Feihu Wang a, Guanqun Han a, Shuai Yuan b, Guodong Li a, De-en Jiang *b and Yujie Sun *a
aDepartment of Chemistry, University of Cincinnati, Cincinnati, OH 45221, USA. E-mail: yujie.sun@uc.edu
bDepartment of Chemical & Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA. E-mail: de-en.jiang@vanderbilt

Received 6th August 2025 , Accepted 2nd November 2025

First published on 3rd November 2025


Abstract

Precision deuteration at metabolically vulnerable sites of pharmaceuticals can enhance drug stability and therapeutic efficacy, yet existing methods often suffer from poor selectivity and inefficiency. Here, we report an electricity-driven bromine-mediated deuteration strategy that enables late-stage site-selective deuteration of pharmaceuticals using D2O as the deuterium source. This approach involves a two-step process: (i) bromination of labile C–H bonds using Br2, followed by (ii) electricity-driven deuterodebromination using a palladium membrane reactor. This design leverages in situ Br2 generation at the anode and selective deuterium permeation through the palladium membrane cathode, thereby significantly improving atom economy and energy efficiency. Our method achieves nearly complete conversion and >90% deuterium incorporation for a range of aryl, heteroaryl, benzylic, and unactivated alkyl bromides, including ten marketed drug molecules. Furthermore, gram-scale synthesis of D-clonidine demonstrates the scalability of this approach. By integrating high selectivity, broad substrate scope, and operational efficiency, this method offers a practical solution for deuterated drug synthesis, with potential applications in pharmaceutical development and metabolic stabilization.


Introduction

Isotopic substitution has long been recognized as a powerful tool in physical and medicinal chemistry, owing to the kinetic isotope effect that influences bond dissociation energies and reaction rates. Deuteration, the selective replacement of hydrogen with its heavier isotope deuterium, has emerged as a crucial strategy in drug development and metabolism studies.1,2 The incorporation of deuterium can significantly alter pharmacokinetic properties by enhancing metabolic stability, reducing toxicity, and prolonging drug half-life.3–5 Over the past few years, several deuterated pharmaceuticals have gained regulatory approval, such as deutetrabenazine (Austedo®),6 deuremidevir (VV116),7 and d1-(R)-pioglitazone (PXL065)8 shown in Fig. 1a. With at least 15 additional deuterated drug candidates currently in clinical trials,9 the pharmaceutical industry continues to explore deuteration as a means to develop safer and more efficacious therapeutics. To facilitate the synthesis of deuterated drugs, various deuteration strategies have been developed. A widely used approach involves deuterated building blocks (see representative examples in Fig. 1b), which introduce deuterium into molecules at early synthetic stages.10 While offering high isotopic enrichment, these methods typically rely on expensive and limited deuterated precursors and require multistep synthetic sequences, leading to high overall cost and operational complexity. Alternatively, direct hydrogen–deuterium exchange (H/D exchange) methods provide a more straightforward route to introduce deuterium into existing molecules. Although such methods can be effective, they often require D2 gas, elevated temperatures, or stoichiometric additives, and may suffer from low site selectivity, incomplete labeling, or over-deuteration of complex scaffolds. With the advancement of transition-metal catalysis, H/D exchange reactions catalyzed by Pd, Ir, Ru, Rh, and Fe have become well-established in synthetic chemistry.11–17 Nevertheless, most homogeneous catalytic systems depend on costly metals and sophisticated ligands and may raise environmental and sustainability concerns, thereby limiting their broader applicability.18
image file: d5sc05956a-f1.tif
Fig. 1 Development of deuteration strategies. (a) Representative deuterated pharmaceuticals. (b) Conventional deuteration strategies. (c) Various catalytic deuteration strategies. (d) Desirable deuteration sites in selected drugs. (e) Our strategy of electricity-driven site-selective deuteration.

To address these challenges, numerous catalytic deuteration strategies have been developed, enabling deuterium incorporation into complex molecules under milder conditions and with greater atom efficiency (Fig. 1c).19 For instance, enzymatic deuteration primarily relies on alcohol dehydrogenases and amine oxidases to achieve regioselective deuterium incorporation from D2O.20,21 Photocatalytic deuteration utilizes visible-light-activated catalysts to enable efficient H/D or X/D exchange,22–30 while electrocatalytic deuteration offers another modular and energy-efficient strategy.31–35 Although substantial progress has been achieved in recent years, the field continues to face intrinsic challenges related to catalyst cost, product purification, undesired side processes, and insufficient compatibility with pharmaceutically relevant functional groups. Thus, the development of a broadly applicable and efficient protocol for late-stage deuteration, particularly at metabolically sensitive sites within drug molecules, remains an outstanding goal in synthetic and medicinal chemistry.

Precision deuteration at metabolically soft positions is critical because many pharmaceuticals undergo rapid clearance via cytochrome P450-mediated hydroxylation.36,37 The strategic replacement of metabolically weak C–H bonds with C–D bonds can significantly modulate the rate of oxidative metabolism through the kinetic isotope effect, ultimately improving drug stability, prolonging systemic exposure, and reducing the required dosage and frequency of administration.38 Moreover, precise and selective deuterium incorporation can fine-tune the pharmacokinetic and pharmacodynamic profiles of drug candidates without altering their fundamental biological targets, providing a powerful means for optimizing clinical performance. Despite these benefits, existing deuteration methods remain inadequate for achieving site-specific isotopic modification in structurally complex drug molecules. Conventional approaches either require pre-installed deuterium at the target site, which limits flexibility in modifying existing drugs, or lead to non-selective over-deuteration, potentially altering molecular properties and introducing unknown pharmaceutical risks. In fact, the desirable deuteration sites of quite a few drug molecules have been determined with representative examples shown in Fig. 1d. However, the development of a general and efficient strategy capable of achieving site-selective deuteration at metabolically relevant positions remains challenging.

Herein, we present a bromine-mediated deuteration strategy that enables precise late-stage deuteration of various drug molecules. This approach consists of a two-step process as shown in Fig. 1e: (i) bromination of metabolically labile sites in drug molecules using Br2, followed by (ii) electricity-driven deuterodebromination in an electrochemical palladium membrane reactor (ePMR). The latter step takes advantage of the selective deuterium permeation from a D2O-based electrolyte into a separate reaction chamber where deuterodebromination takes places.39 Unlike conventional electrocatalysis, this spatially separated deuteration design eliminates product contamination from electrolytes, simplifying purification and improving isotopic purity. Additionally, our method leverages bromide oxidation at the anode, which reduces the required voltage input compared to conventional electrocatalytic deuteration systems usually using water oxidation as the counter reaction. Furthermore, the in situ generated Br2 at the anode can be extracted to brominate additional drug molecules, further enhancing atom economy and sustainability. Beyond enabling the precise deuteration of 10 deuterated drug molecules, our approach is equally applicable in producing a variety of deuterated building blocks. Finally, gram-scale synthesis of deuterated clonidine has been successfully demonstrated, highlighting its great promise and broad utility in pharmaceutical and synthetic chemistry applications.

Results and discussion

Confirmation of hydride transfer

Conventional electrocatalytic hydrodehalogenation (i.e., hydrogenolysis) typically proceeds via initial one-electron reduction of an organic halide, generating a carbon radical intermediate upon halide release. Subsequent proton-coupled electron transfer to this radical species leads to the formation of the hydrogenated product. A competing pathway involves radical dimerization, which can reduce the efficiency of the process. Alternatively, at highly negative potentials, direct two-electron reduction of organic halides generates carbanion intermediates, which subsequently undergo protonation to afford the hydrogenated product. However, such extreme potentials inevitably promote competitive proton reduction, leading to excessive H2 evolution (HER) and diminished faradaic efficiency. Regardless of the mechanistic pathway, hydrodehalogenation fundamentally requires a net 2e/H+ transfer, which can be conceptually framed as a formal hydride (H) transfer process. This perspective aligns with emerging electrochemical strategies that leverage controlled hydrogen atom manipulation to achieve selective transformations.

The ePMR has garnered significant attention for organic hydrogenation due to the rapid diffusion of hydrogen atom through the palladium lattice.40–43 However, previous studies predominantly focused on the hydrogenation of unsaturated substrates (e.g., alkenes, alkynes, aldehydes, and nitriles) via hydrogen atom transfer (HAT) from the palladium membrane electrode (Pdm). While effective for these transformations, HAT alone is insufficient for hydrodehalogenation, which requires an overall hydride transfer process. Given that hydrogen atom disproportionation has been proposed as a route to generate hydrides,44 our initial objective was to confirm the formation and utilization of a hydride species on the hydrogenation side of Pdm (opposite to its electrochemical interface) as illustrated in Fig. 2a. To probe this mechanism, we employed N-methylnicotinamide (NA+) as a hydride acceptor, drawing inspiration from the NAD+/NADH redox cycle. Constant-current electrolysis at −50 mA cm−2 was applied to the electrochemical chamber, facilitating proton/water reduction at the Pdm surface and generating hydrogen atoms. These hydrogen atoms subsequently permeated through the membrane into a separate chamber containing 5 mM NA+ in a phosphate buffer (pH 8) with 30% methanol (v/v). After 24 hours of electrolysis, the UV-visible absorption spectrum of the hydrogenation solution exhibited a pronounced absorption peak at 305 nm (Fig. 2b), indicative of NA+ reduction and subsequent dearomatization. Structural confirmation via1H NMR (Fig. 2c) and mass spectrometry (Fig. 2d) revealed the formation of 1-methylpiperidine-3-carboxamide, a product consistent with hydride transfer to NA+ followed by hydrogenation of two C[double bond, length as m-dash]C bonds—an overall one-hydride–four-hydrogen transfer process. To further validate the hydrogen source, we replaced H2O with D2O in the electrochemical chamber. This modification resulted in the incorporation of five deuterium atoms into the product (Fig. 2c and d), unambiguously confirming that the hydrides (or deuterides) originate from electrochemical H2O/D2O reduction on the Pdm surface. These findings demonstrate the feasibility of hydrogen/deuterium disproportionation in protic solvents using Pdm for hydride/deuteride transfer.


image file: d5sc05956a-f2.tif
Fig. 2 Hydrogenation of N-methylnicotinamide (NA+) using ePMR. (a) Proposed schematics of NA+ hydrogenation following a one-hydride–four-hydrogen transfer mechanism. (b) UV-visible absorption spectra of the hydrogenation solution. (c) 1H NMR spectra of the hydrogenation (top) and deuteration (bottom) solutions. (d) Mass spectroscopies of the hydrogenation (top) and deuteration (bottom) solutions.

Optimization of deuterodebromination

To evaluate the efficacy of the Pdm in facilitating electrocatalytic deuterodebromination, we employed D2O as a cost-effective deuterium source. 2-Bromoquinoline was selected as a model substrate due to the ubiquity of quinoline-based structural motifs in pharmaceuticals. Notably, the C2-position of the quinoline ring, present in the widely used antimalarial drug quinine, is metabolically susceptible to oxidation, making it a relevant target for selective deuteration.45 We first conducted 10-hour electrolysis at −50 mA cm−2 with the chemical chamber containing 20 mM 2-bromoquinoline in various solvents to determine the optimal conditions for deuterium incorporation. Due to the limited solubility of 2-bromoquinoline in water, we explored methanol as a protic solvent. As summarized in Fig. 3a, methanol enabled near-complete conversion (95%) of 2-bromoquinoline, with a deuterium incorporation efficiency of 89%. In contrast, aprotic solvents resulted in significantly lower yields: 72% in tetrahydrofuran and 37% in chloroform. A parallel investigation with 4-bromotoluene under identical conditions revealed that only methanol enabled full conversion (99%) and near-quantitative deuterium incorporation (99%), whereas the other solvents were ineffective. These results suggest that methanol plays a crucial role in promoting deuterodebromination using ePMR. To probe the reaction mechanism, 5 equivalents of 2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPO) were introduced as a radical scavenger into a methanol solution of 20 mM 2-bromoquinoline in the chemical chamber. After electrolysis at −50 mA cm−2 for 10 h, only TEMPO-H was detected by 1H NMR and HR-MS (Fig. S3 and S4). No TEMPO–quinoline adduct or radical dimerization products were observed, suggesting that the deuterobromination in ePMR system does not proceed through a radical pathway. As depicted in Fig. 3b, we hypothesize that methanol facilitates deuterium disproportionation on Pdm, leading to effective deuteride transfer to organic bromides. Additionally, in the case of 2-bromoquinoline, the nitrogen atom can be protonated, inducing the disproportionation process and enabling hydride transfer even in aprotic solvents.
image file: d5sc05956a-f3.tif
Fig. 3 Reaction development. (a) Yields and deuterium (D) incorporations after electrolysis at −50 mA cm−2 for 10 hours. Condition: two-compartment ePMR, Pt mesh as the anode, Pdm as the cathode, 1.0 M NaClO4 in D2O as the electrolyte, 2-bromoquinoline or 4-bromotoluene (20 mM) in different solvents inside the deuteration chamber. (b) Proposed mechanism for the CH3OH-assisted deuterodebromination of organic bromides using ePMR. (c and d) Calculated energy profiles of the hydrodebromination of (c) 2-bromoquinoline and (d) 4-bromotoluene on β-PdH surfaces in the absence and presence of CH3OH. (e) Time-dependent yield and deuterium incorporation. Default condition: two-compartment ePMR, Pt mesh as the anode, Pdm as the cathode, 1.0 M NaClO4 in D2O as the electrolyte, 2-bromoquinoline (20 mM) in CH3OH in the deuteration chamber, −50 mA cm−2. (f) Yield and deuterium incorporation of 2-bromoquinoline under different conditions. Condition 1: default condition, −50 mA cm−2 for 10 hours; condition 2: 10% H2O (v/v) added to the electrolyte solution; condition 3: 10% H2O (v/v) added to the deuteration chamber; condition 4: CD3OD as the solvent in the deuteration chamber.

To gain deeper mechanistic insights, we conducted density functional theory calculations to simulate the hydrogen-saturated Pdm for the hydrodebromination reaction by using surface models created from the bulk β-PdH structure (see SI for details). The results presented in Fig. 3c and d strongly support our hypothesis. In the absence of methanol, protonation of the nitrogen atom in 2-bromoquinoline by an H atom on the PdH surface occurs with an energy barrier of 0.53 eV, followed by a facile hydride transfer from the PdH surface with a barrier of 0.27 eV. When methanol is present, the protonation barrier is only slightly reduced (0.49 eV), and the resulting protonated 2-bromoquinoline is further stabilized via hydrogen bonding, while the hydrodebromination step requires a slightly higher activation energy of 0.68 eV. A striking contrast is observed for 4-bromotoluene, where direct hydride transfer is kinetically infeasible in the absence of methanol, requiring a high activation energy of 1.50 eV. However, introduction of methanol substantially reduces the overall energy barrier, with hydride transfer becoming the rate-determining step at a reduced energy barrier of 0.59 eV—even lower than that for 2-bromoquinoline. Collectively, the above experimental and computational findings establish that, in N-heteroaromatic systems, the substrate nitrogen induces H/D atom disproportionation. By contrast, in hydrocarbon substrates, methanol acts as a critical facilitator of H/D disproportionation, thereby enabling efficient hydride/deuteride transfer using Pdm. This mechanistic insight provides a strong foundation for realizing selective late-stage deuteration strategies in pharmaceuticals and beyond.

Given the solubility constraints of many organic bromides, methanol was chosen as the protic solvent in the deuteration chamber for all subsequent experiments unless otherwise specified. Electrolysis at −50 mA cm−2 using D2O as the electrolyte solvent resulted in a steady conversion of 2-bromoquinoline (20 mM) to quinoline-2-d, reaching >95% conversion after 10 hours (Fig. 3e). The initial deuterium incorporation exceeded 95%, but a slight decrease to ∼89% was observed over time, suggesting a minor source of hydrogen contamination. To systematically identify the origin of this contamination, we conducted control experiments (Fig. 3f). Introducing 10% H2O (v/v) into the D2O electrolyte (condition 2) significantly reduced deuterium incorporation to 37%, indicating that residual H2O in the electrolyte chamber plays a dominant role in hydrogen contamination. In contrast, adding 10% H2O to the deuteration chamber (condition 3) resulted in a less severe contamination, with deuterium incorporation decreasing to 76%. Notably, replacing methanol with CD3OD (condition 4) yielded the highest deuterium incorporation (95%), further confirming that methanol in the deuteration chamber may contribute to limited H/D exchange but to a much lesser extent than electrolyte contamination. Importantly, high conversion efficiencies (>95%) were observed under all conditions, regardless of the presence of H2O in either chamber. These results establish that adventitious H2O in the electrolyte chamber is the primary source of hydrogen contamination, whereas methanol in the deuteration chamber exhibits only a minor influence on isotopic purity.

Substrate scope

Encouraged by the efficient deuterodebromination of 2-bromoquinoline and 4-bromotoluene via ePMR, we sought to evaluate the generality of our strategy across a broad range of organic bromides (Fig. 4). Aryl bromides bearing electron-donating (e.g., methoxy, alkyl) and electron-withdrawing (e.g., ester, amide, chlorine) groups underwent efficient deuterodebromination under standard conditions (20 mM substrate, −50 mA cm−2, 10 h), yielding the corresponding deuterated arenes in high yields (1–9). 2-Bromoanthracene (10) also gave high deuterium incorporation under the standard conditions, despite its relatively low reactivity. Notably, dibromoaryl substrate 11 exhibited near-quantitative deuterium incorporation at both positions, demonstrating the strategy's efficacy for multi-site deuteration. Given the widespread presence of heteroaryl motifs in pharmaceuticals and bioactive molecules, we extended our investigation to pyridine (12–18), quinoline (19, 20), isoquinoline (21), and pyrimidine (22) derivatives. The reaction proved highly effective in most cases, delivering excellent conversion and high deuterium incorporation, highlighting its compatibility with heteroaryl-containing systems. Benzylic bromides, which are frequently encountered in pharmaceuticals and synthetic intermediates, also exhibited high reactivity under our conditions. A diverse set of substituted benzyl bromides (23–31) were smoothly deuterated, yielding the desired benzylic C–D bonds with high efficiency (75–99% yield, 88–99% deuterium incorporation). Unactivated alkyl bromides, often considered challenging substrates due to their low faradaic efficiency in electrochemical deuterodehalogenation,46 were also successfully deuterated. For instance, 1-bromo-4-phenylbutane (32) underwent smooth conversion, affording its deuterated product in 97% yield with 98% deuterium incorporation after 24-hour electrolysis. These results establish ePMR as a highly versatile platform for deuterodebromination, enabling site-selective deuteration across aryl, heteroaryl, benzylic, and unactivated alkyl bromides. This system was successfully applied to produce deuterated pharmaceutical building blocks, including deuterated benzoic acid (33),47p-toluic acid (34),48 and nicotinic acids (35–36),49,50 which can be employed as precursors to synthesize specifically deuterated drug molecules. The broad substrate compatibility, combined with high conversion efficiency and excellent isotopic purity, underscores the potential of this method for the synthesis of high-value deuterated building blocks—with significant implications for pharmaceuticals, agrochemicals, and materials science.
image file: d5sc05956a-f4.tif
Fig. 4 Scope of electricity-driven deuterodebromination of organic bromides using ePMR. Default condition: 20 mM substrate in CH3OH, −50 mA cm−2, 10 hours. Yield and deuterium incorporation (in red brackets) were determined from 1H NMR spectroscopy. at = 24 h. bt = 20 h.

Selective deuteration of pharmaceuticals

The broad substrate compatibility of our deuterodebromination strategy suggests strong potential for late-stage deuteration of drug molecules. Many pharmaceuticals undergo in vivo degradation through cytochrome P450 (CYP)-mediated oxidation, necessitating strategies to minimize metabolic clearance and enhance drug stability.36 Typically, the most labile C–H bonds, often termed metabolic soft sites, are the primary targets for CYP oxidation.37 Replacing these C–H bonds with C–D bonds provides a means to slow systemic clearance, reduce dosing frequency, and enhance pharmacological efficacy.38 Given that bromination may selectively activate these metabolically labile sites, we hypothesized that our two-step bromination–deuterodebromination approach would enable late-stage, site-selective deuteration of pharmaceuticals. To test this hypothesis, we selected ten marketed drugs whose metabolic vulnerabilities are well-documented but the specific deuteration at these sites has not been thoroughly investigated (Fig. 5).
image file: d5sc05956a-f5.tif
Fig. 5 Bromine-assisted selective deuteration of pharmaceuticals using ePMR. Default ePMR deuterodebromination condition: two-compartment ePMR, Pt mesh as the anode, Pdm as the cathode, 1.0 M NaClO4 in D2O as the electrolyte, 20 mM substrate in CH3OH in the deuteration chamber, −50 mA cm−2 for 10 hours. Conversion yields and deuterium incorporation ratios (in red brackets) were determined via1H NMR spectroscopy. a 10 mM substrate in CD3OD in the deuteration chamber, t = 96 h. b CH3OH/CH2Cl2 (v/v = 1/1) in the deuteration chamber. c 10 mM substrate in CH3OH/CH2Cl2 (v/v = 1/1) in the deuteration chamber.

Chlorzoxazone, a muscle relaxant and analgesic, is metabolized predominantly via 6-hydroxylation, a pathway linked to toxic aromatic oxirane intermediates.51 Bromination with Br2 in methanol (50 °C, 12 h) yielded the desired Br-chlorzoxazone (80%), which underwent smooth electrochemical deuterodebromination to afford D-chlorzoxazone with excellent yield (97%) and deuterium incorporation (97%). Similar results were observed for clonidine, an antihypertensive drug primarily metabolized via CYP-mediated 4-hydroxylation.52 Treatment with 0.6 equivalents of Br2 in methyl tert-butyl ether (MTBE) produced Br-clonidine (80%), which, under electrochemical deuterodebromination, furnished D-clonidine (97% yield, 95% D-incorporation). Diclofenac, a non-steroidal anti-inflammatory drug (NSAID) with a short plasma half-life, undergoes metabolism at the C5-position, forming 5-hydroxy-diclofenac as its primary phase I metabolite.53 Bromination of diclofenac methyl ester selectively produced 5-Br-diclofenac methyl ester, which was further converted to D-diclofenac methyl ester under the default electrochemical deuterodebromination condition. Salicylic acid, a precursor to aspirin54 and a plant hormone,55 can also be used to treat various skin disorders.56 Bromination with Br2 in glacial acetic acid at 60 °C for 12 h afforded Br-salicylic acid in 80% yield. Despite the presence of phenolic hydroxyl and carboxyl groups, subsequent electrochemical deuterodebromination in CD3OD successfully delivered D-salicylic acid with 94% yield and 80% deuterium incorporation. Caffeine, a widely consumed stimulant, is primarily metabolized via CYP-mediated demethylation, generating paraxanthine, which subsequently undergoes further oxidation before excretion.57,58 To extend its pharmacokinetic half-life, our strategy selectively incorporated deuterium at the 8′-position, yielding D-caffeine (88% yield, 90% D-incorporation). For drugs containing two metabolically vulnerable positions, such as imipramine59 and dapsone,60 our strategy successfully enabled double deuteration at both target sites, achieving high selectivity and efficiency.

Beyond aromatic C(sp2)–H oxidation, benzylic C(sp3)–H hydroxylation is another prevalent metabolic pathway (Fig. 5b). For example, celecoxib undergoes benzyl hydroxylation,61 a process that can be mitigated by deuterium substitution. Electrochemical deuterodebromination of Br-celecoxib yielded D-celecoxib with 99% yield and 98% deuterium incorporation. Similarly, ibuprofen62 and mexiletine,63 both metabolized at benzylic positions, underwent efficient site-selective deuteration under our optimized conditions. Collectively, these results highlight the broad utility, robustness, and adaptability of our site-selective electricity-driven deuteration strategy across diverse drug scaffolds, establishing it as a promising tool for pharmaceutical development and metabolic optimization.

Gram-scale synthesis of D-clonidine

The efficacy and scalability of our two-step bromination–deuterodebromination strategy were demonstrated in the gram-scale synthesis of deuterated clonidine (D-clonidine). As illustrated in Fig. 6a, bromide oxidation (4.0 M NaBr in electrolyte) at the anode generated Br2in situ, as evidenced by the increase of cell voltage and the characteristic color change observed in the anodic chamber (Fig. S3 and S4). The electrochemically generated Br2 was subsequently extracted with methyl tert-butyl ether and used for selective bromination of clonidine at room temperature, yielding Br-clonidine in 80% yield. Simultaneously, in the deuteration chamber, Br-clonidine (0.1 M in methanol) underwent constant-current electrochemical deuterodebromination using D2O as the deuterium source, affording D-clonidine in 97% yield with 95% deuterium incorporation (0.9 g). The final product required only methanol removal for isolation and purification, streamlining the synthesis. This strategy not only maximizes atom economy by continuously recycling bromine but also enhances energy efficiency. As shown in Fig. 6b, in the absence of NaBr, water oxidation requires a high cell voltage of 3.36 V to achieve 10 mA cm−2. However, in the presence of 4.0 M NaBr, bromide oxidation enables the same current density at a significantly reduced voltage of 2.13 V, saving over 1.2 V in energy input. These results establish our electrocatalytic bromination–deuterodebromination strategy as a practical and energy-efficient approach for scalable late-stage deuteration, with direct implications for the synthesis of isotopically labeled pharmaceuticals.
image file: d5sc05956a-f6.tif
Fig. 6 Two-step bromination-deuterodebromination for the synthesis of D-clonidine. (a) Simplified scheme of ePMR coupled with on-site bromination. Pt mesh as the anode, Pdm as the cathode; the anodic and cathodic chambers are separated by proton-exchange membrane (PEM). The electrolyte is 0.5 M D2SO4 in D2O, with additional 4.0 M NaBr in the anolyte. (b) Linear sweep voltammograms collected in the absence and presence of 4.0 M NaBr in the anode chamber. Condition: two-compartment ePMR, Pt mesh as the anode, Pdm as the cathode, 0.5 M NaClO4 in H2O as the electrolyte, scan rate = 100 mV s−1.

Conclusion

This work presents a selective, scalable, and energy-efficient strategy for precise deuterium incorporation into diverse molecular frameworks. By integrating electricity-driven deuterodebromination with in situ Br2 generation in a palladium membrane reactor, our method enables site-selective deuteration using D2O as the deuterium source. It enhances atom economy, energy efficiency, achieving nearly complete conversion with over 90% deuterium incorporation across aryl, heteroaryl, benzylic, and unactivated alkyl substrates while simplifying product purification. The successful gram-scale synthesis of D-clonidine further demonstrates its scalability. Its broad functional group compatibility and applicability to pharmaceuticals make it a powerful tool for metabolic stabilization and drug development. As demand for deuterated compounds grows, this method offers a practical and versatile solution for high-precision deuteration.

Author contributions

Y. S. conceived the idea and supervised the project. M. Z., F. W., G. H., and G. L. performed experimental work and data analysis. S. Y. and D. J. provided the computational support. M. Z. and Y. S. wrote the manuscript.

Conflicts of interest

The authors declare the following competing financial interest(s): Y. S. has filed a US provisional patent application no. 63/843964 (filed July 14, 2025) on the work presented in this manuscript. The remaining authors declare no other competing interests.

Data availability

Further queries about the data can be directed to the corresponding authors.

The data supporting this article have been included as part of the supplementary information (SI). Supplementary information is available. See DOI: https://doi.org/10.1039/d5sc05956a.

Acknowledgements

Y. S. thanks support from the US National Science Foundation (CBET-2411506) and the Petroleum Research Foundation (66635-ND3). D. J. thanks the US National Science Foundation (CHE-2245564).

References

  1. J. Atzrodt, V. Derdau, W. J. Kerr and M. Reid, Deuterium-and tritium-labelled compounds: applications in the life sciences, Angew. Chem., Int. Ed., 2018, 57, 1758–1784 CrossRef CAS PubMed.
  2. T. Pirali, M. Serafini, S. Cargnin and A. A. Genazzani, Applications of deuterium in medicinal chemistry, J. Med. Chem., 2019, 62, 5276–5297 CrossRef CAS.
  3. E. M. Isin, C. S. Elmore, G. N. Nilsson, R. A. Thompson and L. Weidolf, Use of radiolabeled compounds in drug metabolism and pharmacokinetic studies, Chem. Res. Toxicol., 2012, 25, 532–542 Search PubMed.
  4. A. Katsnelson, Heavy drugs draw heavy interest from pharma backers, Nat. Med., 2013, 19, 656 CrossRef CAS PubMed.
  5. B. Belleau, J. Burba, M. Pindell and J. Reiffenstein, Effect of deuterium substitution in sympathomimetic amines on adrenergic responses, Science, 1961, 133, 102–104 CrossRef CAS PubMed.
  6. C. Schmidt, First deuterated drug approved, Nat. Biotechnol., 2017, 35, 493–494 CrossRef CAS PubMed.
  7. Z. Cao, W. Gao, H. Bao, H. Feng, S. Mei, P. Chen and R. Zhao, VV116 versus nirmatrelvir–ritonavir for oral treatment of Covid-19, N. Engl. J. Med., 2023, 388, 406–417 CrossRef CAS PubMed.
  8. V. Jacques, S. Bolze, S. Hallakou-Bozec, A. W. Czarnik, A. S. Divakaruni, P. Fouqueray, A. N. Murphy, L. H. Van der Ploeg and S. DeWitt, Deuterium-stabilized (R)-pioglitazone (PXL065) Is responsible for pioglitazone efficacy in NASH yet exhibits little to no PPARγ activity, Hepatol. Commun., 2021, 5, 1412–1425 CrossRef CAS PubMed.
  9. R. M. C. Di Martino, B. D. Maxwell and T. Pirali, Deuterium in drug discovery: progress, opportunities, and challenges, Nat. Rev. Drug Discovery, 2023, 22, 562–584 CrossRef CAS PubMed.
  10. J. Steverlynck, R. Sitdikov and M. Rueping, The deuterated “magic methyl” group: A guide to site-selective trideuteromethyl incorporation and labeling by using CD3 reagents, Chem. – Eur. J., 2021, 27, 11751–11772 CrossRef CAS.
  11. C. Teja, S. Kolb, P. Colonna, J. Grover, S. Garcia-Argote, G. K. Lahiri, G. Pieters, D. B. Werz and D. Maiti, Deuteration and Tritiation of Pharmaceuticals by Non-Directed Palladium-Catalyzed C−H Activation in Heavy and Super-Heavy Water, Angew. Chem., Int. Ed., 2024, 63, e202410162 CrossRef CAS PubMed.
  12. L. V. Hale and N. K. Szymczak, Stereoretentive deuteration of α-chiral amines with D2O, J. Am. Chem. Soc., 2016, 138, 13489–13492 CrossRef CAS PubMed.
  13. M. Zhang, X. A. Yuan, C. Zhu and J. Xie, Deoxygenative deuteration of carboxylic acids with D2O, Angew. Chem., Int. Ed., 2019, 58, 312–316 CrossRef CAS PubMed.
  14. R. Pony Yu, D. Hesk, N. Rivera, I. Pelczer and P. J. Chirik, Nature, 2016, 529, 195–199 CrossRef CAS.
  15. W. Li, J. Rabeah, F. Bourriquen, D. Yang, C. Kreyenschulte, N. Rockstroh, H. Lund, S. Bartling, A.-E. Surkus, K. Junge, A. Brückner, A. Lei and M. Beller, Scalable and selective deuteration of (hetero) arenes, Nat. Chem., 2022, 14, 334–341 CrossRef CAS.
  16. Q. K. Kang, Y. Li, K. Chen, H. Zhu, W. Q. Wu, Y. Lin and H. Shi, Rhodium-catalyzed stereoselective deuteration of benzylic C–H bonds via reversible η6-coordination, Angew. Chem., Int. Ed., 2022, 61, e202117381 CrossRef CAS.
  17. J. Dey, S. Kaltenberger and M. van Gemmeren, Palladium (II)-catalyzed nondirected late-stage C(sp2)−H deuteration of heteroarenes enabled through a multi-substrate screening approach, Angew. Chemie Int. Ed., 2024, 63, e202404421 CrossRef CAS.
  18. S. Kopf, F. Bourriquen, W. Li, H. Neumann, K. Junge and M. Beller, Recent developments for the deuterium and tritium labeling of organic molecules, Chem. Rev., 2022, 122, 6634–6718 CrossRef CAS.
  19. W. Ou, C. Qiu and C. Su, Photo-and electro-catalytic deuteration of feedstock chemicals and pharmaceuticals: A review, Chinese J. Catal., 2022, 43, 956–970 CrossRef CAS.
  20. J. S. Rowbotham, M. A. Ramirez, O. Lenz, H. A. Reeve and K. A. Vincent, Bringing biocatalytic deuteration into the toolbox of asymmetric isotopic labelling techniques, Nat. Commun., 2020, 11, 1454 CrossRef CAS.
  21. T. J. Doyon and A. R. Buller, Site-selective deuteration of amino acids through dual-protein catalysis, J. Am. Chem. Soc., 2022, 138, 7327–7336 CrossRef.
  22. Y. Y. Loh, K. Nagao, A. J. Hoover, D. Hesk, N. R. Rivera, S. L. Colletti, I. W. Dacies and D. W. MacMillan, Photoredox-catalyzed deuteration and tritiation of pharmaceutical compounds, Science, 2017, 358, 1182–1187 CrossRef CAS PubMed.
  23. C. Liu, Z. Chen, C. Su, X. Zhao, Q. Gao, G.-H. Ning, H. Zhu, W. Tang, K. Leng, W. Fu, B. Tian, X. Peng, J. Li, Q.-H. Xu, W. Zhou and K. P. Loh, Controllable deuteration of halogenated compounds by photocatalytic D2O splitting, Nat. Commun., 2018, 9, 80 CrossRef.
  24. Z. Zhang, C. Qiu, Y. Xu, Q. Han, J. Tang, K. P. Loh and C. Su, Semiconductor photocatalysis to engineering deuterated N-alkyl pharmaceuticals enabled by synergistic activation of water and alkanols, Nat. Commun., 2020, 11, 4722 Search PubMed.
  25. Y. Li, Z. Ye, Y. M. Lin, Y. Liu, Y. Zhang and L. Gong, Organophotocatalytic selective deuterodehalogenation of aryl or alkyl chlorides, Nat. Commun., 2021, 12, 2894 CrossRef CAS.
  26. R. Zhou, L. Ma, X. Yang and J. Cao, Recent advances in visible-light photocatalytic deuteration reactions, Org. Chem. Front., 2021, 8, 426–444 RSC.
  27. Q. Shi, M. Xu, R. Chang, D. Ramanathan, B. Peñin, I. Funes-Ardoiz and J. Ye, Visible-light mediated catalytic asymmetric radical deuteration at non-benzylic positions, Nat. Commun., 2022, 13, 4453 CrossRef CAS.
  28. N. Li, J. Li, M. Qin, J. Li, J. Han, C. Zhu, W. Li and J. Xie, Highly selective single and multiple deuteration of unactivated C (sp3)-H bonds, Nat. Commun., 2022, 13, 4224 CrossRef CAS.
  29. B. Zhang, Z. Zhang, Y. Wang and D. Zhao, Late-stage deuteration and tritiation through bioinspired cooperative hydrogenolysis, Nat. Synth., 2025, 4, 444–452 CrossRef CAS.
  30. J. Xu, J. Fan, Y. Lou, W. Xu, Z. Wang, D. Li, H. Zhou, X. Lin and Q. Wu, Light-driven decarboxylative deuteration enabled by a divergently engineered photodecarboxylase, Nat. Commun., 2021, 12, 3983 CrossRef CAS.
  31. P. L. Norcott, Current electrochemical approaches to selective deuteration, Chem. Commun., 2022, 58, 2944–2953 RSC.
  32. D. Wood and S. Lin, Deuterodehalogenation under net reductive or redox-neutral conditions enabled by paired electrolysis, Angew. Chem., Int. Ed., 2023, 62, e202218858 CrossRef CAS.
  33. C. Liu, S. Han, M. Li, X. Chong and B. Zhang, Electrocatalytic deuteration of halides with D2O as the deuterium source over a copper nanowire arrays cathode, Angew. Chem. Int. Ed., 2020, 59, 18527–18531 CrossRef CAS.
  34. R. Li, Y. Wu, C. Wang, M. He, C. Liu and B. Zhang, One-pot H/D exchange and low-coordinated iron electrocatalyzed deuteration of nitriles in D2O to α,β-deuterio aryl ethylamines, Nat. Commun., 2022, 13, 5951 CrossRef.
  35. F. Bu, Y. Deng, J. Xu, D. Yang, Y. Li, W. Li and A. Lei, Electrocatalytic reductive deuteration of arenes and heteroarenes, Nature, 2024, 634, 592–599 CrossRef CAS PubMed.
  36. F. P. Guengerich, Cytochrome P450 and chemical toxicology, Chem. Res. Toxicol., 2008, 21, 70–83 Search PubMed.
  37. K. L. Drew and J. Reynisson, The impact of carbon–hydrogen bond dissociation energies on the prediction of the cytochrome P450 mediated major metabolic site of drug-like compounds, Eur. J. Med. Chem., 2012, 56, 48–55 CrossRef CAS.
  38. Z. Zhang and W. Tang, Drug metabolism in drug discovery and development, Acta Pharm. Sin. B, 2018, 8, 721–732 CrossRef.
  39. G. Han, G. Li and Y. Sun, Electrocatalytic hydrogenation using palladium membrane reactors, JACS Au., 2024, 4, 328–343 CrossRef CAS.
  40. H. Inoue, T. Abe and C. Iwakura, Successive hydrogenation of styrene at a palladium sheet electrode combined with electrochemical supply of hydrogen, Chem. Commun., 1996, 55–56 RSC.
  41. R. S. Sherbo, R. S. Delima, V. A. Chiykowski, B. P. MacLeod and C. P. Berlinguette, Complete electron economy by pairing electrolysis with hydrogenation, Nat. Catal., 2018, 1, 501–507 CrossRef CAS.
  42. A. Kurimoto, R. S. Sherbo, Y. Cao, N. W. Loo and C. P. Berlinguette, Electrolytic deuteration of unsaturated bonds without using D2, Nat. Catal., 2020, 3, 719–726 CrossRef CAS.
  43. G. Han, G. Li and Y. Sun, Electrocatalytic dual hydrogenation of organic substrates with a Faradaic efficiency approaching 200%, Nat. Catal., 2023, 6, 224–233 CrossRef CAS.
  44. A. Kurimoto, S. A. Nasseri, C. Hunt, M. Rooney, D. J. Dvorak, N. E. LeSage, R. P. Jansonius, S. G. Withers and C. P. Berlinguette, Bioelectrocatalysis with a palladium membrane reactor, Nat. Commun., 2023, 14, 1814 CrossRef CAS PubMed.
  45. S. R. Marcsisin, X. Jin, T. Bettger, N. McCulley, J. C. Sousa, G. D. Shanks, B. L. Tekwani, R. Sahu, G. A. Reichard, R. J. Sciotti, V. Melendez and B. S. Pybus, CYP450 phenotyping and metabolite identification of quinine by accurate mass UPLC-MS analysis: a possible metabolic link to blackwater fever, Malar. J., 2013, 12, 1–8 CrossRef PubMed.
  46. P. Li, C. Guo, S. Wang, D. Ma, T. Feng, Y. Wang and Y. Qiu, Facile and general electrochemical deuteration of unactivated alkyl halides, Nat. Commun., 2022, 13, 3774 CrossRef CAS.
  47. J. R. Widhalm and N. A. Dudareva, familiar ring to it: biosynthesis of plant benzoic acids, Mol. Plant, 2015, 8, 83–97 CrossRef CAS PubMed.
  48. B. Schweitzer-Chaput, M. A. Horwitz, E. de Pedro Beato and P. Melchiorre, Photochemical generation of radicals from alkyl electrophiles using a nucleophilic organic catalyst, Nat. Chem., 2019, 11, 129–135 CrossRef CAS PubMed.
  49. B. V. Varun, K. Vaithegi, S. Yi and S. B. Park, Nature-inspired remodeling of (aza) indoles to meta-aminoaryl nicotinates for late-stage conjugation of vitamin B3 to (hetero) arylamines, Nat. Commun., 2020, 11, 6308 CrossRef CAS.
  50. Z. Y. You, Y. J. Chen, Y. Y. Wang and C. Chen, Synthesis of Deuterium Labeled Standards of 1-Benzylpiperazine, Fenetylline, Nicocodeine and Nicomorphine, J. Chin. Chem. Soc., 2008, 55, 663–667 CrossRef CAS.
  51. C. Sun, M. Zhang, C. Guan, W. Li, Y. Peng and J. Zheng, In vitro and in vivo metabolic activation and hepatotoxicity of chlorzoxazone mediated by CYP3A, Arch. Toxicol., 2024, 98, 1095–1110 CrossRef CAS PubMed.
  52. S. Amna, T. Øhlenschlæger, E. A. Sædder, J. V. Sigaard and T. K. Bergmann, Review of clinical pharmacokinetics and pharmacodynamics of clonidine as an adjunct to opioids in palliative care, Basic Clin. Pharmacol. Toxicol., 2024, 134, 485–497 CrossRef CAS PubMed.
  53. M. Syed, C. Skonberg and S. H. Hansen, Mitochondrial toxicity of diclofenac and its metabolites via inhibition of oxidative phosphorylation (ATP synthesis) in rat liver mitochondria: Possible role in drug induced liver injury (DILI), Toxicol. in Vitro, 2016, 31, 93–102 CrossRef CAS.
  54. D. Ekinci, M. Şentürk and Ö. İ. Küfrevioğlu, Salicylic acid derivatives: synthesis, features and usage as therapeutic tools, Expert Opin. Ther. Pat., 2011, 21, 1831–1841 CrossRef CAS PubMed.
  55. P. Ding and Y. Ding, Stories of salicylic acid: a plant defense hormone, Trends Plant Sci., 2020, 25, 549–565 CrossRef CAS PubMed.
  56. T. Arif, Salicylic acid as a peeling agent: a comprehensive review. Clin. Cosmet, Investig. Dermatol., 2015, 455–461 CAS.
  57. G. Ngueta, Caffeine and caffeine metabolites in relation to hypertension in US adults, Eur. J. Clin. Nutr, 2020, 74, 77–86 CrossRef CAS.
  58. C. Willson, The clinical toxicology of caffeine: A review and case study, Toxicol. Rep., 2018, 5, 1140–1152 CrossRef CAS.
  59. W. Z. Potter and H. K. Manji, Antidepressants, metabolites, and apparent drug resistance, Clin. Neuropharmacol., 1990, 13, S45–S53 CrossRef PubMed.
  60. G. Wozel and C. Blasum, Dapsone in dermatology and beyond, Arch. Dermatol., 2014, 306, 103–124 CrossRef CAS PubMed.
  61. Y. A. Siu, M. H. Hao, V. Dixit and W. G. Lai, Celecoxib is a substrate of CYP2D6: Impact on celecoxib metabolism in individuals with CYP2C9* 3 variants, Drug Metab. Pharmacokinet., 2018, 33, 219–227 CrossRef CAS.
  62. K. D. Rainsford, Ibuprofen: pharmacology, efficacy and safety, Inflammopharmacology, 2009, 17, 275–342 CrossRef CAS.
  63. M. Nakajima, K. Kobayashi, N. Shimada, S. Tokudome, T. Yamamoto and Y. Kuroiwa, Involvement of CYP1A2 in mexiletine metabolism, Br. J. Clin. Pharmacol., 1998, 46, 55–62 CrossRef CAS PubMed.

Footnote

Equally contributed.

This journal is © The Royal Society of Chemistry 2026
Click here to see how this site uses Cookies. View our privacy policy here.