We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Histamine H2 receptor radioligands: triumphs and challenges

    Steffen Pockes

    *Author for correspondence:

    E-mail Address: steffen.pockes@ur.de

    Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg, D-93053, Germany

    Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA

    Department of Medicinal Chemistry, Institute for Therapeutics Discovery & Development, University of Minnesota, Minneapolis, MN 55414, USA

    &
    Katharina Tropmann

    Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg, D-93053, Germany

    Published Online:https://doi.org/10.4155/fmc-2021-0058

    Abstract

    Since the discovery of the histamine H2 receptor (H2R), radioligands were among the most powerful tools to investigate its role and function. Initially, radiolabeling was used to investigate human and rodent tissues regarding their receptor expression. Later, radioligands gained increasing significance as pharmacological tools in in vitro assays. Although tritium-labeling was mainly used for this purpose, labeling with carbon-14 is preferred for metabolic studies of drug candidates. After the more-or-less successful application of numerous labeled H2R antagonists, the recent development of the G protein-biased radioligand [3H]UR-KAT479 represents another step forward to elucidate the widely unknown role of the H2R in the central nervous system through future studies.

    For more than 50 years now, the histamine H2 receptor (H2R) has been subject of drug research in academia and the pharmaceutical industry. While in 1966 Ash and Schild speculated about its existence [1], the SK&F research group around Sir James W Black was able to prove the presence of the H2R in 1972 with their experiments on histamine-induced gastric acid release [2]. From today's perspective, a rather small number of 700 tested compounds within 9 years of drug development (1964–1972) [3] led via burimamide and metiamide to the world's first blockbuster drug, cimetidine (Figure 1). Cimetidine is known to be the first H2 receptor antagonist on the market in the treatment of peptic ulcer and gastroesophageal reflux disease [3,4]. To date, five additional H2R antagonists (ranitidine [5], famotidine [6], nizatidine [7], roxatidine [8] and lafutidine [9]; Figure 1) with increased potency and better pharmacokinetic properties have been successfully approved [10]. In contrast, H2 receptor agonists have not yet found their way into the world's drug portfolio. Although a large number of highly potent and subtype-selective agonists has already been published, these ligands are predominantly applied in basic research studies within academia [11–18]. However, the aforementioned properties turn them into valuable pharmacological tools that could be of great importance to elucidate the largely unknown role of the H2R in the central nervous system (CNS) [3,15]. Such molecules would be an important addition as counterparts to zolantidine (Figure 1), the only described CNS-penetrable H2R antagonist until today [19]. In addition to this application, there are several studies discussing possible peripheral (acute myeloid leukemia) [14,20] and central (memory and learning) [21,22] therapeutic indications for H2R agonists, although cardiac and gastric side effects should be considered. In particular, the effect on learning and memory occurs via the stimulation of postsynaptic H2Rs, which so far have only been shown via the use of dual-acting acetylcholinesterase inhibitors and H3R antagonists as these molecules initiate this process through inhibition of presynaptic H3-autoreceptors [21,22]. For that reason, the use of CNS-penetrating H2R agonists is of great interest.

    Figure 1. Structures of histamine H2 receptor radioligands and the precursor BMY25368.

    For impromidine and lafutidine, the exact location of the tritium label was not indicated in the literature. BMY-25368 has not been synthesized in radiolabeled form.

    *3H-labeled position(s); **14C-labeled position(s); ***35S-labeled position; ****125I-labeled position.

    Numerous publications with elaborate pharmacological studies and dedicated research were necessary to write the preceding story of success about the H2R. In this regard, radiolabeled compounds played an important role for the examination of histamine receptor expression in the different tissues of the human body. Moreover, characterizations of ligand–receptor interactions and metabolic studies for potential drug candidates were facilitated. Although the number of publications of modern FRET/BRET-based methods for G-protein-coupled receptors (GPCRs) is increasing rapidly, radiolabeled compounds are still an integral part of the pharmacological repertoire of medicinal chemists.

    Radiolabeling

    In principle, there are several ways to introduce a radioisotope into a molecule. First, a decision has to be made on the choice of the isotope. Due to the high prevalence of hydrogen and carbon in organic compounds, their isotopes – namely, tritium (3H) and carbon-14 (14C) – are the first to be considered for labeling. On the other hand, compounds with radioactive isotopes of phosphorus (32P, 33P), sulfur (35S) and iodine (125I) are less frequently encountered. Except for the gamma emitter iodine-125, all isotopes listed here are beta emitters (negative beta decay).

    The decisive factor for the choice of the specific radioisotope is the type of study which should be performed. It is of particular importance whether the radioligand should be applied for a preliminary in vitro characterization in the early phase of drug development or whether the drug candidate is already in a preclinical phase and a metabolic study (e.g., human ADME study) is required. Therefore, depending on the purpose, a thorough assessment of the situation must be made to decide which labeling is the right one. Excellent guidance to make a decision between 3H- and 14C-labeling is provided in the review article by Joel A Krauser, which served as a reference for some of the following points [23]. When comparing the radiochemical properties of the two isotopes, it is obvious that tritium has a significantly higher specific activity (3H: 28.7 Ci/mmol vs 14C: 0.0624 Ci/mmol) is cheaper to acquire and more readily available. In addition, the synthesis of tritiated compounds is much simpler since nowadays the labelling reagents are more easily to handle. Whereas tritium gas was used almost exclusively in the past, highly reactive N-hydroxysuccinimide (NHS) esters are now available, most of which containing a tritiated alkylic side chain. Because of their nongaseous state, these NHS esters can be used easily with exact stoichiometry delivering very high yields due to their reactivity. A disadvantage compared with tritium gas is certainly the dependence of commercially available tritiated side chains of NHS esters, which may not be compatible with the structure of an already existing drug candidate. In addition to the significantly lower acquisition costs, the lower half-life (3H: 12.32 vs 14C: 5730 years) also allows simpler and more economical management of radioactive waste disposal. All these aspects provide reasons to use tritiated compounds in the early phase of drug development, which is why academia works almost exclusively with [3H]radioligands. Although 3H-labeled compounds possess high radiochemical stability, there is a high risk of biological degradation by oxidative cleavage of the 3H label mediated by cytochrome P450 enzymes. Because 14C is integrated into the molecular scaffold, there is a reduced risk of direct label cleavage, which is beneficial for metabolic studies. Furthermore, the isotopic signature of 14C can be analyzed and detected very specifically by mass spectrometry. Conversely, the aforementioned metabolic instability of 3H-labeled compounds inevitably negates their use in such studies. Accordingly, the revealed profile of carbon-14 labeling leads to the fact that studies with 14C-labeled compounds can be predominantly found in the literature in preclinical studies of the pharmaceutical industry. Nevertheless, one should not blindly follow this classification but rather make a detailed and individual evaluation when choosing the right label related to the respective project. Compounds labeled with 35S are rare, certainly also due to the low sulfur content in drugs. However, the application of this isotope is often found in functional GPCR assays, in which [35S]GTPγS is used. 35S is also a beta emitter and possesses a much lower half-life of 87.1 days compared with 3H and 14C. The isotopes 32P and 33P are mainly used to label guanosine-5′-triphosphate (GTP) in functional assays, for example, the [γ-32P]-GTPase or [γ-33P]-GTPase assay, respectively. Both isotopes have extremely short half-lives (32P: 14.3 and 33P: 25.4 days) but show significant differences in decay energy. While phosphorus-32 has a comparatively high decay energy of 1.71 MeV, the energy beta emission of phosphorus-33 is only 0.25 MeV.

    Radioligands for the histamine H2 receptor

    The most versatile molecular tools which successfully promoted pharmacological research on histamine receptors were radioligands. Therefore, in the early 1980s, the well-established H2R antagonists [3H]cimetidine and [3H]ranitidine (Figure 1) were mainly employed [24–27]. However, the general drawback of these ligands was their comparatively low affinity for the H2R (Table 1) despite their high efficacy [28–30]. In addition to the described effect at the H2R, [3H]cimetidine also exhibited considerable affinity for another imidazole recognition site, which precluded selective investigations of the H2R [24,25]. Although [3H]tiotidine (Figure 1) is routinely used in biological in vitro assays, it was reported to have a very high nonspecific binding and to address only a subpopulation of the H2R [31,32]. Similar problems were faced in studies with the endogenous ligand [3H]histamine (Figure 1), which was also found to have low affinity (Table 1) and high nonspecific binding [15,33]. Consequently, large quantities of radioligand had to be used to obtain meaningful results leading to higher risks to health and the environment, as well as inefficiency. Furthermore, [3H]histamine does not exhibit any selectivity within the histamine receptor family showing even higher affinities at the H3R and H4R. This lack of selectivity automatically disqualified [3H]histamine for selective studies at the H2R [15]. The same disadvantage applied for [3H]impromidine (Figure 1), rendering this agonist unsuitable as an appropriate H2R radioligand, as well [34]. Unfortunately, also radiolabeling of the antagonist [3H]ICIA 5165 (Figure 1), an analog of tiotidine, did not lead to any significant improvements over the drawbacks already mentioned [35].

    Table 1. Binding data (pKi values) of selected H2R ligands at the human histamine H2 receptor.
    CompoundpKiRef.
     hH1RhH2RhH3RhH4R 
    Histamine4.65.18.28.1[51]
    Cimetidine4.755.844.695.03[51]
    Ranitidine4.476.674.89<5[51]
    Famotidine<57.56<5[51]
    Tiotidine<47.77<4<5[51]
    Nizatidine6.92§[52]
    Lafutidine7.92[53]
    ICIA 51657.89#[35]
    Roxatidine acetate7.41††[54]
    Zolantidine5.1‡‡7.40[55,28]
    Iodaminopotentidine9.51[29]
    UR-DE257<57.555.42<5[29]
    BMY253687.724.66[29,40]
    UR-NK796.367.946.085.96[14]
    Impromidine5.27.646.997.76[51]
    UR-SB69<47.655.34.4[40]
    UR-KAT479<57.59§§/7.78¶¶4.91<5[17]

    Binding data (pKi) were determined at human HxRs in different assay systems, unless otherwise stated.

    †gpH1R.

    ‡rH3R.

    §pKd from measurements of CRE-SPAP production from CHO-H2-SPAP cells in the presence of histamine.

    ¶Data (pKi) from radioligand competition binding experiments performed in guinea pig cerebral cortex synaptic membrane with [3H]tiotidine.

    #Data (pKi) from radioligand competition binding experiments performed at guinea pig gastric mucosa with [3H]ICIA5165.

    ††H2R antagonistic activity (pA2) on the isolated guinea pig right atrium.

    ‡‡Data (pKi) from competition binding experiments with [3H]mepyramine performed at homogenates of guinea pig cerebral cortex.

    §§Data (pKi) from radioligand competition binding experiments performed on membrane preparations of Sf9 insect cells expressing the hH2R-GsαS with [3H]UR-KAT479.

    ¶¶Data (pKi) from radioligand competition binding experiments performed at HEK293T-hH2R-qs5-HA cells with [3H]UR-KAT479.

    In the 1990s, some experiments with 125I-labeled compounds were performed, resulting in the versatile H2R antagonist [125I]iodoaminopotentidine (Figure 1) [28,36,37]. Thus, [125I]iodoaminopotentidine constituted a potent radioligand at the H2R (Table 1), which could also be used for autoradiography experiments in primates (human/nonhuman brain) and rodents (brain/heart) [36,37]. Its most obvious disadvantage is, however, the use of the iodine-125 isotope (gamma emitter), which involves significantly higher safety precautions during the preparation and usage of the radioligand, compared with tritiated compounds. Moreover, due to the short half-life of the isotope (59.5 days) [38], the radioligand can only be used for 4–6 weeks after preparation [29]. To improve the handling and the safety aspects, the tritiated compound [3H]UR-DE257 (Figure 1) was synthesized, which exhibited a new structural motif with the squaramide modification derived from H2R antagonist BMY25368 (Figure 1 & Table 1) [29]. [3H]UR-DE257 was very well suited to determine pKi values but turned out to be an insurmountable antagonist in functional and kinetic assays, which limited its applicability as a pharmacological tool [29]. The radioligand also possesses high affinity to histamine H2 receptor orthologs (rat, guinea pig) beside the human H2R [29]. Furthermore, [3H]UR-DE257 could be successfully employed for autoradiography of the heart sections of transgenic mice overexpressing the human histamine H2 receptor [39]. The synthesis of [3H]UR-SB69 (Figure 1 & Table 1), a structural combination of the two antagonists famotidine (Figure 1 & Table 1) and UR-DE257, also failed to remedy this situation [40]. In addition to high nonspecific binding, complete displacement of the radioligand could not be achieved in dissociation experiments in this case either [40]. Furthermore, a long-term stability study showed that after 15 months, approximately 50% of the radioligand were already degraded [40]. To circumvent these problems, several attempts were made to radiolabel high affinity H2R agonists with different structural features. A first experiment, starting from the agonistic dimer UR-NK22, resulted in [3H]UR-NK79 (Figure 1 & Table 1) [14]. However, also this ligand did not bind in a saturable manner and showed high non-specific binding in saturation binding experiments [14]. Furthermore, confocal microscopy studies with fluorescent ligands of similar structure indicated that such compounds tend to a pronounced, receptor-independent cellular accumulation [14]. However, since some observations from Kagermeier et al. indicated an improved kinetic behavior, investigations of monomeric carbamoylguanidine-containing amines as promising precursors for H2R radioligands were performed. This led to the development of the highly stable high affinity agonist [3H]UR-KAT479 (Figures 1 & 2, & Table 1), a G protein-biased H2R radioligand with a very good selectivity within the histamine receptor family [17]. In addition to significantly improved kinetics (including complete dissociation from the receptor), [3H]UR-KAT479 showed excellent properties in saturation and competition binding assays [17]. Furthermore, its applicability was demonstrated on H2R orthologs (guinea pig, mouse) by reporting pKi values of standard H2R ligands in whole cells for the first time [17]. Its G protein bias also eliminates the need for specifically adjusted assay conditions (e.g., hypotonic buffers), which can be used to prevent receptor internalization. Finally, the investigated low binding of [3H]UR-KAT479 to red blood cells provides a promising basis for the use of unlabeled carbamoylguanidine-type ligands in animal studies to further explore the role of the H2R in the CNS [17].

    Figure 2. Synthesis of [3H]UR-KAT479 by propionylation of precursor UR-KAT192 with N-succinimidyl [2,3-3H]propionate.

    Radiochemical yield 35%.

    Adapted with permission from [17] © 2020 American Chemical Society.

    In addition to the application of radioligands in basic research, potential H2R drug candidates were also investigated in preclinical studies with regard to their pharmacokinetic properties. In this case, the literature mainly consists of studies with the marketed H2R antagonists. As discussed at the beginning, the selection of the label plays a central role, which is why publications with 14C-labeled compounds are found almost exclusively for metabolic studies. The first work on this topic was published in the early 1980s, studying the excretion and metabolism of [14C]cimetidine in male volunteers [41]. Extensive studies with urinary and fecal samples revealed a comprehensive picture of the metabolic spectrum of cimetidine showing that more than 70% of the 14C was excreted in the urine after 24 h and 5% in the feces [41]. Unchanged cimetidine was the largest urinary component (63%), followed by cimetidine N′-glucuronide (24%) [41]. In addition to the synthesis of 35S- and 14C-labeled famotidine [42], metabolic studies with orally administered [14C]famotidine in man have also been published [43]. Also in this case, urinary and fecal recovery were specifically investigated [43]. Again, renal excretion was the major route of elimination with a recovery of unchanged famotidine of approximately 67% after intravenous administration [43]. The bioavailability averaged 43% of the applied famotidine dose [43]. An interesting ex vivo animal model is provided by the isolated perfused kidney in rats, in which the effect of cationic drugs on the renal secretion of tritiated ranitidine ([3H]ranitidine) was examined [44]. The results indicate that at clinically relevant concentrations the renal tubular secretion of ranitidine is inhibited by trimethoprim, but not by amantadine, pseudoephedrine or triamterene [44]. Besides the classical pharmacokinetic profile in human, pharmacodynamic and hormonal effects of healthy and renally or hepatically impaired patients were also studied with [14C]nizatidine [45]. Nizatidine showed high bioavailability (>90%) with more than 90% being recovered in urine within 16 h of dosing [45]. The drug did not alter hormone concentrations in plasma; however, renal impairment decreased the elimination of nizatidine [45]. Further studies deal with the secretion in human breast milk [46], as well as the localization of the active drug in the parietal cells of gastric mucosa, also here using [14C]nizatidine [47]. The disposition of nizatidine was similar in lactating and nonlactating women with less than 0.1% of the maternal dose being secreted into milk during a 12-h interval after either single or multiple doses [46]. Furthermore, nizatidine was confirmed to be retained as an unchanged drug in the gastric mucosa, supporting the duration of pharmacological effects of nizatidine [47]. Among structurally novel H2R antagonists, metabolic studies with [3H]- and [14C]roxatidine acetate [48], as well as autoradiographic studies of [3H]lafutidine on CGRP-immunoreactive nerves and gastric parietal cells are reported [49]. Roxatidine acetate was almost completely absorbed after oral administration (>95%) and was rapidly converted to roxatidine, its major active plasma and urinary metabolite [48], whereas autoradiographic studies have shown that lafutidine effector sites coincided with the CGRP-immunoreactive nerves as well as the parietal cells [49].

    In addition to the dominant number of pharmacokinetic reports, CNS permeability studies using radiolabeled H2R antagonists have also been performed. Foremost among these is the study by Young et al. that attempts to establish a working model for the passage of histamine H2 receptor antagonists across the blood–brain barrier, where [14C]zolantidine (Table 1), [14C]cimetidine, [14C]ranitidine and [3H]tiotidine, among others, were used [50].

    Conclusion & future perspective

    The aim of this special report was to provide an overview of the use of radioligands at the H2R over the past five decades. Although a lot of progress was made in the field of H2R radioligands, some challenges remain. For example, a recently published study suggests that ligands with similar structural features as [3H]UR-KAT479 possess a high affinity for dopamine receptors of the D2-like family, specifically for the dopamine D3 receptor [18]. This drawback might hamper a possible application in autoradiography experiments.

    Even though H2R agonists have not yet found their way on the drug market, research in this field remains exciting. As described earlier, there are several unanswered questions regarding the human histamine H2 receptor that relate to effects in both the periphery (acute myeloid leukemia) and the CNS (memory and learning), emphasizing the need for the development of appropriate pharmacological tools in this area. Continued efforts in the field of H2R research might eventually lead to the discovery of an agonistic drug for therapeutic purposes, although some studies with regard to bioavailability and blood–brain barrier permeability of the compounds are still pending.

    Executive summary

    Radiolabeling

    • 3H and 14C isotopes are most frequently used for the labeling of potential drug candidates.

    • 3H labeling is mainly used in preliminary in vitro characterization, while 14C labeling is mainly required for metabolic/pharmacokinetic studies (preclinical/clinical phase).

    Radioligands for the histamine H2 receptor

    • 3H-labeled H2R antagonists like [3H]cimetidine, [3H]ranitidine or [3H]tiotidine display comparatively low affinity and high nonspecific binding at the H2R.

    • Squaramide-type H2R antagonist [3H]UR-DE257 with high affinity and lower non-specific binding but insurmountable antagonism.

    • Development of the G protein-biased radioligand [3H]UR-KAT479 (H2R agonist) led to improved kinetic binding properties, beside high affinity and very good receptor subtype selectivity.

    • Recent H2R study shows high affinities of carbamoylguanidine-type agonists such as [3H]UR-KAT479 at dopamine receptors of the D2-like family (mainly D3R).

    • Pharmacokinetic studies of marketed H2R antagonists are mainly performed with 14C-labeled radioligands by pharmaceutical industry in preclinical/clinical stage.

    Financial & competing interests disclosure

    This work was supported by the Deutsche Forschungsgemeinschaft (DFG, Research Training Group GRK 1910). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Ash ASF, Schild HO. Receptors mediating some actions of histamine. Br. J. Pharmacol. Chemo. 27(2), 427–439 (1966).
    • 2. Black JW, Duncan WAM, Durant CJ et al. Definition and antagonism of histamine H2-receptors. Nature. 236(5347), 385–390 (1972).
    • 3. Parsons ME, Ganellin CR. Histamine and its receptors. Br. J. Pharmacol. 147(S1), S127–S135 (2006).
    • 4. Brimblecombe RW, Duncan WA, Durant GJ et al. The pharmacology of cimetidine, a new histamine H2-receptor antagonist. Br. J. Pharmacol. 53(3), 435–436 (1975).
    • 5. Bradshaw J, Brittain RT, Clitherow JW et al. Ranitidine (AH 19065): a new potent, selective histamine H2-receptor antagonist [proceedings]. Br. J. Pharmacol. 66(3), 464P (1979).
    • 6. Takagi T, Takeda M, Maeno H. Effect of a new potent H2-blocker, 3-]]] 2-[(diaminomethylene) amino]-4-thiazolyl] methyl]-thio]-N2-sulfamoylpropionamidine (YM-11170) on gastric secretion induced by histamine and food in conscious dogs. Arch. Int. Pharmacodyn. Ther. 256(1), 49 (1982).
    • 7. Meredith CG, Speeg KV Jr, Schenker S. Nizatidine, a new histamine H2-receptor antagonist, and hepatic oxidative drug metabolism in the rat: a comparison with structurally related compounds. Toxicol. Appl. Pharmacol. 77(2), 315–324 (1985).
    • 8. Tarutani M, Sakuma H, Shiratsuchi K, Mieda M. Histamine H2-receptor antagonistic action of N-(3-[3-(1-piperidinylmethyl)phenoxy]propyl)acetoxyacetamide hydrochloride (TZU-0460). Arzneimittelforschung. 35(4), 703–706 (1985).
    • 9. Shibata M, Yamaura T, Inaba N et al. Gastric antisecretory effect of FRG-8813, a new histamine H2 receptor antagonist, in rats and dogs. Eur. J. Pharmacol. 235(2–3), 245–253 (1993).
    • 10. Tiligada E, Ennis M. Histamine pharmacology: from Sir Henry Dale to the 21st century. Br. J. Pharmacol. 177(3), 469–489 (2020).
    • 11. Kraus A, Ghorai P, Birnkammer T et al. NG-acylated aminothiazolylpropylguanidines as potent and selective histamine H2 receptor agonists. ChemMedChem. 4(2), 232–240 (2009).
    • 12. Ghorai P, Kraus A, Keller M et al. Acylguanidines as bioisosteres of huanidines: NG-acylated Imidazolylpropylguanidines, a new class of histamine H2 receptor agonists. J. Med. Chem. 51(22), 7193–7204 (2008).
    • 13. Birnkammer T, Spickenreither A, Brunskole I et al. The bivalent ligand approach leads to highly potent and selective acylguanidine-type histamine H2 receptor agonists. J. Med. Chem. 55(3), 1147–1160 (2012).
    • 14. Kagermeier N, Werner K, Keller M et al. Dimeric carbamoylguanidine-type histamine H2 receptor ligands: a new class of potent and selective agonists. Bioorg. Med. Chem. 23(14), 3957–3969 (2015).
    • 15. Pockes S, Wifling D, Keller M et al. Highly potent, stable, and selective dimeric hetarylpropylguanidine-type histamine H2 receptor agonists. ACS Omega. 3(3), 2865–2882 (2018).
    • 16. Pockes S, Wifling D, Buschauer A, Elz S. Structure–activity relationship of hetarylpropylguanidines aiming at the development of selective histamine receptor ligands. ChemistryOpen. 8(3), 285–297 (2019).
    • 17. Tropmann K, Höring C, Plank N, Pockes S. Discovery of a G protein-biased radioligand for the histamine H2 receptor with reversible binding properties. J. Med. Chem. 63(21), 13090–13102 (2020). •• Describes the recently developed H2R radioligand with high affinity, selectivity and improved kinetic properties.
    • 18. Biselli S, Bresinsky M, Tropmann K et al. Pharmacological characterization of a new series of carbamoylguanidines reveals potent agonism at the H2R and D3R. Eur. J. Med. Chem. 214, 113190 (2021).
    • 19. Calcutt CR, Ganellin CR, Griffiths R et al. Zolantidine (SK&F 95282) is a potent selective brain-penetrating histamine H2-receptor antagonist. Br. J. Pharmacol. 93(1), 69–78 (1988). • Describes the only reported CNS-penetrable H2R antagonist.
    • 20. Martner A, Thorén FB, Aurelius J et al. Immunotherapy with histamine dihydrochloride for the prevention of relapse in acute myeloid leukemia. Expert Rev. Hematol. 3(4), 381–391 (2010).
    • 21. Khan N, Saad A, Nurulain SM et al. The dual-acting H3 receptor antagonist and AChE inhibitor UW-MD-71 dose-dependently enhances memory retrieval and reverses dizocilpine-induced memory impairment in rats. Behav. Brain Res. 297, 155–164 (2016).
    • 22. Sadek B, Khan N, Darras FH et al. The dual-acting AChE inhibitor and H3 receptor antagonist UW-MD-72 reverses amnesia induced by scopolamine or dizocilpine in passive avoidance paradigm in rats. Physiol. Behav. 165, 383–391 (2016).
    • 23. Krauser JA. A perspective on tritium versus carbon-14: ensuring optimal label selection in pharmaceutical research and development. J. Label Compd. Radiopharm. 56(9–10), 441–446 (2013). •• Excellent review with guidance on deciding between 3H- and 14C-labeling of ligands depending on intended use.
    • 24. Warrander SE, Norris DB, Rising TJ, Wood TP. 3H-cimetidine and the H2-receptor. Life Sci. 33(12), 1119–1126 (1983).
    • 25. Rising TJ, Norris DB, Warrander SE, Wood TP. High affinity 3H-cimetidine binding in guinea-pig tissues. Life Sci. 27(3), 199–206 (1980).
    • 26. Smith IR, Cleverley MT, Ganellin CR, Metters KM. Binding of [3H] cimetidine to rat brain tissue. Agents Actions. 10(5), 422–426 (1980).
    • 27. Bristow DR, Hare JR, Hearn JR, Martin LE. Radioligand binding studies using [3H]-cimetidine and [3H]-ranitidine. Br. J. Pharmacol. 72(3), P547–P548 (1981).
    • 28. Leurs R, Smit MJ, Menge WM, Timmerman H. Pharmacological characterization of the human histamine H2 receptor stably expressed in Chinese hamster ovary cells. Br. J. Pharmacol. 112(3), 847–854 (1994).
    • 29. Baumeister P, Erdmann D, Biselli S et al. [3H]UR-DE257: development of a tritium-labeled squaramide-type selective histamine H2 receptor antagonist. ChemMedChem. 10(1), 83–93 (2015). •• Describes a squaramide-type H2R radioligand with insurmountable binding properties.
    • 30. Grätz L, Tropmann K, Bresinsky M et al. NanoBRET binding assay for histamine H2 receptor ligands using live recombinant HEK293T cells. Sci. Rep. 10(1), 13288 (2020).
    • 31. Gajtkowski GA, Norris DB, Rising TJ, Wood TP. Specific binding of 3H-tiotidine to histamine H2 receptors in guinea pig cerebral cortex. Nature. 304(5921), 65–67 (1983).
    • 32. Kelley MT, Bürckstümmer T, Wenzel-Seifert K et al. Distinct interaction of human and guinea pig histamine H2-receptor with guanidine-type agonists. Mol. Pharmacol. 60(6), 1210–1225 (2001).
    • 33. Sinkins WG, Kandel M, Kandel SI et al. G protein-linked receptors labeled by [3H] histamine in guinea pig cerebral cortex. I. Pharmacological characterization [corrected]. Mol. Pharmacol. 43(4), 569–582 (1993).
    • 34. Schwartz J-C, Arrang J-M, Garbarg M et al. Histaminergic transmission in the mammalian brain. Physiol. Rev. 71(1), 1–51 (1991).
    • 35. Nielsen ST. Binding of [3H] ICIA 5165, an H2-receptor antagonist to guinea pig gastric mucosa. Agents Actions. 18(5), 524–531 (1986).
    • 36. Ruat M, Traiffort E, Bouthenet ML et al. Reversible and irreversible labeling and autoradiographic localization of the cerebral histamine H2 receptor using [125I] iodinated probes. Proc. Natl Acad. Sci. USA 87(5), 1658–1662 (1990).
    • 37. Martinez-Mir MI, Pollard H, Moreau J et al. Three histamine receptors (H1, H2 and H3) visualized in the brain of human and non-human primates. Brain Res. 526(2), 322–327 (1990).
    • 38. Unterweger MP, Hoppes DD, Schima FJ. New and revised half-life measurements results. Nucl. Instrum. Methods Phys. Res. 312(1), 349–352 (1992).
    • 39. Gergs U, Bernhardt G, Buchwalow IB et al. Initial characterization of transgenic mice overexpressing human histamine H2 receptors. J Pharmacol. Exp. Ther. 369(1), 129–141 (2019).
    • 40. Biselli S. Synthesis and Pharmacological Characterization of Subtype-Selective Ligands, Including Radio- and Fluorescence Labeled Ligands, for the Histamine H2 Receptor [PhD thesis]. University of Regensburg, Germany (2019).
    • 41. Mitchell SC, Idle JR, Smith RL. The metabolism of [14C]cimetidine in man. Xenobiotica. 12(5), 283–292 (1982). • Extensive work about the metabolism of cimetidine in man using carbon-14 labeling.
    • 42. Tamazawa K, Arima H. Synthesis of 35S- and 14C-labeled famotidine, a new potent histamine H2 receptor antagonist. J. Label. Compd. Radiopharm. 20(10), 1193–1200 (1983).
    • 43. Yeh KC, Chremos AN, Lin JH et al. Single-dose pharmacokinetics and bioavailability of famotidine in man. Results of multicenter collaborative studies. Biopharm. Drug Dispos. 8(6), 549–560 (1987).
    • 44. Nation RL, Evans AM, Cabot JL et al. Effect of cationic drugs on the renal secretion of ranitidine in the rat isolated perfused kidney. Clin. Exp. Pharmacol. Physiol. 25(1), 33–37 (1998).
    • 45. Callaghan JT, Bergstrom RF, Rubin A et al. A pharmacokinetic profile of nizatidine in man. Scand. J. Gastroenterol. 22(136), 9–17 (1987).
    • 46. Obermeyer BD, Bergstrom RF, Callaghan JT et al. Secretion of nizatidine into human breast milk after single and multiple doses. Clin. Pharmacol. Ther. 47(6), 724–730 (1990).
    • 47. Sano H, Mera Y, Arai K et al. Localization of 14C-nizatidine in the gastric wall of rats. Arzneimittelforschung. 41(9), 958–961 (1991).
    • 48. Collins JD, Pidgen AW. Pharmacokinetics of roxatidine in healthy volunteers. Drugs. 35(3), 41–47 (1988).
    • 49. Nakamura M, Akiba Y, Matsui H et al. Autoradiographic and confocal laser microscopic observation show lafutidine binds to CGRP-immunoreactive nerves as well as gastric parietal cells. Inflammopharmacology. 10(4–6), 495–503 (2002).
    • 50. Young RC, Mitchell RC, Brown TH et al. Development of a new physicochemical model for brain penetration and its application to the design of centrally acting H2 receptor histamine antagonists. J. Med. Chem. 31(3), 656–671 (1988). • Describes an important brain penetration model for centrally acting H2R antagonists.
    • 51. Panula P, Chazot PL, Cowart M et al. International Union of Basic and Clinical Pharmacology. XCVIII. Histamine receptors. Pharmacol Rev. 67(3), 601–655 (2015).
    • 52. Baker JG. A study of antagonist affinities for the human histamine H2 receptor: H2 antagonist affinities. Bri. J. Pharmacol. 153(5), 1011–1021 (2008).
    • 53. Inaba N, Shibata M, Onodera S et al. [Studies on histamine H2-receptor antagonistic property of FRG-8813, a novel anti-ulcer drug]. Nihon Yakurigaku Zasshi. 105(4), 231–241 (1995).
    • 54. Li L, Kracht J, Peng S et al. Synthesis and pharmacological activity of fluorescent histamine H2 receptor antagonists related to potentidine. Bioorg. Med. Chem. Lett. 13(10), 1717–1720 (2003).
    • 55. Calcutt CR, Ganellin CR, Griffiths R et al. Zolantidine (SK&F 95282) is a potent selective brain-penetrating histamine H2-receptor antagonist. Br. J. Pharmacol. 93(1), 69–78 (1988).