The present disclosure relates to the treatment of pharmacoresistant epilepsy with propynones, propynals, propynols, propynes, propenones, amides, quinolinones, naphthyridinones, thiopyranoxides, pyrazolopyridines, and indazoles.
More than 70 million people worldwide are affected by epilepsy, a common severe neurological disease that is characterized by an enduring predisposition of the brain to generate epileptic seizures and the associated neurobiological, cognitive, psychological, and social consequences (1-4). The first-line treatment consists of pharmacotherapy with antiseizure drugs (ASDs) to control the seizures in terms of incidence and severity (4, 5). Unfortunately, despite the availability of more than 25 approved ASDs, approximately 30% of epilepsy patients fail to achieve seizure freedom due to pharmacoresistance, also known as drug-resistance (4, 6-8). Besides, first-line ASDs are associated with important adverse effects that can significantly impact daily life (5, 9). Hence, there is an unmet medical need for safer and more effective ASDs.
The search for new and improved ASDs, effective against drug-resistant seizures, has proven particularly challenging (10). An important limitation is the fact that the more clinically-relevant animal models of drug-resistant seizures, such as the rat lamotrigine-resistant amygdala kindling model, are typically chronic models, which are not suited for drug screening because they are low-throughput and labour-intensive (11). With the exception of the pharmacoresistant acute mouse 6-Hz (44 mA) psychomotor seizure model (12), either drug-sensitive acute rodent models (e.g., the maximal electroshock seizure test (MES)) or in vitro target-based assays are used for drug screening. However, they are prone for the selection of ‘me-too’ drug candidates.
The lack of rodent models that can capture pharmacoresistant epileptic seizures and are suitable for high-throughput drug screening calls for additional drug discovery strategies to identify clinically relevant hits. In this regard the larval zebrafish model has gained interest as a small vertebrate that combines the strengths of high-throughput drug screening with in vivo testing (13, 14). Moreover, the use of a lower vertebrate for screening purposes is ethically preferable to higher vertebrates (15).
Recently, several drug-resistant larval zebrafish epilepsy and seizure models have been reported (13, 16-18). Among them is the zebrafish ethyl ketopentanoate (EKP) seizure model, a novel pharmacoresistant animal model of chemically-induced seizures that was developed by the Laboratory for Molecular Biodiscovery (Prof. P. de Witte), pharmacologically characterized, and shown to have the potential to select innovative antiseizure compounds (16). EKP is a lipid-permeable inhibitor of glutamic acid decarboxylase (GAD) that converts glutamate into γ-aminobutyric acid (GABA) (16). GAD is a key enzyme in the dynamic regulation of neural network excitability (16). Importantly, the decrease of GAD activity in zebrafish is clinically relevant as lowered GAD activity is associated with several forms of epilepsy which are often treatment resistant (19-25).
In our search for novel antiseizure compounds with the potential to treat drug-resistant seizures, we selected the zebrafish ethyl ketopentanoate (EKP) seizure model for hit identification and as critical gate-keeper for further investigation in rodent models because: (1) it is validated for the presence of behavioural and non-behavioural seizure biomarkers, (2) it has been pharmacologically characterized and demonstrated to be pharmacoresistant, (3) it has a novel and clinically relevant underlying mechanism of seizure induction, and (4) it is suitable for high-throughput screening (16, 26).
Using our zebrafish-based drug discovery approach, we found by automated behavioural analysis propynones that are active against EKP-induced drug-resistant seizures. To further investigate their antiseizure activities, improve our understanding of the structural necessities, and select hits with an optimal safety-efficacy profile, a compound library of 56 structurally related small molecules was synthesized in a systematic manner. Many of these structures are novel and/or have been synthesized for the first time. Their tolerability in zebrafish larvae was assessed and besides the initial behavioural antiseizure analysis, electrophysiological antiseizure analysis was performed using non-invasive local field potential (LFP) recordings. In total, 11 structurally related, novel classes of antiseizure compounds were discovered, namely, propynones, propynals, propynols, propynes, propenones, amides, quinolinones, naphthyridinones, thiopyranoxides, pyrazolopyridines, and indazoles, including more than 30 hits. Among the novel antiseizure compounds identified were rac-3-(4-(tert-butyl)phenyl)-1-phenylprop-2-yn-1-ol (compound 3.3) and 3-((3-chlorophenyl)ethynyl)-1H-pyrazolo[3,4-b]pyridine (compound 10.1), which were well tolerated in vivo and did not demonstrate apparent off-targets after in vitro pharmacological profiling. Moreover, their potential against drug-resistant seizures was validated in the mouse 6-Hz (44 mA) seizure model and their ADME and pharmacokinetic profiles were determined. Finally, also compounds (2-aminopyridin-3-yl)(4-(3-chlorophenyl)piperazin-1-yl)methanone (compound 6.1) and 2-(4-(tert-butyl) phenyl)-1,8-naphthyridin-4(1H)-one (compound 8.1) were selected for testing and validated in the mouse 6-Hz (44 mA) seizure model.
The limited success of current antiseizure drug therapies against pharmacoresistant epilepsy calls for new drug discovery strategies to identify clinically relevant hits. The larval zebrafish model is of particular interest as it combines the strengths of high-throughput drug screening with in vivo testing. In this study, we used the larval zebrafish ethyl ketopentanoate (EKP) seizure model, a novel animal model of drug-resistant seizures with the potential to select innovative antiseizure compounds. Here, we present the discovery of several novel classes of antiseizure compounds (i.e., propynones, propynals, propynols, propynes, propenones, amides, quinolinones, naphthyridinones, thiopyranoxides, pyrazolopyridines, and indazoles), the synthesis of a compound library of 56 (mostly novel) structurally related small molecules that resulted from a systematic behavioural antiseizure analysis using automated video tracking, their electrophysiological antiseizure analysis using non-invasive local field potential recordings, and the in-depth investigation of the novel antiseizure compounds rac-3-(4-(tert-butyl)phenyl)-1-phenylprop-2-yn-1-ol (compound 3.3) and 3-((3-chlorophenyl)ethynyl)-1H-pyrazolo[3,4-b]pyridine (compound 10.1) in terms of efficacy and safety. Compounds 3.3 and 10.1 were selected from the identified hits because they were well tolerated and highly effective. They were tested in the mouse 6-Hz (44 mA) psychomotor seizure model, a gold standard that functions as gatekeeper within the Epilepsy Therapy Screening Program (ETSP), and underwent in vitro and in vivo pharmacokinetic analysis and in vitro safety profiling. The potential of compounds 3.3 and 10.1 against drug-resistant seizures, as identified in the zebrafish EKP model, was validated in the mouse 6-Hz (44 mA) seizure model as both showed dose-dependent antiseizure activity. Moreover, among 47 common off-targets tested in vitro, no apparent off-targets were observed. Besides, also compounds (2-aminopyridin-3-yl)(4-(3-chlorophenyl)piperazin-1-yl)methanone (compound 6.1) and 2-(4-(tert-butyl)phenyl)-1,8-naphthyridin-4(1H)-one (compound 8.1) were selected for testing in the mouse 6-Hz (44 mA) seizure model, based on their structural properties, and showed dose-dependent antiseizure activity as well. Taken together, these data show that the novel classes of antiseizure compounds, identified in this study, can be of interest in the search for new and improved drug therapies against pharmacoresistant epilepsy. Continued antiseizure characterization and target identification will be key to unravel their potential. Finally, this study supports the relevance of the use of the larval zebrafish EKP seizure model as a pre-rodent animal model in drug discovery.
Embodiments herein are summarised in the following statements:
The present disclosure relates to compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy.
A first aspect relates to compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy with general structure (I)
Herein X is selected from C═O, CH—OH and CH2.
Furthermore X can be C═N—, whereby the N atom of C═N— is bound to a N atom of a R1. When X is C═N—R1 is has an amino phenyl, or amino pyridyl moiety, typically an 2-amino phenyl, or 2-amino pyridyl moiety. More specifically R1 is 2-amino phenyl, or 2-amino pyridyl.
In compounds with general structure (I), R1 is selected from the group consisting of
In embodiments hereof, R1 is phenyl, optionally substituted with OH, NO2, NH2, OCH3, CH2—NH2, CH2—CH2—NH2, and a halogen.
In embodiments hereof, R1 is phenyl, substituted with a C1-C6 carbon alkyl wherein carbon is optionally replaced by oxygen and/or optionally comprise OH or ═O substituents. Examples of such substituents are methyl and —(C═O)—O—CH2—CH3.
In embodiments hereof, R1 is phenyl, substituted with an aliphatic 6 membered ring, with optional 1 or 2 hetero atoms.
In specific embodiments the phenyl has an N containing substituent bound to the nitrogen of C═N— in the general formula (I).
In embodiments hereof, R1 is an aromatic 5 membered ring, optionally comprising a sulphur heteroatom, or optionally comprising 1 or 2 nitrogen hetero atoms. The substituents are typically at the 2, 4, or 6 position.
In specific embodiments, substituents on the R1 phenyl results in benzocyclohexyl optionally comprising 1 or 2 oxygen heteroatoms (as indicated in compound 1.3.
In C2-C6 carbon alkyls optionally carbon is replaced by oxygen and/or comprise OH or ═O substituents.
Examples of R1 are hydrogen, [1,4]dioxin-6-yl, 1H-imidazol-2-yl, 2-(dimethylamino) pyridin-3-yl), 2-(methylamino)pyridin-3-yl), 2,3-dihydrobenzo[b], 2-amino-5-methylphenyl, 2-aminophenyl, 2-aminopyridin-3-yl, 2-hydroxyphenyl, 2-methoxyphenyl, 2-morpholinopyridin-3-yl, 3-aminopyridin-2-yl, 3-fluorophenyl, 4-(ethoxycarbonyl), 4-aminophenyl, 4-methoxyphenyl, 4-methylpyridin-3-yl, 4-nitrophenyl, isoquinolin-4-yl, phenyl, pyridin-3-yl, and thiophen-3-yl.
In the compounds with general structure (1), R2 is a phenyl, optionally comprising one or two hetero atoms and/or optionally further substituted with OH, OCH3, methyl, halomethyl, one or more halogens, or with a linear or branched C2-C8 alkyl, optionally further substituted with ═O or wherein a carbon atom in the C2-C8 is replace with a halogen.
R2 can be a linear or branched C1-C10 alkyl, C1-C8 alkyl, C1-C6 alkyl, C1-C10 alkene, C1-C8 alkene, C1-C6 alkene, C1-C10 alkyne, C1-C8 alkyne, C1-C6 alkyne, wherein optionally a carbon atom is replaced by a Si atom. R2 can be a C3 to C6 cycloalkyl, such a cyclopropyl.
Examples of R2 are 2-chlorophenyl, 2-methoxyphenyl, 3-(trifluoromethyl), 3,4-dichlorophenyl, 3-chlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 4-(methoxycarbonyl), 4-(tert-butyl)phenyl, 4-(trifluoromethyl), 4-chlorophenyl, 4-fluorophenyl, 4-methoxyphenyl, cyclohexyl, cyclopropyl, isopropyl, m-tolyl, n-hexyl, n-pentyl, o-tolyl, phenyl, and p-tolyl.
In specific embodiments the compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy are propynones, wherein X in general structure (I) is C=0.
Examples hereof are compounds 1.1. to 1.41 depicted in
In specific embodiments compounds for the treatment of epilepsy, in particular drug-resistant epilepsy are 1, 2, 3, 5, 6, 7, 8, 9, 10, 15, 20, 25 compounds selected from the group consisting of 1-(4-methoxyphenyl)-3-(p-tolyl)prop-2-yn-1-one (I.1), 1-(4-nitrophenyl)-3-(p-tolyl)prop-2-yn-1-one (I.2), 1-(4-aminophenyl)-3-(4-(tert-butyl)phenyl)prop-2-yn-1-one (I.4), 1-(2-aminophenyl)-3-(4-(tert-butyl)phenyl)prop-2-yn-1-one (I.5), 3-(4-(tert-butyl)phenyl)-1-(thiophen-3-yl)prop-2-yn-1-one (I.8), 1-(thiophen-3-yl)non-2-yn-1-one (I.9), 3-cyclopropyl-1-(thiophen-3-yl)prop-2-yn-1-one (I.10), 3-(4-(tert-butyl)phenyl)-1-(4-methylpyridin-3-yl)prop-2-yn-1-one (I.11), and 3-(4-(tert-butyl)phenyl)-1-(2-(methylamino)phenyl)prop-2-yn-1-one (I.14) 1-(2-aminopyridin-3-yl)-3-phenylprop-2-yn-1-one (1.1), 1-(2-aminopyridin-3-yl)-3-(4-fluorophenyl)prop-2-yn-1-one (1.2), 1-(2-aminopyridin-3-yl)-3-(triisopropylsilyl)prop-2-yn-1-one (1.3), 1-(2-aminopyridin-3-yl)-3-cyclohexylprop-2-yn-1-one (1.4), 1-(2-aminopyridin-3-yl)oct-2-yn-1-one (1.5), 1-(2-aminopyridin-3-yl)-4-methylpent-2-yn-1-one (1.6), 1-(2-aminopyridin-3-yl)-3-(4-chlorophenyl)prop-2-yn-1-one (1.7), methyl 4-(3-(2-aminopyridin-3-yl)-3-oxoprop-1-yn-1-yl)benzoate (1.8), 1-(2-aminopyridin-3-yl)-3-(4-methoxyphenyl)prop-2-yn-1-one (1.9), 1-(2-aminopyridin-3-yl)-3-(3-chlorophenyl)prop-2-yn-1-one (1.10), 1-(2-aminopyridin-3-yl)-3-(4-(trifluoromethyl)phenyl)prop-2-yn-1-one (1.11), 1-(2-aminopyridin-3-yl)-3-(p-tolyl)prop-2-yn-1-one (1.12), 1-(2-aminopyridin-3-yl)-3-(3,4-dichlorophenyl)prop-2-yn-1-one (1.13), 1-(2-aminophenyl)-3-(4-(tert-butyl)phenyl)prop-2-yn-1-one (1.14), 3-(4-(tert-butyl)phenyl)-1-(pyridin-3-yl)prop-2-yn-1-one (1.15), 3-(4-(tert-butyl)phenyl)-1-phenylprop-2-yn-1-one (1.16), 1-(2-aminopyridin-3-yl)-3-(3-(trifluoromethyl)phenyl)prop-2-yn-1-one (1.17), 3-(4-(tert-butyl)phenyl)-1-(2-methoxyphenyl)prop-2-yn-1-one (1.18), 3-(4-(tert-butyl)phenyl)-1-(2-hydroxyphenyl)prop-2-yn-1-one (1.19), 3-(3-chlorophenyl)-1-(2-chloropyridin-3-yl)prop-2-yn-1-one (1.20), 3-(3-chlorophenyl)-1-(2-morpholinopyridin-3-yl)prop-2-yn-1-one (1.21), 1-(2-aminopyridin-3-yl)-3-(4-(tert-butyl)phenyl)prop-2-yn-1-one (1.22), 1-(2-aminopyridin-3-yl)non-2-yn-1-one (1.23), 1-(3-aminopyridin-2-yl)-3-(4-(tert-butyl)phenyl)prop-2-yn-1-one (1.24) 3-(3-chlorophenyl)-1-(pyridin-3-yl)prop-2-yn-1-one (1.25), 3-(3-chlorophenyl)-1-(1H-imidazol-2-yl)prop-2-yn-1-one (1.26), 3-(3-chlorophenyl)-1-(2-(dimethylamino)pyridin-3-yl)prop-2-yn-1-one (1.27), 1,3-diphenylprop-2-yn-1-one (1.28), 3-(4-methoxyphenyl)-1-phenylprop-2-yn-1-one (1.29), 1-phenyl-3-(p-tolyl)prop-2-yn-1-one (1.30), 3-(4-chlorophenyl)-1-phenylprop-2-yn-1-one (1.31), 3-(3-methoxyphenyl)-1-phenylprop-2-yn-1-one (1.32), 1-phenyl-3-(m-tolyl)prop-2-yn-1-one (1.33), 3-(3-chlorophenyl)-1-phenylprop-2-yn-1-one (1.34) 3-(3-fluorophenyl)-1-phenylprop-2-yn-1-one (1.35), 3-(2-methoxyphenyl)-1-phenylprop-2-yn-1-one (1.36), 1-phenyl-3-(o-tolyl)prop-2-yn-1-one (1.37), 3-(2-chlorophenyl)-1-phenylprop-2-yn-1-one (1.38), 3-(4-(tert-butyl)phenyl)-1-(3-fluorophenyl)prop-2-yn-1-one (1.39), 1-(2-aminophenyl)-3-(3-chlorophenyl)prop-2-yn-1-one (1.40), 1-(2-chlorophenyl)-3-(3-chlorophenyl)prop-2-yn-1-one (1.41).
In specific embodiments the compounds and their use in the treatment of epilepsy, in particular drug resistant epilepsy are propynals, wherein X in general structure (I) is C=0 and R1 is hydrogen.
An example hereof is compound 2.1. as depicted in
In specific embodiments the compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy are propynols, wherein X in general structure (I) is CH—OH.
Examples hereof are compounds 3.1. to 3.4 in
In specific embodiments the compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy are propynes, wherein X is CH2.
An example hereof is compound 4.1 as depicted in
A second aspect relates to propenones for the treatment of epilepsy, in particular drug-resistant epilepsy with general structure (II)
Herein R1 and R2 are as defined for formula I of the first aspect, and R3=R2.
Examples, of R1 are hydrogen, methyl, ethyl, [1,4]dioxin-6-yl, 1H-imidazol-2-yl, 2-(dimethylamino), 2-(methylamino), 2,3-dihydrobenzo[b], 2-amino-5-methylphenyl, 2-aminophenyl, 2-aminopyridin-3-yl, 2-hydroxyphenyl, 2-methoxyphenyl, 2-morpholinopyridin-3-yl, 3-aminopyridin-2-yl, 3-fluorophenyl, 4-(ethoxycarbonyl), 4-aminophenyl, 4-methoxyphenyl, 4-methylpyridin-3-yl, 4-nitrophenyl, isoquinolin-4-yl, phenyl, pyridin-3-yl, and thiophen-3-yl.
Examples of R2 and R3 are independently selected from the group consisting of 2-chlorophenyl, 2-methoxyphenyl, 3-(trifluoromethyl), 3,4-dichlorophenyl, 3-chlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 4-(methoxycarbonyl), 4-(tert-butyl)phenyl, 4-(trifluoromethyl), 4-chlorophenyl, 4-fluorophenyl, 4-methoxyphenyl, cyclohexyl, cyclopropyl, isopropyl, m-tolyl, n-hexyl, n-pentyl, o-tolyl, phenyl, and p-tolyl.
Examples hereof are compounds 5.1 and 5.2 depicted in
A third aspect relates to amides for the treatment of epilepsy, in particular drug-resistant epilepsy with general structure (III)
Herein R1 and R2 are as defined for formula I of the first aspect,
Examples of R1 are hydrogen, methyl, ethyl, [1,4]dioxin-6-yl, 1H-imidazol-2-yl, 2-(dimethylamino) pyridin-3-yl), 2-(methylamino) pyridin-3-yl), 2,3-dihydrobenzo[b], 2-amino-5-methylphenyl, 2-aminophenyl, 2-aminopyridin-3-yl, 2-hydroxyphenyl, 2-methoxyphenyl, 2-morpholinopyridin-3-yl, 3-aminopyridin-2-yl, 3-fluorophenyl, 4-(ethoxycarbonyl), 4-aminophenyl, 4-methoxyphenyl, 4-methylpyridin-3-yl, 4-nitrophenyl, isoquinolin-4-yl, phenyl, pyridin-3-yl, and thiophen-3-yl.
Examples of R2 are 2-chlorophenyl, 2-methoxyphenyl, 3-(trifluoromethyl), 3,4-dichlorophenyl, 3-chlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 4-(methoxycarbonyl), 4-(tert-butyl)phenyl, 4-(trifluoromethyl), 4-chlorophenyl, 4-fluorophenyl, 4-methoxyphenyl, cyclohexyl, cyclopropyl, isopropyl, m-tolyl, n-hexyl, n-pentyl, o-tolyl, phenyl, and p-tolyl.
An example hereof is compound 6.1 depicted in
A fourth aspect relates to compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy with general structure (IV)
Herein Y is nitrogen or carbon,
Herein R2 is as defined for formula (I) of the first aspect,
Examples of R2 are 2-chlorophenyl, 2-methoxyphenyl, 3-(trifluoromethyl), 3,4-dichlorophenyl, 3-chlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 4-(methoxycarbonyl), 4-(tert-butyl)phenyl, 4-(trifluoromethyl), 4-chlorophenyl, 4-fluorophenyl, 4-methoxyphenyl, cyclohexyl, cyclopropyl, isopropyl, m-tolyl, n-hexyl, n-pentyl, o-tolyl, phenyl, and p-tolyl.
Examples hereof are quinoline-4-(1H)-ones such as compound 7.1 depicted in
Examples hereof are 1s,8-Naphthyridin-4-(1H)-ones such as compound 8.1 depicted in
A sixth aspect relates to thiopyran 1-oxides for the treatment of epilepsy, in particular drug-resistant epilepsy, with general structure (V)
Herein R1 and R2 are as defined for formula I of the first aspect,
Examples of R1 are hydrogen, methyl, ethyl, [1,4]dioxin-6-yl, 1H-imidazol-2-yl, 2-(dimethylamino) pyridin-3-yl), 2-(methylamino) pyridin-3-yl), 2,3-dihydrobenzo[b], 2-amino-5-methylphenyl, 2-aminophenyl, 2-aminopyridin-3-yl, 2-hydroxyphenyl, 2-methoxyphenyl, 2-morpholinopyridin-3-yl, 3-aminopyridin-2-yl, 3-fluorophenyl, 4-(ethoxycarbonyl), 4-aminophenyl, 4-methoxyphenyl, 4-methylpyridin-3-yl, 4-nitrophenyl, isoquinolin-4-yl, phenyl, pyridin-3-yl, and thiophen-3-yl.
Examples of R2 are 2-chlorophenyl, 2-methoxyphenyl, 3-(trifluoromethyl), 3,4-dichlorophenyl, 3-chlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 4-(methoxycarbonyl), 4-(tert-butyl)phenyl, 4-(trifluoromethyl), 4-chlorophenyl, 4-fluorophenyl, 4-methoxyphenyl, cyclohexyl, cyclopropyl, isopropyl, m-tolyl, n-hexyl, n-pentyl, o-tolyl, phenyl, and p-tolyl.
An example hereof is compound 9.1. depicted in
A seventh aspect relates to compounds and their use in the treatment of epilepsy, in particular drug-resistant epilepsy with general structure (VI)
Wherein Z is carbon or nitrogen,
R4 are the same R1 as defined for formula (I) of the first aspect,
Examples of R4 are hydrogen, methyl, ethyl, [1,4]dioxin-6-yl, 1H-imidazol-2-yl, 2-(dimethylamino) pyridin-3-yl), 2-(methylamino) pyridin-3-yl), 2,3-dihydrobenzo[b], 2-amino-5-methylphenyl, 2-aminophenyl, 2-aminopyridin-3-yl, 2-hydroxyphenyl, 2-methoxyphenyl, 2-morpholinopyridin-3-yl, 3-aminopyridin-2-yl, 3-fluorophenyl, 4-(ethoxycarbonyl), 4-aminophenyl, 4-methoxyphenyl, 4-methylpyridin-3-yl, 4-nitrophenyl, isoquinolin-4-yl, phenyl, pyridin-3-yl, thiophen-3-yl, methyl, and alkyl.
R2 is as defined for formula (I) of the first aspect,
Example of R2 are 2-chlorophenyl, 2-methoxyphenyl, 3-(trifluoromethyl), 3,4-dichlorophenyl, 3-chlorophenyl, 3-fluorophenyl, 3-methoxyphenyl, 4-(methoxycarbonyl), 4-(tert-butyl)phenyl, 4-(trifluoromethyl), 4-chlorophenyl, 4-fluorophenyl, 4-methoxyphenyl, cyclohexyl, cyclopropyl, isopropyl, m-tolyl, n-hexyl, n-pentyl, o-tolyl, phenyl, and p-tolyl.
Examples hereof are pyrazolo-[3,4-b] pyridines such as compounds 10.1 and 10.2 depicted in
Other examples hereof are indazoles such as compound 11.1 depicted in
The present disclosure further relates to methods of treatment comprising an effective amount of a compound according to any one of the above aspects to an individual suffering from epilepsy, more particular drug resistant epilepsy.
The propynones, propynals, propynols and propynes depicted in
Pyrazolo [3,4-b]pyridines and indazoles are structurally related in that the oxygen, hydroxyl or hydrogen in the propynones, propynals, propynols and propynes and is replaced by a nitrogen forming a bond with a nitrogen R1 substituents, thereby forming a five membered ring with two nitrogens.
“Seizure” refers to a brief episode of signs or symptoms due to abnormal excessive or synchronous neuronal activity in the brain. The outward effect can vary from uncontrolled jerking movement (tonic-clonic seizure) to as subtle as a momentary loss of awareness (absence seizure).
Seizure types are typically classified on observation (clinical and EEG) rather than the underlying pathophysiology or anatomy.
A more recent classification is published in Fisher et al. (2017) Epilepsia 58(4), 522-530.
“Epilepsy” is a condition of the brain marked by a susceptibility to recurrent seizures. There are numerous causes of epilepsy including, but not limited to birth trauma, perinatal infection, anoxia, infectious diseases, ingestion of toxins, tumours of the brain, inherited disorders or degenerative disease, head injury or trauma, metabolic disorders, cerebrovascular accident and alcohol withdrawal.
A large number of subtypes of epilepsy have been characterized and categorized. The classification and categorization system, that is widely accepted in the art, is that adopted by the International League Against Epilepsy's (“ILAE”) Commission on Classification and Terminology [See e.g., Berg et al. (2010), “Revised terminology and concepts for organization of seizures,” Epilepsia, 51(4), 676-685]:
A more recent classification can be found in Scheffer et al. (2017) Epilepsia. 58, 512-521.
“Drug-resistant epilepsy (DRE)” is defined by Kwan et al. (2010) Epilepsia 52(6), 1069-1077, as “failure of adequate trials of two tolerated and appropriately chosen and used antiepileptic drugs (AED schedules) (whether as monotherapies or in combination) to achieve sustained seizure freedom.”
More particularly Drug-resistant epilepsy (DRE) is an epilepsy wherein two of the below list fails to achieve sustained seizure freedom.
A non-exhaustive list of anti-epileptic compounds includes Paraldehyde; Stiripentol; Barbiturates (such as Phenobarbital, Methylphenobarbital, Barbexaclone; Benzodiazepines (such as Clobazam, Clonazepam, Clorazepate, Diazepam Midazolam and Lorazepam); Potassium bromide; Felbamate; Carboxamides (such as Carbamazepine Oxcarbazepine and Eslicarbazepine acetate); fatty-acids (such as valproic acid, sodium valproate, divalproex sodium, Vigabatrin, Progabide and Tiagabine); Topiramate; Hydantoins (such as Ethotoin, Phenytoin, Mephenytoin and Fosphenytoin); Oxazolidinediones (such as Paramethadione Trimethadione and Ethadione); Beclamide; Primidone; Pyrrolidines such as Brivaracetam Etiracetam Levetiracetam; Seletracetam; Succinimides (such as Ethosuximide, Phensuximide and Mesuximide); Sulfonamides (such as Acetazolamide, Sultiame Methazolamide and Zonisamide); Lamotrigine; Pheneturide; Phenacemide; Valpromide; Valnoctamide; Perampanel; Stiripentol; Pyridoxine.
The compound as claimed and their use refers to the chemical formula with general structure as defined, and pharmaceutically accepted derivatives thereof. These may be used as a free acid or base, and/or in the form of a pharmaceutically acceptable acid-addition and/or base-addition salt (e.g. obtained with non-toxic organic or inorganic acid or base), in the form of a hydrate, solvate and/or complex, and/or in the form of a pro-drug or pre-drug, such as an ester. As used herein and unless otherwise stated, the term “solvate” includes any combination which may be formed by a pharmaceutical composition of this disclosure with a suitable inorganic solvent (e.g. hydrates) or organic solvent, such as but not limited to alcohols, ketones, esters, and the like. Such salts, hydrates, solvates, etc. and the preparation thereof will be clear to the skilled person.
“treatment” relates to any medical benefit and in the context of epilepsy to less severe seizures shorter seizure periods or a reduced frequency of seizures.
In the present disclosure a zebrafish model is used as a model for drug-resistant epilepsy. The lipid-permeable glutamic acid decarboxylase (GAD)-inhibitor, ethyl ketopentanoate (EKP), is used that induces drug-resistant seizures in zebrafish. GAD, converting glutamate into GABA, is a key enzyme in the dynamic regulation of neural network excitability. Clinical evidence has shown that lowered GAD activity is associated with several forms of epilepsy that are often treatment resistant. This EKP-induced epilepsy zebrafish model has been validated as a model for drug-resistant epilepsy and was used to demonstrate anticonvulsant activity of various anti-epileptic drugs (AEDs).
The compound library was synthesized by the laboratory of MolDesignS (Prof. W. De Borggraeve) using a variety of synthetic strategies. Each compound was designed based on the potency of the prior candidates, which was determined via automated behavioural antiseizure analysis. All synthetic protocols are described in detail in examples 14-27.
EKP was synthesized in several batches by the laboratory of MolDesignS (Prof. W. De Borggraeve), using an in-house-optimized literature procedure (16) (
For experiments with zebrafish larvae, dry compounds were dissolved in 100% dimethyl sulfoxide (DMSO, spectroscopy grade, Acros Organics (Geel, Belgium)) as 100-fold concentrated stocks and diluted in embryo medium to a final concentration of 1% DMSO content. Control groups were treated with 1% DMSO (VHC) in accordance with the final solvent concentration of tested compounds. For mouse experiments, a mixture of 8% solutol, 12% polyethylene glycol M.W. 200 (PEG200, >95% purity, Acros Organics (Geel, Belgium)) and 80% water was used as solvent and VHC. First, compounds were dissolved in 40% solutol/60% PEG200, after which the solution or suspension was diluted 5-fold in water. Valproate (sodium valproate, ≥98% purity) was purchased from Sigma-Aldrich (Overijse, Belgium).
All animal experiments carried out were in accordance with Directive 2010/63/EU, implemented in 2020 by the Commission Implementing Decision (EU) 2020/569, and approved by the Ethics Committee of the University of Leuven (approval numbers 023/2017 and 027/2017) and by the Belgian Federal Department of Public Health, Food Safety & Environment (approval number LA1210261).
Adult zebrafish (Danio rerio) stocks of AB strain (Zebrafish International Resource Center, Oregon, WA, USA) were maintained at 28° C., on a 14/10 h light/dark cycle under standard aquaculture conditions. Fertilized eggs were collected via natural spawning and raised in embryo medium (1.5 mM HEPES, pH 7.2, 17.4 mM NaCl, 0.21 mM KCl, 0.12 mM MgSO4, 0.18 mM Ca(NO3)2, and 0.6 μM methylene blue) at 28° C., under a 14/10-h light/dark cycle.
Male NMRI mice (weight 16-20 g) were acquired from Charles River Laboratories (France) and housed in polyacrylic cages under a 14/10-h light/dark cycle at 21° C. The animals were fed a pellet diet and water ad libitum and were allowed to acclimatize for one week before experimental procedures were conducted. Prior to the experiment, mice were isolated in polyacrylic cages with a pellet diet and water ad libitum for habituation overnight in the experimental room, to minimize stress.
Prior to behavioural and electrophysiological antiseizure analysis of compounds, their tolerability in zebrafish larvae was assessed at 10 and 2 μM by water immersion using 12 replicates per condition. After 20 (±2) h of exposure, the larvae were visually evaluated for signs of toxicity under a light microscope. Overall morphology, posture, touch response, oedema, signs of necrosis, swim bladder, and heartbeat were checked. A compound at a certain concentration was defined to be tolerated when no signs of toxicity were observed in comparison to VHC-treated larvae. When tolerance was observed at 10 μM, the tolerability of 50 μM was tested as well.
Experiments were performed as described in (16). In brief, a single 5 or 7 dpf larva was placed in each well of a 96-well plate and treated with either VHC (embryo medium with 1% DMSO) or test compound (dissolved in embryo medium, final solvent concentration of 1% DMSO) in a 100 μL volume. Larvae were incubated in the dark for 2 h at 28° C., whereafter 100 μL of either VHC (embryo medium with 1% DMSO) or 600 μM EKP (dissolved in embryo medium, final solvent concentration of 1% DMSO, 300 μM working concentration) was added to each well. Next, within 5 min the 96-well plate was placed in an automated tracking device (ZebraBox Viewpoint, France) and larval behaviour was video recorded for 30 min. The complete procedure was performed in dark conditions using infrared light. Total locomotor activity was recorded by ZebraLab software (Viewpoint, France) and expressed in actinteg units, which is the sum of pixel changes detected during the defined time interval (5 min). Larval behaviour was depicted as mean actinteg units per 5 min during the 30 min recording period and over consecutive time intervals. Data are pooled from independent experiments and expressed as mean±SD.
Non-invasive LFP recordings were measured from the midbrain (optic tectum) of 7 dpf zebrafish larvae pre-incubated with VHC only, EKP only, or compound and EKP. Larvae were incubated for approximately 22 h with VHC (embryo medium with 1% DMSO) or test compound (dissolved in embryo medium, final solvent concentration of 1% DMSO) in a 100 μL volume. After incubation, VHC (embryo medium with 1% DMSO) or 600 μM EKP (dissolved in embryo medium, final solvent concentration of 1% DMSO, 300 μM working concentration) was added to the well for 15 min prior to recording. These steps occurred at 28° C., while further manipulation and electrophysiological recordings occurred at room temperature (±21° C.) and were performed as described before (16, 27). Each recording lasted 600 seconds.
A power spectral density (PSD) analysis of the recordings was performed using MatLab R2019 software (MATrix LABoratory, USA) as described in (26). The PSD estimate of each LFP recording was summed over each 10 Hz frequency band, ranging from 0 to 160 Hz. Next, the PSD estimates were normalized against the VHC control. Data are expressed as mean±SEM PSD per larva and per condition over the 20-90 Hz region. Outliers were identified via the ROUT test (Q=1%).
The antiseizure activity of compounds was investigated in the mouse 6-Hz (44 mA) psychomotor seizure model as described before (27). In brief, male NMRI mice (average body weight 30 g, range 23.5-38 g) were i.p. injected with 200 μL (injection volume was adjusted to the individual weight) of VHC (8% solutol/12% PEG200/80% water) or treatment (valproate or test compound dissolved in VHC) 30 min before seizure induction by corneal electrical stimulation (6 Hz, 0.2 ms rectangular pulse width, 3 s duration, 44 mA) using an ECT Unit 5780 (Ugo Basile, Comerio, Italy). Seizure behaviour was video recorded and seizure durations were determined by blinded video analysis by experienced researchers, familiar with the different seizure characteristics. Data are expressed as mean±SD.
In vitro ADME profiling of compounds was done by Eurofins Panlabs Inc (St Charles, MO, USA) using their ADME-Tox service (Cat Ref P375, Tier 1 ADME Panel). The following was tested: (1) aqueous solubility in PBS (pH 7.4), simulated intestinal fluid and simulated gastric fluid at 200 μM, (2) protein binding (plasma, human) at 10 μM, (3) A-B and B-A permeability (Caco-2, pH 6.5 and 7.4) at 10 μM, and (4) intrinsic clearance (liver microsomes, human) at 100 nM. Of note, the ADME-Tox service includes the determination of log D values, however, these could not be defined as the concentration of test compound in the aqueous buffer was below the limit of quantitation for both molecules. Hence, c Log P values were calculated based on the corresponding SMILES using Actelion's free OSIRIS DataWarrior software version 5.2.1. (28). The atom-based log P calculation method, called OsirisP, uses as an increment system and adds contributions of every atom based on its atom type. The prediction engine distinguishes a total of 368 atom types and was optimized using a training set of more than 5000 molecules with experimentally determined log P values. The free prediction engine has proven to outperform many alternative calculation methods (29).
SGF, SIF and PBS buffers were prepared as follows: 34.2 mM NaCl, 84.7 mM HCl, 3.2 g/L pepsin (pH 2) for SGF, 50 mM KH2PO4, 38 mM NaOH, 10 g/L pancreatin (pH 7.5) for SIF and 137 mM NaCl, 2.7 mM KCl, 8.1 mM Na2HPO4 and 1.5 mM KH2PO4 (pH 7.4) for PBS. Test compounds were prepared at 200 μM concentrations in the corresponding buffer from a 10 mM stock solution (final solvent concentration of 2% DMSO). Buffer samples were mixed thoroughly followed by a 24 h incubation at room temperature. Samples were centrifuged and the supernatant was used for HPLC analysis. A calibration standard of the test compound was prepared at 200 μM in methanol/water (3:2 v/v) from the stock solution on the day of the analysis. Metoprolol, rifampicin, ketoconazole, phenytoin, haloperidol, simvastatin, diethylstilbestrol and tamoxifen were included in each assay. Aqueous solubility (μM) was determined by comparing peak areas.
Human plasma, used as the protein containing matrix, was spiked with the test compound at 10 μM (final solvent concentration of 1% DMSO). The assay was performed in a 96-well format in a dialysis block (Teflon) with the dialysate compartment containing PBS (pH 7.4) and the sample side containing an equal volume of the spiked protein matrix. The plate was incubated at 37° C. for 4 h. After incubation, samples were taken from both compartments, diluted with the phosphate buffer, followed by the addition of acetonitrile and centrifugation. The supernatants were then used for HPLC-MS/MS analysis. Acebutolol, quinidine and warfarin were tested in each assay. The percentage bound to proteins and the recovery were calculated as follows:
Where Areap, Areab and Areac are the peak area of the analyte in the protein matrix, the peak area of the analyte in the assay buffer and the peak area of the analyte in control sample, respectively.
Caco-2 cells were derived from a human colorectal adenocarcinoma. For permeability assays, cells were seeded at 1×105 cells/cm2 in 96 Multiscreen™ plates (Millipore) and used at days 21-25 post-seeding. Cells were used for 15 consecutive passages in culture. HBSS with 10 mM MES at pH 6.5 (apical side) or HBSS with HEPES at pH 7.4 (basolateral side) were used as the transport buffers. Test compounds were added at 10 μM concentration (final solvent concentration of 1% DMSO) to the apical side to determine apical to basolateral (A-B) transport and to the basal side to determine basolateral to apical (B-A) transport. For inhibition studies, 100 μM verapamil was included on both the A and B sides. Aliquots were taken from the donor side (A-B transport) at time zero and the end point, and from the receiver side (B-A transport) at the end point. Propranolol, labetalol, ranitidine, and colchicine (P-glycoprotein substrate) were included in each assay. Samples were analysed by HPLC-MS/MS for quantification. Papp (cm/sec) was calculated from the following equation:
Where VR is the volume of the receiver, C is the concentration of the test compound (either at the donor or receiver side and at mid-point or end point of the incubation), Δt is the incubation time (seconds) and A is the surface area of the cell monolayer (0.11 cm2).
Metabolic stability of the test compounds was evaluated in pooled liver microsomes from human species. Test compounds were pre-incubated with liver microsomes in phosphate buffer (pH 7.4) in a 37° C. shaking water bath for 5 min and an NADPH-generating system was added to initiate the reaction. Samples were collected at time points of 0, 15, 30, 45 and 60 min and extracted with acetonitrile/methanol. After centrifugation, the supernatants were analysed by HPLC-MS/MS. T1/2 was calculated from the slope of the line obtained by plotting the natural logarithmic percentage (Ln %) of the test compound remaining in the reaction mixture vs. incubation time (min). CLint (μL/min/mg protein) was calculated from T1/2 using following equation:
In vitro pharmacological profiling of compounds was done against 78 functional assays for 47 common off-targets (30) by Eurofins DiscoverX Corporation (Fremont, CA, USA) using their SAFETYscan E/IC50 ELECT service (Cat Ref 87-1003DR, Safety47 Panel Dose Response). The following assays were performed: (1) GPCR cAMP modulation, (2) calcium mobilization, (3) nuclear hormone receptor assays, (4) KINOMEscan binding assays, (5) ion channel assays, (6) transporter assays and (7) enzymatic assays. Compounds were tested in a range of 10 concentrations, following a 3-fold serial dilution, starting from 10 μM. The assays were performed utilizing the PathHunter enzyme fragment complementation (EFC) technology, FLIPR®-based cellular screening assays, and KINOMEscan kinase binding assays.
GPCR cAMP Modulation
cAMP Hunter cell lines were expanded from freezer stocks. Prior to testing, cells were seeded in a total volume of 20 μL into white walled 384-well microplates and incubated at 37° C. cAMP modulation was determined using the DiscoverX HitHunter cAMP XS+ assay. For Gs agonist determination, cells were incubated with sample to induce response. For Gi agonist determination, cells were incubated with sample in the presence of EC80 forskolin to induce response. For both conditions, media was aspirated from cells and replaced with 15 μL 2:1 HBSS/10 mM HEPES:cAMP XS+ Ab reagent. Intermediate dilution of sample stocks was performed to generate 4× sample in assay buffer (containing 4×EC80 forskolin in the case of antagonist determination). 5 μL of 4× sample was added to cells and incubated at 37° C. or room temperature (RT) for 30 or 60 min. Final assay vehicle concentration was 1%. For antagonist determination, cells were pre-incubated with sample followed by agonist challenge at the EC80 concentration. Media was aspirated from cells and replaced with 10 μL 1:1 HBSS/HEPES:cAMP XS+ Ab reagent. 5 μL of 4× compound was added to the cells and incubated at 37° C. or RT for 30 min. 5 μL of 4×EC80 agonist was added to the cells and incubated at 37° C. or RT for 30 or 60 min. For Gi coupled GPCRs, EC80 forskolin was included. After appropriate compound incubation, assay signal was generated through incubation with 20 μL cAMP XS+ ED/CL lysis cocktail for 1 h followed by incubation with 20 μL cAMP XS+ EA reagent for 3 h at RT. Microplates were read following signal generation with a PerkinElmer Envision™ instrument for chemiluminescent signal detection. Compound activity was analyzed using CBIS data analysis suite (ChemInnovation, CA). For Gs agonist mode assays, percentage activity was calculated using the following equation:
For Gs antagonist mode assays, percentage inhibition was calculated using the following equation:
For Gi agonist mode assays, percentage activity was calculated using the following equation:
For Gi antagonist or negative allosteric mode assays, percentage inhibition was calculated using the following equation:
Prior to testing, cells (10,000 cells/well) were expanded from freezer stocks and seeded into a total volume of 50 μL into black walled, clear-bottom, Poly-D-lysine coated 384-well microplates and incubated at 37° C. (≤0.2% DMSO concentration). Assays were performed in 1× dye loading buffer consisting of 1× Dye (DiscoverX, Calcium No WashPLUS kit, Catalog No. 90-0091), 1× Additive A and 2.5 mM Probenecid in HBSS/20 mM HEPES. Probenecid was prepared fresh. Prior to testing, media was aspirated from cells and replaced with 25 μL dye loading buffer. Cells were incubated for 45 min at 37° C. and then 20 min at RT. For agonist determination, 25 μL of 2× compound in HBSS/20 mM HEPES was added using a FLIPR Tetra (MDS). For antagonist determination, cells were pre-incubated with sample followed by agonist challenge at the EC80 concentration. 25 μL of 2× sample was added and cells were incubated for 30 min at RT in the dark to equilibrate plate temperature. After incubation, antagonist determination was initiated by addition of 25 μL of 1× compound with 3×EC80 agonist using FLIPR. For both agonist and antagonist formats, activity was measure on a FLIPR Tetra (MDS). Calcium mobilization was monitored for 2 min with a 5 sec baseline read.
FLIPR read—Area under the curve, was calculated for the entire 2 min read. Compound activity was analysed using CBIS data analysis suite (ChemInnovation, CA). For agonist mode assays, percentage activity was calculated using the following equation:
For antagonist mode assays, percentage inhibition was calculated using the following equation:
Prior to testing, PathHunter NHR cell lines were expanded from freezer stocks and cells were seeded in a total volume of 20 μL into white walled 384-well microplates and incubated at 37° C. For agonist determination, cells were incubated with sample to induce response and an intermediate dilution of sample stocks was performed to generate 5× sample in assay buffer. 5 μL of 5× sample was added to the cells and incubated at 37° C. or RT for 3-16 h. Final assay vehicle concentration was 1%. For antagonist determination, cells were pre-incubated with antagonist followed by agonist challenge at the EC80 concentration. Intermediate dilution of sample stocks was performed to generate 5× sample in assay buffer. 5 μL of 5× sample was added to the cells and incubated at 37° C. or RT for 60 min (vehicle concentration was 1%), followed by the addition of 5 μL of 6×EC80 agonist in assay buffer and incubation at 37° C. or RT for 3-16 h. Assay signals were generated through a single addition of 12.5 or 15 μL (50% v/v) of PathHunter Detection reagent cocktail followed by a 1 h incubation at RT. Microplates were read following signal generation with a PerkinElmer Envision™ instrument for chemiluminescent signal detection. Compound activity was analysed using CBIS data analysis suite (ChemInnovation, CA). For agonist mode assays, percentage activity was calculated using the following equation:
For antagonist mode assays, percentage inhibition was calculated using the following equation:
For select assays, the ligand response produced a decrease in receptor activity (inverse agonist with a constitutively active target). For those assays inverse agonist activity was calculated by the following equation:
For most assays, kinase-tagged T7 phage strains were grown in parallel in 24-well blocks in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage from a frozen stock (multiplicity of infection=0.4) and incubated with shaking at 32° C. until lysis (90-150 min). The lysates were centrifuged (6,000×g) and filtered (0.2 μm) to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 min at RT to generate affinity resins for kinase assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce non-specific phage binding. Binding reactions were assembled by combining kinases, liganded affinity beads and test compounds in 1× loading buffer (20% SeaBlock, 0.17×PBS, 0.05% Tween 20, 6 mM DTT). All reactions were performed in polypropylene 384-well plates in a final volume of 0.02 mL. The assay plates were incubated at room temperature with shaking for 1 h and the affinity beads were washed with wash buffer (1×PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (1×PBS, 0.05% Tween 20, 0.5 μM non-biotinylated affinity ligand) and incubated at RT with shaking for 30 min. The kinase concentration in the eluates was measured by qPCR.
The kinase concentration in the eluates was measured by qPCR. qPCR reactions were assembled by adding 2.5 μL of kinase eluate to 7.5 μL of qPCR master mix containing 0.15 μM amplicon primers and 0.15 μM amplicon probe. The qPCR protocol consisted of a 10 min hot start at 95° C., followed by 35 cycles of 95° C. for 15 sec, 60° C. for 1 min.
Percentage of response was calculated by the following equation:
Percentage of control was converted to percentage of response using the following formula:
Binding constants (Kds) were calculated with a standard dose-response curve using the Hill equation:
Where the Hill slope was set to −1 and curves were fitted using a non-linear least square fit with the Levenberg-Marquardt algorithm.
Prior to testing, cell lines were expanded from freezer stocks and cells were seeded in a total volume of 20 μL into black walled, clear-bottom, Poly-D-lysine coated 384-well microplates and incubated at 37° C. As described in (2), assays were performed in 1× dye loading buffer consisting of 1× Dye and 2.5 mM Probenecid when applicable. Probenecid was prepared fresh. For agonist (opener) determination, cells were incubated with sample to induce response and an intermediate dilution of sample stocks was performed to generated 2-5× sample assay in buffer. 10-25 μL of 2-5× sample was added to the cells and incubated at 37° C. or RT for 30 min. Final assay vehicle concentration was 1%. For antagonist (blocker) determination, cells were pre-incubated with sample. Intermediate dilution of sample stocks was performed to generate 2-5× sample in assay buffer. After dye loading, cells were removed from the incubator and 10-25 μL of 2-5× sample was added to the cells in the presence of EC80 agonist when appropriate. Cells were incubated for 30 min at RT in the dark to equilibrate the plate temperature. Vehicle concentration was 1%. Again, compound activity was measured as described earlier, on a FLIPR Tetra (MDS). Compound activity was analysed using CBIS data analysis suite (ChemInnovation, CA). For agonist mode assays, percentage activity was calculated using the following equation:
For antagonist mode, percentage of inhibition was calculated as follows:
Prior to testing, cell lines were expanded from freezer stocks and cells were seeded in a total volume of 25 μL into black walled, clear-bottom, Poly-D-lysine coated 384-well microplates and incubated at 37° C. After cell plating and incubation, media was removed an 25 μL of 1× compound in 1×HBSS/0.1% BSA was added. Compounds were incubated with cells for 30 min at 37° C. After compound incubation, 25 μL of 1× dye loading buffer (1× dye, 1×HBSS/20 mM HEPES) was added to the wells. Cells were incubated for 30-60 min at 37° C. After incubation, microplates were transferred to a PerkinElmer Envision™ instrument for fluorescent signal detection. Compound activity was analysed using CBIS data analysis suite (ChemInnovation, CA). For blocker mode assays, percentage of inhibition was calculated using the following formula:
Enzyme preparations were sourced from various vendors-AChE (R&D Systems), COX1 and COX2 (BPS Bioscience), MAOA (Sigma), PDE3A and PDE4D2 (Signal Chem). AChE: enzyme and test compound were pre-incubated for 15 min at RT before substrate addition. Acetylthiocholine and DTNB were added and incubated at RT for 30 min. Signal was detected by measuring absorbance at 405 nm. COX1 and COX2: enzyme stocks were diluted in assay buffer (40 mM Tris-HCl, 1×PBS, 0.5 mM Phenol, 0.01% Tween 20 and 10 nM Hematin) and allowed to equilibrate with compounds at RT for 30 min (binding incubation). Arachidonic acid (1.7 μM) and Ampliflu Red (2.5 μM) were prepared and dispended into a reaction plate. Plates were read immediately on a fluorimeter with the emission detection at 590 nm and excitation wavelength 544 nm. MAOA: enzyme and test compound were pre-incubated for 15 min at 37° C. before substrate addition. The reaction was initiated by addition of kynuramine and incubated at 37° C. for 30 min. The reaction was terminated by addition of NaOH. The amount of 4-hydroquinoline formed was determined through spectrofluorometric readout with the emission detection at 380 nm and excitation wavelength 310 nm. PDE3A and PDE4D2: enzyme and test compound were pre-incubated for 15 min at RT before substrate addition. cAMP substrate (at a concentration equal to EC80) was added and incubated at RT for 30 min. Enzyme reaction was terminated by addition of 9 mM IBMX. Signal was detected using the HitHunter® cAMP detection kit. Microplates were transferred to a PerkinElmer Envision™ instrument and read out as described for each assay. Compound activity was analysed using CBIS data analysis suite (ChemInnovation, CA). For enzyme activity assays, percentage inhibition was calculated using the following equation:
In our search for novel antiseizure compounds with the potential to treat drug-resistant seizures, the zebrafish EKP seizure model was used for hit identification and as critical gate-keeper for further investigation in the pharmacoresistant mouse 6-Hz (44 mA) psychomotor seizure model. An automated behavioural analysis was done of 7-day-old zebrafish larvae using video tracking (ViewPoint, France). In the course of our drug discovery process, several propynones (i.e., compounds I.1, I.2, I.4, I.5, I.8, I.9, I.10, I.11, I.14, and 1.22), a quinolinone (i.e., compound 7.1), and a naphthyridinone (i.e., compound 8.1) were observed to be active against EKP-induced drug-resistant seizures (
To further investigate the antiseizure activities of propynones in general, and of compound 1.22 in particular, to improve our understanding of the structural necessities, and to select hits with an optimal safety-efficacy profile, a compound library of 56 structurally related small molecules was synthesized (
An overview of the behavioural antiseizure activity data is given in
Given the challenge to differentiate between hits in terms of efficacy, all compounds underwent electrophysiological antiseizure analysis (
Prior to electrophysiological antiseizure analysis of the compounds, their tolerability in zebrafish larvae was assessed (Table 1). Compounds were administered to 6 dpf zebrafish larvae via water immersion at 10 and 2 μM for 20 (±2) h. At 7 dpf the larvae were visually evaluated for signs of toxicity under a light microscope. A compound was defined to be tolerated at the concentration used when no signs of toxicity were observed compared to VHC-treated larvae. When tolerance was observed at 2 and 10 μM, the tolerability of 50 μM was tested as well. All compounds were considered safe at 2 μM, except for compounds 1.13, 1.20, 1.21, 1.31, 1.35, and 1.41. These compounds are active and tolerated in the behavioural assay, which is performed on 5 dpf larvae and after 2 h (instead of 7 dpf, 22 h): 1.13, 1.20, 1.21, 1.31, and 1.35
An overview of the electrophysiological antiseizure activity data, also referred to as anti-epileptiform activity, is given in
Although many compounds show potential against EKP-induced drug-resistant seizures in the zebrafish model, compounds 3.3, 10.1, and 10.2 show the most optimal tolerability-efficacy profile. Compounds 3.3 and 10.1 were selected for further investigation in terms of safety (i.e., in vitro pharmacological profiling for 47 common off-targets) and efficacy (i.e., behavioural antiseizure analysis in the mouse 6-Hz (44 mA) psychomotor seizure model, in vitro ADME profiling, and pharmacokinetic analysis in naïve mice). In addition, given the chemical diversity present among the numerous antiseizure hits, compounds 6.1 and 8.1 were also selected for behavioural antiseizure analysis in the mouse 6-Hz (44 mA) psychomotor seizure model. This to investigate whether the activity of these four new classes of antiseizure compounds in the zebrafish EKP model of drug-resistant seizures translates to a standard mouse seizure model of drug-resistant seizures. Selection criteria included heterocycles versus carbocycles, monocyclic compounds versus bicyclic compounds, a diverse set of functional groups, and—feasibly—distinctive pharmacological profiles.
We aimed to investigate whether the anti-epileptiform activity of compounds 3.3, 6.1, 8.1, and 10.1 observed in the zebrafish EKP model, translates to a standard mouse seizure model of drug-resistant seizures. Among the rodent seizure models available, the mouse 6-Hz (44 mA) model was selected. The 6-Hz (44 mA) mouse model is a gold standard in current antiseizure drug discovery that can detect compounds with novel antiseizure mechanisms and with potential activity against pharmacoresistant seizures (12, 38, 39). The 6-Hz 44 mA model is an acute model of pharmacoresistant focal impaired awareness seizures, previously referred to as complex partial or psychomotor seizures that are induced by a low frequency, long duration corneal electrical stimulation (6 Hz, 0.2 ms rectangular pulse width, 3 s duration, 44 mA). Seizures are typically characterized by a clonic phase and stereotypical automatic behaviours like stun, forelimb clonus, Straub tail and vibrissae twitching. For the experiments with compounds 3.3 and 10.1 (
Mice that were injected with compound 3.3 displayed a dose-response relationship (
Mice that were injected with compound 10.1 displayed a dose-response relationship (
Mice that were injected with compound 6.1 displayed a dose-response relationship (
Mice that were injected with compound 8.1 displayed a dose-response relationship (
We can conclude that the anti-epileptiform activity that was observed in the larval EKP-model translates to a standard mouse model of pharmacoresistant seizures. This demonstrates the effectiveness of the zebrafish-based antiseizure drug discovery approach and the potential of the investigated compounds.
To characterize the absorption, distribution, metabolism and excretion (ADME) properties of compounds 3.3 and 10.1, in vitro ADME profiling was performed by Eurofins Panlabs Inc (St Charles, MO, USA) using their ADME-Tox service (Cat Ref P375, Tier 1 ADME Panel) (Table 2). The Log D values could not be defined as the concentration of test compound in the aqueous buffer was below the limit of quantitation for both molecules. Hence, c Log P values were calculated based on the corresponding SMILES using Actelion's free OSIRIS DataWarrior software version 5.2.1. (28) (Table 2). Compound 3.3 displayed a c Log P value of 4.635, which points out a low hydrophilicity and high lipophilicity. Compound 10.1, on the other hand, showed a lower c Log P value of 2.996, and is thus less lipophilic. The solution properties showed high plasma protein binding for both compound 3.3 and 10.1 of 99.8% and 99.65%, respectively, and an acceptable solubility. Solubility studies were performed in PBS (pH 7.4), simulated intestinal fluid (pH 7.5), and simulated gastric fluid (pH 2.0). Compound 3.3 showed a solubility of 17.7 μM in PBS, 102.5 μM in simulated intestinal fluid, and 24.5 μM in simulated gastric fluid. Compound 10.1 had a lower solubility in PBS of <0.1 μM and in simulated intestinal fluid of 26.7 μM. In simulated gastric fluid compound 10.1 had a higher solubility of 26.7 μM. In vitro absorption studies in Caco-cells showed for compound 3.3 a transport activity from the apical to basolateral direction of 0.4×10−6 cm/s and from the basolateral to apical direction of 0.1×10−6 cm/s. For compound 10.1, a transport activity from the apical to basolateral direction of 3.3×10−6 cm/s and from basolateral to apical direction of 0.7×10−6 cm/s was observed. A percentual recovery of 6 and 13% from the apical to basolateral direction and of 15 and 44% from basolateral to apical direction was observed for compounds 3.3 and 10.1, respectively. Overall, the permeability is rather low for both compounds. Finally, in vitro metabolism studies were done to determine the intrinsic clearance in human liver microsomes and the half-life. A half-life of 19 min was observed for compound 3.3 who showed an intrinsic clearance of 380.6 μL/min/mg, and a half-life of 10 min was observed for compound 10.1 who showed an intrinsic clearance of 748.5 μL/min/mg. Taken together, both compounds show an acceptable ADME profile, which could be improved in a hit-to-lead optimization process.
To identify potential off-target effects of compounds 3.3 and 10.1 in vitro pharmacological profiling was performed by Eurofins Panlabs using their Safety47 Screen. The in vitro pharmacological profiles are shown in table 3, showing the IC50 and EC50 values, as well as the maximal response. For compound 3.3, the following IC50 values were obtained: 4.3 μM for the muscarinic acetylcholine receptor M2 (CHRM2), 7.9 μM for the dopamine receptor D2 (DRD2S), 6.4 μM for the 5-hydroxytryptamine 1B (HTR1B) receptor, and 6.9 μM for the alpha-4 beta-2 nicotinic acetylcholine receptors. For compound 10.1, on the other hand, no IC50 or EC50 values were observed at the highest test concentration of 10 μM. Thus, while compound 3.3 shows 4 potential off-targets compound 10.1 has none. Of note, the potencies of compound 3.3 are low with IC50 values of 4-8 μM, which is typically considered safe. Nonetheless, these findings should be further investigated.
All reagents were obtained from commercially available sources (Sigma-Aldrich, Acros Organics, J&K Scientific, AK Scientific, Fisher Scientific, Manchester Organics, Fluorochem, Janssen Chimica and Iris Biotech) and were used without any further purification, unless stated otherwise. 3-bromopyridin-2-amine purchased from Acros was purified via column chromatography on silica gel prior to use. Dry triethylamine used in the carbonylative Sonogashira reactions was previously distilled over sodium or CaH2 and stored under argon atmosphere. Dry DCM and dry dioxane were purchased via Acros Organics in 500 mL glass bottles equipped with an AcroSeal® and were stabilized with amylene (approximately 50 ppm) and BHT (2-5 ppm), respectively, and stored over molecular sieves. Dry THF (unstabilized) and dry toluene were bought via Sigma-Aldrich in 18 L steel drums and were dispensed using a MBRAUN MB-SPS-800 Solvent Purification System.
Unless stated otherwise, all reactions were performed under N2 or Ar atmosphere and stirred magnetically with PTFE-coated magnetic stirring bars at 350-450 rpm. With the exception of the EKP synthesis (see p. S54), solvents were evaporated under reduced pressure using a rotary evaporator at a bath temperature of 50° C. Final compounds were dried under high vacuum (10−3 mbar) at room temperature. Yields refer to isolated compounds after purification with a purity ≥99.8 mol % based on 1H NMR analysis.
1H and 13C nuclear magnetic resonance (NMR) spectra were recorded on a Bruker Avance 300 spectrometer with a Bruker 300 UltraShield™ magnet system (operating at a 1H basic frequency of 300.13 MHz), a Bruker Avance III HD 400 spectrometer with a Bruker Ascend™ 400 magnet system (operating at a 1H basic frequency of 400.17 MHz) or a Bruker Avance II+ 600 spectrometer with a Bruker 600 UltraShield™ magnet system (operating at a 1H basic frequency of 600.13 MHz) in chloroform-d (CDCl3) or DMSO-d6. The data were recorded at room temperature using Bruker TopSpin 3.6.1 and processed and analyzed using Bruker TopSpin 4.1.1. The δ-values are expressed in parts per million (ppm). 1H data were calibrated using tetramethylsilane (TMS) as an internal reference, while 13C data were calibrated using the deuterated solvents as internal reference (for CDCl3 a 1:1:1 triplet at 77.16 ppm and for DMSO-d6 a 1:3:6:7:6:3:1 septet at 39.52 ppm). The following acronyms were used: s (singlet), d (doublet), t (triplet), q (quartet), quint (quintet), sext (sextet), sept (septet), ABq (AB quartet), m (multiplet), br (broadened). The prefix app. denotes the apparent multiplicity of a signal.
Thin layer chromatography (TLC) analysis was performed using Sigma-Aldrich 20×20 cm precoated, glass TLC plates with fluorescent indicator at 254 nm (article number 99571: layer thickness 250 μm, particle size 8.0-12.0 μm, average pore diameter 60 Å). Visualization of the products was achieved by UV-radiation at 254 nm or using a staining solution of saturated o-dianisidine in glacial acetic acid.
Flash column chromatography (medium pressure liquid chromatography, MPLC) was performed using a Büchi Sepacore® flash system, consisting of a Büchi C-660 Fraction Collector, a Büchi C-615 Pump Manager controlling two Büchi C-605 Pump Modules, a Knauer WellChrom K-2501 spectrophotometer (operating at 254 nm), and a Linseis D120S plotter. Büchi PP cartridges (40/150 mm) were filled with 90 g of Acros ultra-pure silica gel for column chromatography (article number 360050300, particle size 40-60 μm, average pore diameter 60 Å) using a Büchi C-670 Cartridger. Unless stated otherwise, the eluent flow rate was set to 25 mL/min.
Column chromatography was performed using Acros silica gel for chromatography article number 240370300, particle size 0.060-0.200 mm, average pore diameter 60 Å).
Microwave-assisted reactions were performed using a single-mode CEM Discover® LabMate operating at 2.465 GHz. The reaction mixture was magnetically stirred and continuously irradiated at a power of 0 to 300 W using the standard absorbance level of 100 W. The reactions were carried out in 10 mL glass microwave vials, sealed with a snap-on cap with septum. When the reaction was finished, the vial was cooled down to ambient temperature under a stream of compressed air. The reaction parameters (temperature, pressure, output power and reaction time) were monitored by a computer using Synergy 1.39 software.
High-resolution mass spectra (HR-MS) were acquired on a quadrupole orthogonal acceleration time-of-flight mass spectrometer (Synapt G2 HDMS, Waters, Milford, MA). Samples were infused at 3 μL/min and spectra were obtained in positive or negative ionization mode with a resolution of 15000 (FWHM) using leucine enkephalin as lock mass.
HR-MS data were acquired for all new compounds and a small number of known compounds for which no HR-MS data were available in literature.
Low resolution mass spectra (LR-MS) were recorded using an Agilent 1100 HPLC system, consisting of a G1311A quaternary pump and solvent module, a G1313A automatic liquid sampler (ALS), a G1315A diode-array detector (DAD, operating at 215, 254, 280, 320 and 365 nm) and a G1316A thermostated column compartment (TCC, kept at a constant temperature of 25° C.) without an HPLC column (direct injection method). The HPLC system was coupled to an Agilent 6110 single-quadrupole mass spectrometer with an electrospray ionization (ESI) source (capillary voltage 3500 V), operating in the positive mode. Samples were prepared by dissolving the compound in methanol to an approximate concentration of 1 mM. Each sample was automatically injected onto the HPLC system (injection volume 10 μL) and run isocratically in 100% methanol (LC-MS grade, Fisher Scientific) with a flow rate of 0.2 mL/min. Data were acquired using Agilent LC/MSD ChemStation software rev. B.04.03-SP2 [105] and processed and analyzed using ACD/Spectrus Processor 2019.1.2.
LR-MS data were acquired for all compounds for which HR-MS data were already available in literature.
Attenuated total reflection (ATR) Fourier-transformed infrared (FT-IR) spectra were recorded on a Bruker Alpha-P FT-IR spectrometer with single reflection Platinum ATR accessory. Samples were analyzed neat in solid or liquid state without any further manipulations. The data were recorded at room temperature using Bruker OPUS 7.5 and processed and analyzed using ACD/Spectrus Processor 2019.1.2. The ν-values are reported in units of reciprocal centimeters (cm−1).
Melting points (MP) were recorded using an Electrothermal™ IA9300 digital melting point apparatus. Samples were analyzed in 1.5 mm outer diameter capillaries with a sample height of 1 mm. The Tm-values values are uncorrected and reported in units of degrees Celsius (° C.).
Protocol based on Zhang et al. Sci. Rep. 2017, 7 (1also).
Ethyl 2-oxopent-4-enoate (ethyl ketopentanoate, EKP) was prepared by adding dropwise boron trifluoride diethyletherate (6.34 mL, 50.00 mmol, 1.00 equiv.) to a stirring solution of ethyl glyoxylate (˜50% in toluene, 9.91 mL, 50.00 mmol, 1.00 equiv.) and allyltrimethylsilane (15.89 mL, 100.00 mmol, 2.00 equiv.) in dry DCM (120 mL) at 0° C. The solution was allowed to warm up to ambient temperature and was stirred for an additional 8 h. After this time, the reaction was quenched with a saturated aqueous NH4Cl and extracted with DCM (3×50 mL). The combined organic extracts were washed with brine (100 mL), dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to yield the crude ethyl 2-hydroxypent-4-enoate (ethyl hydroxypentanoate, EHP) intermediate as a yellow oil.
To a solution of this crude ethyl 2-hydroxypent-4-enoate in DCM (250 mL) was added Dess-Martin periodinane (24.56 g, 55.00 mmol, 1.10 equiv.) while stirring at room temperature. After 18 h, the mixture was quenched with a 1/1 mixture of 10% aqueous Na2S2O3/saturated aqueous NaHCO3 and extracted with DCM (3×50 mL). The combined organic layers were washed with water and brine, dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. To the residue was added Et2O (100 mL) to precipitate out the Dess-Martin iodinane by-products. The suspension was filtered on a glass filter and the EKP-containing filtrate was concentrated under reduced pressure. The residue was purified via column chromatography on silica gel (95/5 pentane/Et2O), furnishing the desired product as a light yellow oil in 26-38% yield.
1H NMR (400 MHz, CDCl3): δ 6.77 (ddd, J=17.2, 11.2, 10.3 Hz, 1H), 6.22 (ddd, J=11.2, 0.9, 0.8 Hz, 1H), 5.92 (br, s, 1H), 5.42 (ddd, J=17.2, 1.8, 0.9 Hz, 1H), 5.26 (ddd, J=10.3, 1.8, 0.8 Hz, 1H), 4.31 (q, J=7.1 Hz, 2H), 1.35 (t, J=7.1 Hz, 3H). 13C NMR (101 MHz, CDCl3): δ 165.65, 139.38, 129.78, 120.16, 112.15, 62.35, 14.02. IR (neat): ν 3424.92 (O—H stretch), 3090.25 (C—H stretch), 2982.60 (C—H stretch), 2941.26 (C—H stretch), 2910.25 (C—H stretch), 1688.69 (C═O stretch). LR-MS (ESI): m/z calculated for [M+Na]+ 165.0528, found 165.1.
All data are in accordance with Zhang cited above. Note that EKP exists solely as its tautomeric enol form when dissolved in CDCl3.
EKP degrades at temperatures higher than 40° C. and in the presence of (Lewis) acids and nucleophiles. Therefore, rotary evaporation was always performed at 35° C. Furthermore, a significant portion of the yield is lost during column chromatography on silica gel. Hence, the elution time should be kept as short as possible. Other methods of purification (e.g. vacuum distillation and kugelrohr) proved even less successful.
Note that EKP is a highly toxic substance with a high vapor pressure. Care should be taken during the entire synthetic procedure or when handling the product. EKP should be stored at a temperature of −25° C. or lower, which freezes the compound. In this fashion, the thermal degradation rate is decreased and the vapor pressure is reduced.
Protocol based on Neumann et al. Org. Lett. 2014, 16 (8), 2216-2219 and Veryser et al. React. Chem. Eng. 2016, 1 (2), 142-146.
To the right chamber of a flame-dried two-chamber reactor (COWare)4-5 was added the aryl halide (0.50 mmol, 1.00 equiv.), palladium(II) chloride (4.4 mg, 0.025 mmol, 5.0 mol %) and Xantphos (14.5 mg, 0.025 mmol, 5.0 mol %) [Hermange et al. J. Am. Chem. Soc. 2011, 133 (15), 6061-6071; Friis et al. J. Am. Chem. Soc. 2011, 133 (45), 18114-18117]. The reactor was closed with two screw caps and septa and was evacuated and backfilled with argon three times. Dry, degassed toluene (3 mL) was added to the left chamber, followed by formic acid (29 μL, 0.75 mmol, 1.50 equiv.) and mesylchloride (58 μL, 0.75 mmol, 1.50 equiv.). To the right chamber, dry, degassed dioxane (3 mL) was added, followed by the terminal alkyne (0.75 mmol, 1.50 equiv.) and dry triethylamine (0.21 mL, 1.50 mmol, 3.00 equiv.) [If solid, the alkyne was added to the two-chamber reactor before it was closed with the screw cap]. The reaction was initiated by the addition of triethylamine (0.21 mL, 1.50 mmol, 3.00 equiv.) to the left chamber of the reactor. Immediately after the addition of the triethylamine, the reactor was placed in an oil bath at 80 or 100° C. for 18 h. When the reaction was finished, the crude reaction mixture was filtered over a pad of Celite® 535. The filtrate was concentrated in vacuo and purified via column chromatography on silica gel.
Protocol based on WO2012065963 and Yamaji et al. Phys. Chem. Chem. Phys. 2017, 19 (26), 17028-17035.
At 0° C., EtMgBr (3.0 M in Et2O, 1.00 ml, 3.00 mmol, 3.00 equiv.) was added to a solution of the terminal alkyne (3.00 mmol, 3.0 equiv.) in dry THF (8 mL) and stirred for 5 min at 0° C. and then for 30 min at room temperature. This solution was slowly added to a solution of the aldehyde (1.00 mmol, 1.00 equiv.) in dry THF (10 mL) under a nitrogen atmosphere at room temperature. After 2 h, the mixture was quenched with saturated aqueous NH4Cl, extracted with DCM and the organic layers were washed with brine and dried over Na2SO4. The resulting organic solution was filtered and the filtrate was concentrated under reduced pressure.
The crude residue was dissolved in DCM (10 mL) and stirred overnight at room temperature in the presence of manganese dioxide (609 mg, 7.00 mmol, 7.00 equiv.). The solution was filtered over Celite® 535 and the solvent was evaporated in vacuo. The residue was purified via column chromatography on silica gel.
General procedure A for the synthesis of propynones was followed using 1-bromo-4-methoxybenzene (63 μL) and 1-ethynyl-4-methylbenzene (95 μL) at 80° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5), furnishing 72% of the desired product as a yellow solid.
1H NMR (300 MHz, CDCl3): δ 8.23-8.14 (m, 2H), 7.57 (app. d, J=8.0 Hz, 2H), 7.21 (app. d, J=8.0 Hz, 2H), 7.01-6.94 (m, 2H), 3.89 (s, 3H), 2.40 (s, 3H). 13C NMR (75 MHz, CDCl3): δ 176.77, 164.41, 141.32, 133.01, 131.96, 130.40, 129.47, 117.23, 113.86, 93.01, 86.78, 55.60, 21.77. IR (neat): ν 2995.00 (C—H stretch), 2968.13 (C—H stretch), 2837.91 (C—H stretch), 2188.89 (C≡C stretch), 1616.35 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 251.1072, found 251.1071. MP: Tm 87.5-88.3.
All data are in accordance with CN107602361.
General procedure A for the synthesis of propynones was followed using 1-bromo-4-nitrobenzene (101 mg) and 1-ethynyl-4-methylbenzene (95 μL) at 80° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5), yielding 12% of the desired product as a yellow solid.
1H NMR (300 MHz, CDCl3): δ 8.37 (app. s, 4H), 7.61 (app. d, J=8.1 Hz, 2H), 7.27 (app. d, J=8.1 Hz, 2H), 2.43 (s, 3H). 13C NMR (75 MHz, CDCl3): δ 175.92, 150.81, 142.44, 141.14, 133.39, 130.42, 129.69, 123.85, 116.27, 96.26, 86.56, 21.88. IR (neat): ν 3102.48 (C—H stretch), 2922.65 (C—H stretch), 2850.31 (C—H stretch), 2195.09 (C≡C stretch), 1641.15 (C═O stretch), 1510.93 (NO2 stretch). HR-MS (ESI): m/z calculated for [M+H]+ 266.0817, found 266.0812. MP: Tm 159.6-160.7.
All data are in accordance with Sarkae et al. Appl. Organomet. Chem. 2020, 34 (7), e5646.
General procedure A for the synthesis of propynones was followed using 6-bromo-2,3-dihydrobenzo[b][1,4]dioxine (67 μL) and ethynylbenzene (82 μL) at 80° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5), furnishing the desired compound as a beige solid in 43% yield.
1H NMR (300 MHz, CDCl3): δ 7.80-7.73 (m, 2H), 7.69-7.64 (m, 2H), 7.52-7.36 (m, 3H), 6.99-6.93 (m, 1H), 4.37-4.26 (m, 4H). 13C NMR (75 MHz, CDCl3): δ 176.54, 149.15, 143.40, 133.01, 131.01, 130.66, 128.67, 123.95, 120.27, 118.87, 117.40, 92.38, 86.87, 64.78, 64.08. IR (neat): ν 3063.21 (C—H stretch), 2935.06 (C—H stretch), 2877.18 (C—H stretch), 2203.36 (C≡C stretch), 1630.82 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 265.0865, found 265.0859. MP: Tm 98.1-100.8 (literature 98.5-99.7).
All data are in accordance with Levashov et al. Russ. J. Gen. Chem. 2017, 87(7), 1627-1630 and Wu et al. Chemistry—A European Journal 2010, 16 (40), 12104-12107.
General procedure A for the synthesis of propynones was followed using 4-bromoaniline (86 mg) and 1-(tert-butyl)-4-ethynylbenzene (135 μL) at 80° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2), furnishing 64% of the desired product as a dark brown solid.
1H NMR (400 MHz, CDCl3): δ 8.05 (app. d, J=8.5 Hz, 2H), 7.58 (app. d, J=8.3 Hz, 2H), 7.41 (app. d, J=8.3 Hz, 2H), 6.67 (app. d, J=8.5 Hz, 2H), 4.37 (br, s, 2H), 1.32 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 176.51, 154.12, 152.49, 132.82, 132.40, 132.05, 125.74, 117.60, 113.79, 92.27, 87.00, 35.11, 31.15. IR (neat): ν 3350.51 (N—H stretch), 3222.36 (N—H stretch), 2957.79 (C—H stretch), 2864.78 (C—H stretch), 2190.96 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 278.1545, found 278.1539. MP: Tm 137.9-139.7.
General procedure A for the synthesis of propynones was followed using 2-bromoaniline (86 mg) and 1-(tert-butyl)-4-ethynylbenzene (135 μL.) at 80° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 98/2), yielding 54% of the desired product as a yellow oil.
1H NMR (400 MHz, CDCl3): δ 8.20 (app. d, J=8.1 Hz, 1H), 7.61 (app. d, J=8.2 Hz, 2H), 7.43 (app. d, J=8.2 Hz, 2H), 7.31 (app. t, J=8.1 Hz, 1H), 6.73 (app. t, J=8.1 Hz, 1H), 6.67 (app. d, J=8.1 Hz, 1H), 6.39 (br, s, 2H), 1.34 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 179.75, 154.17, 151.16, 135.31, 134.59, 132.79, 125.77, 119.08, 117.58, 116.87, 116.19, 92.99, 87.02, 35.15, 31.18. IR (neat): ν 3445.59 (N—H stretch), 3338.11 (N—H stretch), 3036.34 (C—H stretch), 2961.93 (C—H stretch), 2904.05 (C—H stretch), 2866.85 (C—H stretch), 2199.23 (C≡C stretch), 1620.48 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 278.1545, found 278.1540.
General procedure A for the synthesis of propynones was followed using 2-bromoaniline (86 mg) and oct-1-yne (149 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5), furnishing the desired product as a brown oil in 48% yield.
1H NMR (400 MHz, CDCl3): δ 8.07 (app. dd, J=8.3, 1.0 Hz, 1H), 7.27 (app. t, J=7.2 Hz, 1H), 6.67 (app. t, J=7.2 Hz, 1H), 6.62 (app. d, J=8.3 Hz, 1H), 6.32 (br, s, 2H), 2.47 (t, J=7.1 Hz, 2H), 1.65 (app. quint, J=7.1 Hz, 2H), 1.46 (app. quint, J=7.1 Hz, 2H), 1.37-1.27 (m, 4H), 0.90 (t, J=6.8 Hz, 3H). 13C NMR (101 MHz, CDCl3): δ 179.94, 150.96, 135.01, 134.65, 118.86, 116.67, 115.94, 95.85, 79.87, 31.25, 28.65, 27.86, 22.51, 19.20, 14.03. IR (neat): ν 3437.32 (N—H stretch), 3243.03 (N—H stretch), 2926.79 (C—H stretch), 2856.51 (C—H stretch), 2219.89 (C≡C stretch), 1618.41 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 230.1545, found 230.1544.
General procedure A for the synthesis of propynones was followed using ethyl 4-bromobenzoate (82 μL) and oct-1-yne (149 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 99/1), yielding 21% of the desired product as a brown oil.
1H NMR (400 MHz, CDCl3): δ 8.16 (app. d, J=8.5 Hz, 2H), 8.13 (app. d, J=8.5 Hz, 2H), 4.40 (q, J=7.1 Hz, 2H), 2.51 (t, J=7.1 Hz, 2H), 1.69 (app. quint, J=7.2 Hz, 2H), 1.53-1.46 (m, 2H), 1.41 (t, J=7.1 Hz, 3H), 1.38-1.29 (m, 4H), 0.90 (t, J=6.7 Hz, 3H). 13C NMR (101 MHz, CDCl3): δ 177.47, 165.73, 139.94, 134.87, 129.65, 129.36, 98.10, 79.69, 61.52, 31.23, 28.66, 27.74, 22.50, 19.29, 14.28, 14.02. IR (neat): ν 2955.73 (C—H stretch), 2928.86 (C—H stretch), 2858.58 (C—H stretch), 2199.23 (C≡C stretch), 1719.69 (C═O stretch ester), 1647.35 (C═O stretch ketone). HR-MS (ESI): m/z calculated for [M+H]+ 287.1647, found 287.1638.
General procedure A for the synthesis of propynones was followed using 3-bromothiophene (47 μL) and 1-(tert-butyl)-4-ethynylbenzene (180 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 98/2 to 9/1), furnishing 39% of the desired product as a brown oil.
1H NMR (400 MHz, CDCl3): δ 8.36 (app. dd, J=2.9, 0.6 Hz, 1H), 7.68 (app. dd, J=5.1, 0.6 Hz, 1H), 7.61 (app. d, J=8.2 Hz, 2H), 7.45 (d, J=8.2 Hz, 2H), 7.34 (app. dd, J=5.1, 3.0 Hz, 1H), 1.35 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 171.53, 154.57, 143.14, 135.26, 132.95, 126.87, 126.77, 125.79, 116.99, 92.04, 87.20, 35.11, 31.08. IR (neat): ν 3106.61 (C—H stretch), 2961.93 (C—H stretch), 2866.85 (C—H stretch), 2186.82 (C≡C stretch), 1626.68 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 269.1000, found 269.0997.
General procedure A for the synthesis of propynones was followed using 3-bromothiophene (47 μL) and oct-1-yne (149 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 98/2 to 95/5), furnishing the desired product as a brown oil in 53% yield.
1H NMR (300 MHz, CDCl3): δ 8.22 (app. dd, J=3.0, 1.2 Hz, 1H), 7.59 (app. dd, J=5.1, 1.2 Hz, 1H), 7.30 (app. dd, J=5.1, 3.0 Hz, 1H), 2.45 (t, J=7.1 Hz, 2H), 1.66 (app. quint, J=7.1 Hz, 2H), 1.52-1.40 (m, 2H), 1.39-1.26 (m, 4H), 0.98 (t, J=6.8 Hz, 3H). 13C NMR (75 MHz, CDCl3): δ 171.71, 143.13, 135.19, 126.77, 126.59, 94.98, 80.18, 31.22, 28.63, 27.78, 22.48, 19.10, 14.02. IR (neat): ν 3106.61 (C—H stretch), 2953.66 (C—H stretch), 2926.79 (C—H stretch), 2856.51 (C—H stretch), 2199.23 (C≡C stretch), 1632.88 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 221.1000, found 221.0995.
General procedure A for the synthesis of propynones was followed using 3-bromothiophene (47 μL) and ethynylcyclopropane (85 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 98/2 to 8/2), furnishing 60% of the desired product as a brown solid.
1H NMR (300 MHz, CDCl3): δ 8.19 (app. dd, J=3.0, 1.2 Hz, 1H), 7.56 (app. dd, J=5.1, 1.2 Hz, 1H), 7.29 (app. dd, J=5.1, 3.0 Hz, 1H), 1.57-1.45 (m, 1H), 1.07-0.95 (m, 4H). 13C NMR (75 MHz, CDCl3): δ 171.55, 143.14, 135.04, 126.84, 126.63, 99.20, 76.00, 9.91, −0.01. IR (neat): ν 3085.94 (C—H stretch), 3005.33 (C—H stretch), 2920.59 (C—H stretch), 2850.31 (C—H stretch), 2207.49 (C≡C stretch), 1661.82 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 177.0374, found 177.0368. MP: Tm 78.8-80.6.
General procedure A for the synthesis of propynones was followed using 3-bromo-4-methylpyridine (56 μL) and 1-(tert-butyl)-4-ethynylbenzene (180 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM to DCM/MeOH 95/5), furnishing the desired product as a dark brown oil in 15% yield.
1H NMR (300 MHz, CDCl3) δ 9.46 (app. s, 1H), 8.60 (app. d, J=5.1 Hz, 1H), 7.62 (app. d, J=8.6 Hz, 2H), 7.44 (app. d, J=8.6 Hz, 2H), 7.21 (app. d, J=5.1 Hz, 1H), 2.68 (s, 3H), 1.33 (s, 9H). 13C NMR (75 MHz, CDCl3): δ 178.19, 154.90, 153.85, 152.58, 149.44, 133.10, 131.85, 126.82, 125.83, 116.65, 94.00, 87.60, 35.14, 31.04, 21.36. IR (neat): ν 3046.67 (C—H stretch), 2961.93 (C—H stretch), 2906.12 (C—H stretch), 2868.91 (C—H stretch), 2195.09 (C≡C stretch), 1641.15 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 278.1545, found 278.1541.
General procedure A for the synthesis of propynones was followed using 4-bromoisoquinoline (104 mg) and 1-(tert-butyl)-4-ethynylbenzene (180 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM to DCM/MeOH 95/5), furnishing 41% of the desired product as a dark yellow solid.
1H NMR (400 MHz, CDCl3): δ 9.61 (app. s, 1H), 9.41 (app. s, 1H), 9.20 (app. d, J=8.7 Hz, 1H), 8.06 (app. d, J=8.2 Hz, 1H), 7.89 (app. t, J=7.8 Hz, 1H), 7.71 (app. t, J=7.5 Hz, 1H), 7.66 (app. d, J=8.3 Hz, 2H), 7.46 (app. d, J=8.3 Hz, 2H), 1.34 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 178.87, 158.07, 154.94, 150.65, 133.40, 133.24, 131.62, 128.60, 128.49, 128.26, 126.66, 125.96, 125.45, 116.92, 93.58, 87.77, 35.26, 31.17. IR (neat): ν 3048.74 (C—H stretch), 2957.79 (C—H stretch), 2904.05 (C—H stretch), 2864.78 (C—H stretch), 2193.02 (C≡C stretch), 1628.75 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 314.1545, found 314.1541. MP: Tm 102.5-104.4.
General procedure A for the synthesis of propynones was followed using 2-bromo-4-methylaniline (62 μL) and 1-(tert-butyl)-4-ethynylbenzene (180 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5), furnishing the title compound as a dark brown oil in 29% yield.
1H NMR (400 MHz, CDCl3): δ 7.96 (s, 1H), 7.61 (app. d, J=8.3 Hz, 2H), 7.44 (app. d, J=8.3 Hz, 2H), 7.15 (app. d, J=8.4 Hz, 1H), 6.60 (app. d, J=8.4 Hz, 1H), 6.25 (br, s, 2H), 2.30 (s, 3H), 1.35 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 179.51, 153.97, 149.08, 136.58, 133.79, 132.60, 125.65, 125.06, 118.85, 117.60, 116.88, 92.72, 87.01, 35.03, 31.08, 20.42. IR (neat): ν 3445.59 (N—H stretch), 3333.98 (N—H stretch), 3036.34 (C—H stretch), 2959.86 (C—H stretch), 2904.05 (C—H stretch), 2866.85 (C—H stretch), 2190.96 (C≡C stretch), 1628.75 (C═O stretch). LR-MS (ESI): m/z calculated for [M+H]+ 292.1701, found 292.2.
All data are in accordance with Veryser et al. Adv. Synth. Catal. 2017, 359 (8), 1271-1276.
General procedure A for the synthesis of propynones was followed using 2-bromo-N-methylaniline (59 μL) and 1-(tert-butyl)-4-ethynylbenzene (180 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 98/2), furnishing the desired product as a dark yellow solid in 41% yield.
1H NMR (400 MHz, CDCl3): δ 8.80 (br, d, J=5.1 Hz, 1H), 8.24 (app. dd, J=8.0, 1.2 Hz, 1H), 7.61 (app. d, J=8.4 Hz, 2H), 7.45-7.38 (m, 3H), 6.72-6.63 (m, 2H), 2.97 (d, J=5.1 Hz, 3H), 1.35 (s, 9H). 13C NMR (151 MHz, CDCl3): δ 179.49, 153.93, 152.75, 135.90, 135.29, 132.63, 125.65, 118.59, 117.65, 114.45, 111.02, 92.80, 86.98, 35.07, 31.11, 29.35. IR (neat): ν 3317.44 (N—H stretch), 3081.81 (C—H stretch), 2955.73 (C—H stretch), 2924.72 (C—H stretch), 2868.91 (C—H stretch), 2819.31 (C—H stretch), 2199.23 (C≡C stretch), 1616.35 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 292.1701, found 292.1696. MP: Tm 141.6-142.8.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and ethynylbenzene (87 μL) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 9/1 to 6/4), furnishing the desired product as a dark yellow solid in 41% yield.
1H NMR (300 MHz, CDCl3): δ 8.46 (app. dd, J=7.8, 1.9 Hz, 1H), 8.29 (app. dd, J=4.7, 1.9 Hz, 1H), 7.69-7.64 (m, 2H), 7.53-739 (m, 3H), 6.74 (app. dd, J=7.8, 4.7 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 178.43, 159.15, 155.21, 143.26, 133.04, 130.90, 128.86, 120.25, 114.36, 112.99, 93.66, 86.50. IR (neat): ν 3400.12 (N—H stretch), 3253.37 (N—H stretch), 3092.14 (C—H stretch), 2916.45 (C—H stretch), 2850.31 (C—H stretch), 2190.96 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 223.0871, found 223.0864. MP: Tm 146.2-147.8.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-ethynyl-4-fluorobenzene (87 μL) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2 to 7/3), yielding 50% of the desired product as a light brown solid.
1H NMR (400 MHz, CDCl3): δ 8.42 (app. dd, J=7.9, 1.9 Hz, 1H), 8.29 (app. dd, J=4.7, 1.9 Hz, 1H), 7.70-7.63 (m, 2H), 7.16-7.09 (m, 2H), 6.73 (app. dd, J=7.9, 4.7 Hz, 1H). 13C NMR (101 MHz, CDCl3): δ 178.26, 164.13 (d, J=253.8 Hz), 159.15, 155.25, 143.17, 135.29 (d, J=8.8 Hz), 116.41 (d, J=22.2 Hz), 116.35 (d, J=3.7 Hz), 114.24, 112.96, 92.55, 86.39. IR (neat): ν 3398.05 (N—H stretch), 3263.70 (N—H stretch), 3112.81 (C—H stretch), 2924.72 (C—H stretch), 2190.96 (C≡C stretch), 1620.48 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 241.0777, found 241.0770. MP: Tm 172.5-173.5.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and ethynyltriisopropylsilane (173 μL) at 80° C. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2 to 7/3), yielding 23% of the desired product as a bright yellow solid.
1H NMR (300 MHz, CDCl3): δ 8.41 (app. dd, J=7.9, 1.9 Hz, 1H), 8.27 (app. dd, J=4.7, 1.9 Hz, 1H), 6.70 (app. dd, J=7.9, 4.7 Hz, 1H), 1.16 (sept, J=4.2 Hz, 3H), 1.15 (d, J=4.6 Hz, 18H). 13C NMR (101 MHz, CDCl3): δ 177.94, 159.12, 154.99, 143.31, 114.28, 112.98, 102.65, 98.50, 18.72, 11.26. IR (neat): ν 3395.98 (N—H stretch), 3271.97 (N—H stretch), 3125.22 (C—H stretch), 2945.39 (C—H stretch), 2862.71 (C—H stretch), 2151.69 (C≡C stretch), 1612.21 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 303.1893, found 303.1889. MP: Tm 94.6-95.7.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and ethynylcyclohexane (100 μL) at 80° C. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM/EtOAc 90/10), followed by column chromatography on silica gel (DCM to DCM/EtOAc/MeOH 8/1/1), yielding 56% of the title compound as a bright yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.34 (app. dd, J=7.8, 1.8 Hz, 1H), 8.25 (app. dd, J=4.7, 1.8 Hz, 1H), 6.69 (app. dd, J=7.8, 4.7 Hz, 1H), 2.74-2.62 (m, 1H), 1.98-1.86 (m, 2H), 1.82-1.70 (m, 2H), 1.69-1.49 (m, 4H), 1.46-1.32 (m, 3H). 13C NMR (151 MHz, CDCl3): δ 178.89, 159.09, 154.76, 143.43, 114.43, 112.84, 100.82, 79.17, 31.86, 29.53, 25.78, 24.89. IR (neat): ν 3398.05 (N—H stretch), 3265.77 (N—H stretch), 3125.22 (C—H stretch), 2924.72 (C—H stretch), 2852.38 (C—H stretch), 2201.29 (C≡C stretch), 1608.08 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 229.1341, found 229.1335. MP: Tm 137.5-138.6.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine13 (88 mg) and hept-1-yne (100 μL) at 80° C. The crude reaction mixture was purified via column chromatography on silica gel (DCM/EtOAc 9/1 to 8/2), yielding 29% of the desired product as a bright yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.34 (app. dd, J=7.8, 1.8 Hz, 1H), 8.25 (app. dd, J=4.7, 1.8 Hz, 1H), 6.69 (app. dd, J=7.8, 4.7 Hz, 1H), 2.49 (t, J=7.2 Hz, 2H), 1.73-1.62 (m, 2H), 1.50-1.31 (m, 4H), 0.93 (t, J=7.2 Hz, 3H). 13C NMR (151 MHz, CDCl3): δ 178.80, 159.11, 154.85, 143.43, 114.35, 112.85, 97.35, 79.30, 31.28, 27.66, 22.27, 19.32, 14.06. IR (neat): ν 3400.12 (N—H stretch), 3259.57 (N—H stretch), 3116.95 (C—H stretch), 2959.86 (C—H stretch), 2930.92 (C—H stretch), 2852.38 (C—H stretch), 2211.63 (C≡C stretch), 1610.15 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 217.1341, found 217.1337. MP: Tm 110.7-111.7.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 3-methylbut-1-yne (81 μL) at 80° C. The crude reaction mixture was purified via column chromatography on silica gel (DCM/MeOH 99.5/0.5), yielding 26% of the desired product as a dark yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.33 (app. dd, J=7.8, 1.8 Hz, 1H), 8.25 (app. dd, J=4.7, 1.8 Hz, 1H), 6.69 (app. dd, J=7.8, 4.7 Hz, 1H), 2.85 (sept, J=7.0 Hz, 1H), 1.32 (d, J=6.9 Hz, 6H). 13C NMR (151 MHz, CDCl3): δ 178.85, 159.10, 154.84, 143.42, 114.33, 112.84, 101.86, 78.38, 22.18, 21.12. IR (neat): ν 3416.65 (N—H stretch), 3271.97 (N—H stretch), 3131.42 (C—H stretch), 2970.19 (C—H stretch), 2926.79 (C—H stretch), 2868.91 (C—H stretch), 2203.36 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 189.1028, found 189.1023. MP: Tm 91.7-92.8.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-chloro-4-ethynylbenzene (102 mg) at 80° C. The crude reaction mixture was purified via column chromatography on silica gel (DCM), yielding 35% of the desired product as a bright yellow solid.
1H NMR (600 MHz, CDCl3): δ 8.43 (app. dd, J=7.8, 1.8 Hz, 1H), 8.32 (app. dd, J=4.7, 1.8 Hz, 1H), 7.62 (app. dd, J=8.6, 2.0 Hz, 2H), 7.43 (app. dd, J=8.6, 2.0 Hz, 2H), 6.76 (app. dd, J=7.8, 4.7 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 178.13, 159.15, 155.35, 143.17, 137.29, 134.20, 129.33, 118.69, 114.19, 112.98, 92.19, 87.18. IR (neat): ν 3412.52 (N—H stretch), 3261.63 (N—H stretch), 3085.94 (C—H stretch), 2916.45 (C—H stretch), 2848.24 (C—H stretch), 2205.43 (C≡C stretch), 1610.15 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 257.0482, found 257.0474. MP: Tm 183.3-184.2.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and methyl 4-ethynylbenzoate (123 mg) at 80° C. The crude reaction mixture was purified via column chromatography on silica gel (Et2O), furnishing the desired product as a dark yellow solid in 18% yield.
1H NMR (400 MHz, CDCl3): δ 8.43 (app. dd, J=7.9, 1.8 Hz, 1H), 8.31 (app. dd, J=4.7, 1.8 Hz, 1H), 8.09 (app. d, J=8.3 Hz, 2H), 7.72 (app. d, J=8.3 Hz, 2H), 6.74 (app. dd, J=7.9, 4.7 Hz, 1H), 3.95 (s, 3H). 13C NMR (151 MHz, CDCl3): δ 178.00, 166.23, 159.17, 155.50, 143.21, 132.83, 131.88, 129.88, 124.71, 114.16, 113.04, 91.90, 88.19, 52.63. IR (neat): ν 3418.72 (N—H stretch), 3259.57 (N—H stretch), 3116.95 (C—H stretch), 2922.65 (C—H stretch), 2852.38 (C—H stretch), 2203.36 (C≡C stretch), 1719.69 (C═O stretch ester), 1612.21 (C═O stretch ketone). HR-MS (ESI): m/z calculated for [M+H]+ 281.0926, found 281.0926. MP: Tm 182.4-183.7 (decomposition).
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-ethynyl-4-methoxybenzene (100 mg) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (DCM/Et2O 95/5 to 9/1), yielding 73% of the desired product as a yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.44 (app. dd, J=7.8, 1.8 Hz, 1H), 8.27 (app. dd, J=4.7, 1.8 Hz, 1H), 7.65-7.58 (m, 2H), 7.10-6.78 (m, 2H), 6.72 (app. dd, J=7.8, 4.7 Hz, 1H), 3.85 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.50, 161.81, 159.14, 154.90, 143.17, 135.04, 114.57, 114.42, 112.86, 111.99, 94.73, 86.39, 55.58. IR (neat): ν 3402.19 (N—H stretch), 3261.63 (N—H stretch), 3131.42 (C—H stretch), 2930.92 (C—H stretch), 2835.84 (C—H stretch), 2188.89 (C≡C stretch), 1599.81 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 253.0977, found 253.0972. MP: Tm 175.7-177.0.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-chloro-3-ethynylbenzene (96 μL) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (DCM/MeOH 99.9/0.1). Spectroscopic analysis of the product fraction indicated the presence of some residual aryl halide. Therefore, the product mixture was dissolved in Et2O and subsequently washed with 0.5 M aqueous HCl, saturated aqueous NaHCO3, 0.5 M aqueous HCl, saturated aqueous NaHCO3 and brine. The organic phase was dried over MgSO4, filtered and concentrated in vacuo. The title compound was obtained as a dark yellow solid in 23% yield.
1H NMR (300 MHz, CDCl3): δ 8.41 (app. dd, J=7.9, 1.9 Hz, 1H), 8.30 (app. dd, J=4.7, 1.9 Hz, 1H), 7.65 (app. t, J=1.7 Hz, 1H), 7.55 (app. dd, J=7.5, 2.6 Hz, 1H), 7.47 (app. ddd, J=8.1, 2.1, 1.2 Hz, 1H), 7.37 (app. t, J=7.8 Hz, 1H), 6.74 (app. dd, J=7.9, 4.7 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 178.01, 159.15, 155.40, 143.22, 134.80, 132.64, 131.14, 131.09, 130.13, 121.96, 114.18, 113.03, 91.53, 86.99. IR (neat): ν 3418.72 (N—H stretch), 3267.83 (N—H stretch), 3112.81 (C—H stretch), 2920.59 (C—H stretch), 2852.38 (C—H stretch), 2199.23 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 257.0482, found 257.0475. MP: Tm 150.1-150.8.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-ethynyl-4-(trifluoromethyl)benzene (125 μL) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM to DCM/EtOAc 95/5). Spectroscopic analysis of the product fraction indicated the presence of some residual aryl halide. Therefore, the product mixture was dissolved in Et2O and subsequently washed with 0.5 M aqueous HCl, saturated aqueous NaHCO3, 0.5 M aqueous HCl, saturated aqueous NaHCO3 and brine. The organic phase was dried over MgSO4, filtered and concentrated in vacuo. The title compound was obtained as a dark yellow oil in 21% yield.
1H NMR (600 MHz, CDCl3): δ 8.42 (app. dd, J=7.9, 1.9 Hz, 1H), 8.30 (app. dd, J=4.7, 1.9 Hz, 1H), 7.77 (app. d, J=8.1 Hz, 2H), 7.69 (app. d, J=8.1 Hz, 2H), 6.75 (app. dd, J=7.9, 4.7 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 177.73, 159.01, 155.29, 143.11, 133.01, 132.25 (q, J=33.0 Hz), 125.66 (q, J=3.8 Hz), 123.90, 123.55 (q, J=272.5 Hz), 113.99, 112.87, 91.01, 87.58. IR (neat): ν 3404.25 (N—H stretch), 3278.17 (N—H stretch), 3131.42 (C—H stretch), 2955.73 (C—H stretch), 2920.59 (C—H stretch), 2852.38 (C—H stretch), 2207.49 (C≡C stretch), 1612.21 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 291.0745, found 291.0739.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-ethynyl-4-methylbenzene (97 μL) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM/EtOAc 95/5 to 9/1), yielding 74% of the desired product as a bright yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.45 (app. dd, J=7.8, 1.9 Hz, 1H), 8.28 (app. dd, J=4.7, 1.9 Hz, 1H), 7.56 (app. d, J=8.0 Hz, 2H), 7.23 (app. d, J=8.0 Hz, 2H), 6.73 (app. dd, J=7.8, 4.7 Hz, 1H), 2.41 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.52, 159.13, 155.05, 143.27, 141.64, 133.07, 129.65, 117.12, 114.43, 112.95, 94.34, 86.36, 21.92. IR (neat): ν 3418.72 (N—H stretch), 3255.43 (N—H stretch), 3119.01 (C—H stretch), 3034.27 (C—H stretch), 2916.45 (C—H stretch), 2850.31 (C—H stretch), 2197.16 (C≡C stretch), 1606.01 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 237.1028, found 237.1022. MP: Tm 158.3-159.2.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1,2-dichloro-4-ethynylbenzene (132 mg) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM to DCM/EtOAc 9/1 to 6/4), followed by column chromatography on silica gel (DCM/EtOAc 8/2), furnishing the desired product as a bright yellow solid in 17% yield.
1H NMR (400 MHz, CDCl3): δ 8.38 (app. dd, J=7.9, 1.9 Hz, 1H), 8.31 (app. dd, J=4.7, 1.9 Hz, 1H), 7.75 (app. dd, J=1.7, 0.4 Hz, 1H), 7.51 (app. dd, J=8.3, 0.4 Hz, 1H), 7.48 (app. dd, J=8.3, 1.7 Hz, 1H), 6.74 (app. dd, J=7.9, 4.7 Hz, 1H). 13C NMR (101 MHz, CDCl3): δ 177.80, 159.15, 155.55, 143.12, 135.70, 134.40, 133.38, 131.93, 131.03, 120.15, 114.07, 113.06, 90.43, 87.58. IR (neat): ν 3418.72 (N—H stretch), 3263.70 (N—H stretch), 3104.55 (C—H stretch), 3026.00 (C—H stretch), 2197.16 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 291.0092, found 291.0085. MP: Tm 182.1-182.9.
General procedure A for the synthesis of propynones was followed using 2-bromoaniline (58 μL) and 1-(tert-butyl)-4-ethynylbenzene (141 μL) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/DCM 5/5), furnishing the desired product as a yellow solid in 36% yield.
1H NMR (300 MHz, CDCl3): δ 8.19 (app. dd, J=8.1, 1.4 Hz, 1H), 7.61 (app. d, J=8.6 Hz, 2H), 7.43 (app. d, J=8.6 Hz, 2H), 7.32 (app. ddd, J=8.5, 7.1, 1.6 Hz, 1H), 6.72 (app. ddd, J=8.1, 7.1, 1.0 Hz, 1H), 6.66 (app. dd, J=8.4, 0.6 Hz, 1H), 6.36 (br, s, 2H), 1.34 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 179.63, 154.05, 151.04, 135.19, 134.48, 132.68, 125.65, 118.96, 117.46, 116.76, 116.08, 92.87, 86.90, 35.03, 31.07. IR (neat): ν 3426.99 (N—H stretch), 3317.44 (N—H stretch), 3075.61 (C—H stretch), 2959.86 (C—H stretch), 2920.59 (C—H stretch), 2850.31 (C—H stretch), 2186.82 (C≡C stretch), 1616.35 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 278.1545, found 278.1539. MP: Tm 74.1-75.2.
General procedure A for the synthesis of propynones was followed using 3-bromopyridine (50 μL) and 1-(tert-butyl)-4-ethynylbenzene (141 μL) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via flash column chromatography on silica gel (DCM to DCM/MeOH 99/1), yielding 24% of the title compound as a brown oil.
1H NMR (300 MHz, CDCl3): δ 9.46 (app. dd, J=2.2, 0.8 Hz, 1H), 8.84 (app. dd, J=4.8, 1.7 Hz, 1H), 8.44 (app. dt, J=8.0, 2.0 Hz, 1H), 7.65 (app. d, J=8.6 Hz, 2H), 7.48 (app. dd, J=7.9, 0.9 Hz, 1H), 7.47 (app. d, J=8.6 Hz, 2H), 1.35 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 176.63, 155.34, 154.27, 151.64, 136.33, 133.38, 132.51, 126.06, 123.69, 116.62, 95.69, 86.36, 35.34, 31.19. IR (neat): ν 3040.47 (C—H stretch), 2957.79 (C—H stretch), 2926.79 (C—H stretch), 2852.38 (C—H stretch), 2195.09 (C≡C stretch), 1641.15 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 264.1388, found 264.1392.
General procedure A for the synthesis of propynones was followed using bromobenzene (1.07 mL, 10.00 mmol, 1.00 equiv.) and 1-(tert-butyl)-4-ethynylbenzene (2.82 mL, 15.00 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/DCM 8/2), yielding 80% of the title compound as a yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.26-8.20 (m, 2H), 7.65-7.58 (m, 3H), 7.54-7.48 (m, 2H), 7.46-7.41 (m, 2H), 1.33 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 178.07, 154.58, 137.01, 134.00, 132.99, 129.54, 128.59, 125.76, 117.03, 93.80, 86.75, 35.09, 31.05. IR (neat): ν 3038.40 (C—H stretch), 2963.99 (C—H stretch), 2904.05 (C—H stretch), 2866.85 (C—H stretch), 2190.96 (C≡C stretch), 1634.95 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 263.1436, found 263.1431. MP: Tm 49.2-50.2.
All data are in accordance with Wu et al. 2010 cited above and Liu et al. Org. Lett. 2008, 10 (18), 3933-3936.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-ethynyl-3-(trifluoromethyl)benzene (111 μL) at 80° C. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (DCM/Et2O 9/1), furnishing the desired product as a bright yellow solid in 26% yield.
1H NMR (400 MHz, CDCl3): δ 8.42 (app. dd, J=7.9, 1.9 Hz, 1H), 8.31 (app. dd, J=4.7, 1.9 Hz, 1H), 7.91 (app. s, 1H), 7.83 (app. d, J=7.8 Hz, 1H), 7.73 (app. d, J=7.9 Hz, 1H), 7.57 (app. dd, J=7.9, 7.8 Hz, 1H), 6.75 (app. dd, J=7.9, 4.7 Hz, 1H). 13C NMR (101 MHz, CDCl3): δ 177.89, 159.16, 155.50, 143.21, 135.99, 131.62 (q, J=33.1 Hz), 129.65 (q, J=3.8 Hz), 129.51, 127.34 (q, J=3.7 Hz), 123.53 (q, J=272.7 Hz), 121.30, 114.10, 113.06, 91.11, 87.18. IR (neat): ν 3414.59 (N—H stretch), 3269.90 (N—H stretch), 3108.68 (C—H stretch), 2918.52 (C—H stretch), 2201.29 (C≡C stretch), 1612.21 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 291.0745, found 291.0736. MP: Tm 168.3-168.9.
General procedure A for the synthesis of propynones was followed using 1-bromo-2-methoxybenzene (1.28 mL, 10.00 mmol, 1.00 equiv.) and 1-(tert-butyl)-4-ethynylbenzene (2.82 mL, 15.00 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (heptane/DCM 4/6), yielding 9% of the title compound as a dark brown oil.
1H NMR (600 MHz, CDCl3): δ 8.08 (app. dd, J=7.9, 1.6 Hz, 1H), 7.58 (app. d, J=8.3 Hz, 2H), 7.54 (app. ddd, J=8.4, 7.4, 1.6 Hz, 1H), 7.42 (app. d, J=8.3 Hz, 2H), 7.05 (app. ddd, J=7.9, 7.4 Hz, 1H), 7.02 (app. d, J=8.4 Hz, 1H), 3.97 (s, 3H), 1.33 (s, 9H). 13C NMR (151 MHz, CDCl3): δ 176.90, 159.79, 154.18, 134.87, 132.91, 132.64, 126.96, 125.67, 120.31, 117.64, 112.21, 92.29, 89.05, 55.95, 35.07, 31.08. IR (neat): ν 3071.47 (C—H stretch), 2961.93 (C—H stretch), 2866.85 (C—H stretch), 2837.91 (C—H stretch), 2195.09 (C≡C stretch), 1618.41 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 293.1542, found 293.1538.
Protocol based on Ma et al. Org. Lett. 2016, 18 (6), 1322-1325.
In a round-bottom flask, rac-2-(3-(4-(tert-butyl)phenyl)-1-hydroxyprop-2-yn-1-yl)phenol (3.2, 800 mg, 2.85 mmol, 1.00 equiv.) was stirred in the presence of manganese dioxide (1.736 g, 19.97 mmol, 7.00 equiv.) for 3 h at room temperature. The suspension was filtered over Celite® 535 and the filtrate was concentrated under reduced pressure. The residue was purified via column chromatography (heptane/EtOAc 95/5). The title compound was obtained as a bright yellow solid in 22% yield.
1H NMR (600 MHz, CDCl3): δ 11.79 (s, 1H), 8.13 (app. dd, J=7.9, 1.3 Hz, 1H), 7.64 (app. d, J=8.4 Hz, 2H), 7.53 (app. ddd, J=8.8, 7.2, 1.7 Hz, 1H), 7.46 (app. d, J=8.4 Hz, 2H), 7.02-6.98 (m, 2H), 1.35 (s, 9H). 13C NMR (151 MHz, CDCl3): δ 182.51, 162.94, 155.21, 137.16, 133.22, 133.17, 126.00, 121.00, 119.50, 118.26, 116.74, 96.94, 85.72, 35.31, 31.18. IR (neat): ν 3046.67 (C—H stretch), 2957.79 (C—H stretch), 2924.72 (C—H stretch), 2868.91 (C—H stretch), 2197.16 (C≡C stretch), 1618.41 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 279.1385, found 279.1378. MP: Tm 131.1-136.7 (decomposition).
General procedure B for the synthesis of propynones was followed using 2-chloronicotinaldehyde (283 mg, 2.00 mmol, 1.00 equiv.) and 1-chloro-3-ethynylbenzene (0.74 mL, 6.00 mmol, 3.00 equiv.). The quantities of the other reagents and solvents were adapted accordingly. The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2). The title compound was obtained as an off-white solid in 83% overall yield.
1H NMR (400 MHz, CDCl3): δ 8.63-8.53 (m, 1H), 8.37-8.29 (m, 1H), 7.65 (app. s, 1H), 7.59-7.52 (m, 1H), 7.52-7.46 (m, 1H), 7.46-7.41 (m, 1H), 7.38 (app. t, J=7.8 Hz, 1H). 13C NMR (101 MHz, CDCl3): δ 175.34, 152.66, 149.67, 140.84, 134.85, 132.87, 132.48, 131.68, 131.37, 130.18, 122.59, 121.43, 93.47, 88.36. IR (neat): ν 3119.01 (C—H stretch), 3065.27 (C—H stretch), 2203.36 (C≡C stretch), 1632.88 (C═O stretch). IR (neat): ν 3119.01 (C—H stretch), 3065.27 (C—H stretch), 2203.36 (C≡C stretch), 1632.88 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 275.9983, found 275.9979. MP: Tm 114.1-114.8.
General procedure B for the synthesis of propynones was followed using 2-morpholinonicotinaldehyde (384 mg, 2.00 mmol) and 1-chloro-3-ethynylbenzene (0.74 mL, 6.00 mmol). The quantities of the other reagents and solvents were adapted accordingly. The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/EtOAc 75/25). The purified product was obtained as a yellow liquid, which could be solidified into a bright yellow solid by crash precipitation in n-pentane. The title compound was obtained in 92% overall yield.
1H NMR (400 MHz, CDCl3): δ 8.39-8.33 (m, 2H), 7.61 (app. s, 1H), 7.55-7.50 (m, 1H), 7.48-7.43 (m, 1H), 7.39-7.33 (m, 1H), 6.85 (m, 1H), 3.84 (br, app. t, J=4.3 Hz, 4H), 3.52 (br, app. t, J=4.3 Hz, 4H). 13C NMR (101 MHz, CDCl3): δ 176.40, 159.15, 152.32, 143.04, 134.80, 132.72, 131.14, 131.12, 130.14, 122.01, 119.51, 114.43, 89.84, 88.42, 66.94, 50.20. IR (neat): ν 3065.27 (C—H stretch), 2970.19 (C—H stretch), 2856.51 (C—H stretch), 2197.16 (C≡C stretch), 1628.75 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 327.0900, found 327.0891. MP: Tm 87.6-88.5.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and 1-(tert-butyl)-4-ethynylbenzene (181 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5 to 6/4), yielding 64% of the desired product as a bright yellow solid.
1H NMR (400 MHz, CDCl3): δ 8.45 (app. d, J=7.8 Hz, 1H), 8.26 (app. d, J=3.2 Hz, 1H), 7.60 (app. d, J=8.2 Hz, 2H), 7.43 (app. d, J=8.2 Hz, 2H), 6.72 (app. dd, J=7.8, 3.2 Hz, 1H), 1.33 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 178.43, 159.16, 154.74, 154.63, 143.38, 132.89, 125.87, 117.06, 114.44, 112.75, 94.27, 86.27, 35.20, 31.16. IR (neat): ν 3389.78 (N—H stretch), 3263.70 (N—H stretch), 3133.48 (C—H stretch), 3038.40 (C—H stretch), 2959.86 (C—H stretch), 2866.85 (C—H stretch), 2186.82 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 279.1497, found 279.1494. MP: Tm 164.1-164.9.
General procedure A for the synthesis of propynones was followed using 3-bromopyridin-2-amine (88 mg) and oct-1-yne (147 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 9/1 to 8/2), yielding 49% of the desired product as a brown solid.
1H NMR (400 MHz, CDCl3) δ 8.35 (app. dd, J=7.9, 1.4 Hz, 1H), 8.23 (app. dd, J=4.7, 1.4 Hz, 1H), 6.67 (app. dd, J=7.9, 4.7 1H), 2.48 (t, J=7.1 Hz, 2H), 1.65 (app. quint, J=7.4 Hz, 2H), 1.45 (app. quint, J=7.1 Hz, 2H), 1.31 (m, 4H), 0.90 (t, J=7.1 Hz, 3H). 13C NMR (101 MHz, CDCl3): δ 178.66, 159.15, 154.34, 143.69, 114.44, 112.59, 97.33, 79.25, 31.33, 28.77, 27.89, 22.60, 19.31, 14.12. IR (neat): ν 3420.79 (N—H stretch), 3305.04 (N—H stretch), 2953.66 (C—H stretch), 2918.52 (C—H stretch), 2852.38 (C—H stretch), 2213.69 (C≡C stretch), 1620.48 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 231.1497, found 231.1494. MP: Tm 75.8-77.6.
General procedure A for the synthesis of propynones was followed using 2-bromopyridin-3-amine (88 mg) and 1-(tert-butyl)-4-ethynylbenzene (181 μL, 1.00 mmol, 2.00 equiv.) at 100° C. In both chambers, 2 mL of solvent was used. The crude reaction mixture was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2), furnishing the title compound as a light brown solid in 25% yield.
1H NMR (400 MHz, CDCl3): δ 8.02 (m, 1H), 7.51 (app. d, J=8.3 Hz, 2H), 7.38 (app. d, J=8.3 Hz, 2H), 7.02 (m, 2H), 4.31 (br, s, 2H), 1.32 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 152.34, 144.40, 140.08, 132.00, 131.66, 128.85, 125.58, 125.54, 123.79, 121.10, 119.41, 95.20, 84.67, 34.94, 31.23. IR (neat): ν 3360.85 (N—H stretch), 3048.74 (C—H stretch), 2959.86 (C—H stretch), 2901.99 (C—H stretch), 2866.85 (C—H stretch), 2215.76 (C≡C stretch), 1686.62 (C═O stretch). MP: Tm 125.8-127.2.
General procedure B for the synthesis of propynones was followed using nicotinaldehyde (93 μL) and 1-chloro-3-ethynylbenzene (0.37 mL). The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/acetone 97/3). The purified product was obtained as a dark yellow solid in 99% overall yield.
1H NMR (400 MHz, CDCl3): δ 9.42 (app. s, 1H), 8.86 (app. dd, J=4.7, 1.1 Hz, 1H), 8.42 (app. dt, J=7.8, 1.6 Hz, 1H), 7.7-7.66 (m, 1H), 7.61-7.56 (m, 1H), 7.52-7.45 (m, 2H), 7.42-7.35 (m, 1H). 13C NMR (101 MHz, CDCl3): δ 176.15, 154.46, 151.43, 136.22, 134.77, 132.83, 132.02, 131.49, 131.27, 130.10, 123.61, 121.29, 92.42, 86.69. IR (neat): ν 3048.74 (C—H stretch), 2961.93 (C—H stretch), 2201.29 (C≡C stretch), 1632.88 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 242.0373, found 242.0369. MP: Tm 106.2-106.6.
All data are in accordance with CN102256487
General procedure B for the synthesis of propynones was followed using 1H-imidazole-2-carbaldehyde (96 mg) and 1-chloro-3-ethynylbenzene (0.37 mL). The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/EtOAc 5/5). The purified product was obtained as a beige solid in 53% overall yield.
1H NMR (400 MHz, DMSO-d6): δ 13.71 (br, s, 1H), 7.82-7.77 (m, 1H), 7.72-7.63 (m, 2H), 7.59 (br s, 1H), 7.57-7.51 (m, 1H), 7.31 (br, s, 1H). 13C NMR (101 MHz, DMSO-d6): δ 166.35, 145.63, 133.63, 132.26, 132.00, 131.49, 131.30, 131.02, 123.30, 121.25, 89.89, 87.71. IR (neat): ν 3121.08 (C—H stretch), 2966.06 (C—H stretch), 2850.31 (C—H stretch), 2199.23 (C≡C stretch), 1622.55 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 231.0325, found 231.0322. MP: Tm 170.1-170.8.
General procedure B for the synthesis of propynones was followed using 2-(dimethylamino)nicotinaldehyde (151 mg) and 1-chloro-3-ethynylbenzene (0.37 mL). The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5). The purified product was obtained as a dark yellow solid in 91% overall yield.
1H NMR (400 MHz, CDCl3): δ 8.37 (app. dd, J=7.5, 1.8 Hz, 1H), 8.32 (app. dd, J=4.7, 1.8 Hz, 1H), 7.62 (app. s, 1H), 7.55-7.49 (m, 1H), 7.46-7.41 (m, 1H), 7.35 (app. t, J=7.8 Hz, 1H), 6.72 (app. dd, J=7.9, 4.6 Hz, 1H), 3.08 (s, 6H). 13C NMR (101 MHz, CDCl3): δ 175.18, 158.43, 151.88, 142.46, 134.51, 132.44, 130.90, 130.71, 129.93, 122.07, 117.12, 111.84, 89.21, 88.31, 41.40. IR (neat): ν 3063.21 (C—H stretch), 2922.65 (C—H stretch), 2850.31 (C—H stretch), 2199.23 (C≡C stretch), 1614.28 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 285.0795, found 285.0794. MP: Tm 49.4-52.1.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and ethynylbenzene (0.41 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 9/1 to 7/3), furnishing the title compound as a yellow solid in 76% yield.
1H NMR (400 MHz, CDCl3): δ 8.26-8.20 (m, 2H), 7.72-7.67 (m, 2H), 7.64 (app. t, J=7.4 Hz, 1H), 7.56-7.46 (m, 3H), 7.43 (app. t, J=7.3 Hz, 2H). 13C NMR (101 MHz, CDCl3): δ 178.05, 136.93, 134.14, 133.10, 130.82, 129.61, 128.72, 128.65, 120.17, 93.13, 86.92. IR (neat): ν 3050.81 (C—H stretch), 2195.09 (C≡C stretch), 1630.82 (C═O stretch). LR-MS (ESI): m/z calculated for [M+H]+ 207.0810, found 207.1. MP: Tm 48.4-48.9 (literature 46-48).
All data are in accordance with Wu et al. 2010 cited above, Liu et al. 2008 cited above, Iman et al. Synthesis 1990, 1990 (07), 631-632, Chen et al, Tetrahedron 2009, 65 (49), 10134-10141 and Zhang et al. Org. Biomol. Chem. 2020, 18 (6), 1073-1077.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-4-methoxybenzene (0.49 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 9/1 to 3/7) and subsequent recrystallization (heptane/i-PrOH 8/2), furnishing the title compound as a yellow solid in 83% yield.
1H NMR (400 MHz, CDCl3): δ 8.25-8.19 (m, 2H), 7.68-7.59 (m, 3H), 7.52 (app. t, J=7.6 Hz, 2H), 6.94 (app. d, J=8.8 Hz, 2H), 3.87 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.21, 161.90, 137.24, 135.31, 134.05, 129.66, 128.72, 114.59, 112.10, 94.46, 87.04, 55.61. IR (neat): ν 3052.87 (C—H stretch), 3013.60 (C—H stretch), 2842.04 (C—H stretch), 2182.69 (C≡C stretch), 1624.62 (C═O stretch). LR-MS (ESI): m/z calculated for [M+Na]+ 259.0735, found 259.1. MP: Tm 81.5-82.6 (literature 80-82).
All data are in accordance with with Wu et al. 2010 cited above, Liu et al. 2008 cited above, Iman et al. 1990 cited above, Chen et al 2009 cited above, Zhang et al. Org. Biomol. Chem. 2020, 18 (6), 1073-1077, Bishop et al. Synthesis 2004, 2004 (01), 43-52, Jeong et al. J. Org. Chem. 2014, 79 (14), 6444-6455, and Zhang et al. Org. Biomol. Chem. 2014, 12 (47), 9702-9706.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-4-methylbenzene (0.48 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 8/2 to 5/5) and subsequent recrystallization (heptane), furnishing the title compound as a yellow solid in 64% yield.
1H NMR (400 MHz, CDCl3): δ 8.26-8.19 (m, 2H), 7.67-7.56 (m, 3H), 7.52 (app. t, J=7.6 Hz, 2H), 7.23 (app. d, J=7.9 Hz, 2H), 2.41 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.23, 141.71, 137.45, 134.15, 133.28, 129.70, 129.64, 128.74, 117.18, 93.96, 86.94, 21.92. IR (neat): ν 3023.93 (C—H stretch), 2916.45 (C—H stretch), 2850.31 (C—H stretch), 2193.02 (C≡C stretch), 1626.68 (C═O stretch). LR-MS (ESI): m/z calculated for [M+Na]+ 243.0786, found 243.1. MP: Tm 59.8-60.6 (literature 67-71).
All data are in accordance with Wu et al. 2010 cited above, Liu et al. 2008 cited above, Chen et al 2009 cited above, Bishop et al. 2004 cited above, Jeon et al. 2014 cited above and Zhang et al. 2014 cited above.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-chloro-4-ethynylbenzene (512 mg, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 8/2 to 6/4) and subsequent recrystallization (heptane/i-PrOH 8/2), furnishing the title compound as a brown solid in 49% yield.
1H NMR (400 MHz, CDCl3): δ 8.25-8.17 (m, 2H), 7.69-7.59 (m, 3H), 7.53 (app. t, J=7.7 Hz, 2H), 7.42 (app. d, J=8.5 Hz, 2H). 13C NMR (101 MHz, CDCl3): δ 177.98, 137.37, 136.91, 134.42, 134.41, 129.74, 129.34, 128.84, 118.77, 91.76, 87.75. IR (neat): ν 3085.94 (C—H stretch), 3065.27 (C—H stretch), 3057.01 (C—H stretch), 2197.16 (C≡C stretch), 1632.88 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 241.0420, found 241.0423. MP: Tm 104.2-105.8 (literature 105-108).
All data are in accordance with Chen et al. 2009 cited above and Bishop et al. 2004 cited above.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-3-methoxybenzene (0.45 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 7/3 to 3/7), furnishing the title compound as a yellow solid in 55% yield.
1H NMR (400 MHz, CDCl3): δ 8.27-8.19 (m, 2H), 7.64 (app. t, J=7.4 Hz, 1H), 7.53 (app. t, J=7.7 Hz, 2H), 7.37-7.27 (m, 2H), 7.20 (app. s, 1H), 7.07-7.01 (m, 1H), 3.85 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.17, 159.63, 137.04, 134.30, 129.95, 129.75, 128.79, 125.73, 121.23, 117.75, 117.71, 93.16, 86.73, 55.59. IR (neat): ν 3011.53 (C—H stretch), 2961.93 (C—H stretch), 2935.06 (C—H stretch), 2839.98 (C—H stretch), 2190.96 (C≡C stretch), 1632.88 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 237.0916, found 237.0911. MP: Tm 73.9-74.8.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-3-methylbenzene (0.48 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 8/2 to 5/5), furnishing the title compound as a brown solid in 55% yield.
1H NMR (400 MHz, CDCl3): δ 8.23 (app. d, J=7.4 Hz, 2H), 7.63 (app. t, J=7.2 Hz, 1H), 7.56-7.47 (m, 4H), 7.34-7.27 (m, 2H), 2.39 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.21, 138.68, 137.11, 134.21, 133.71, 131.89, 130.38, 129.72, 128.76, 128.74, 120.09, 93.65, 86.82, 21.34. IR (neat): ν 3061.14 (C—H stretch), 3032.20 (C—H stretch), 2945.39 (C—H stretch), 2916.45 (C—H stretch), 2188.89 (C≡C stretch), 1630.82 (C═O stretch). LR-MS (ESI): m/z calculated for [M+H]+ 221.0966, found 221.1. MP: Tm 30.8-31.8.
All data are in accordance with Zhang et al. 2020 cited above.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-chloro-3-ethynylbenzene (0.46 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 7/3), furnishing the title compound as a beige solid in 29% yield.
1H NMR (600 MHz, CDCl3): δ 8.26-8.20 (m, 2H), 7.71-7.65 (m, 2H), 7.59 (app. d, J=7.6 Hz, 1H), 7.55 (app. t, J=7.7 Hz, 2H), 7.51-7.46 (m, 1H), 7.39 (app. t, J=7.9 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 177.84, 136.81, 134.76, 134.47, 132.79, 131.25, 131.16, 130.11, 129.73, 128.84, 122.00, 91.02, 87.51. IR (neat): ν 3065.27 (C—H stretch), 2201.29 (C≡C stretch), 1630.82 (C═O stretch). LR-MS (ESI): m/z calculated for [M+H]+ 241.0420, found 241.0. MP: Tm 75.1-75.4.
All data are in accordance with Jeong et al. 2014 cited above.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-3-fluorobenzene (0.43 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 8/2 to 6/4), furnishing the title compound as a yellow solid in 20% yield.
1H NMR (600 MHz, CDCl3): δ 8.24 (app. d, J=7.2 Hz, 2H), 7.67 (app. t, J=7.4 Hz, 1H), 7.56 (app. t, J=7.7 Hz, 2H), 7.50 (app. d, J=7.6 Hz, 1H), 7.46-7.38 (m, 2H), 7.23 (app. td, J=8.4, 1.6 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 177.78, 162.33 (d, J=248.1 Hz), 136.72, 134.34, 130.45 (d, J=8.5 Hz), 129.62, 128.95 (d, J=3.2 Hz), 128.72, 121.97 (d, J=9.3 Hz), 119.69 (d, J=23.2 Hz), 118.25 (d, J=21.2 Hz), 91.08 (d, J=3.5 Hz), 87.14. IR (neat): ν 3061.14 (C—H stretch), 2195.09 (C≡C stretch), 1649.42 (C═O stretch). LR-MS (ESI): m/z calculated for [M+Na]+ 247.0535, found 247.1. MP: Tm 59.3-59.8 (literature 48-61.0).
All data are in accordance with Zhang et al. 2014 cited above and Yilmaz et al. Chemistry Select 2019, 4 (37), 11043-11047.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-2-methoxybenzene (0.49 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 7/3 to 3/7), furnishing the title compound as a yellow solid in 56% yield.
1H NMR (600 MHz, CDCl3): δ 8.36-8.33 (m, 2H), 7.67-7.61 (m, 2H), 7.54 (app. t, J=7.7 Hz, 2H), 7.50-7.45 (m, 1H), 7.01 (app. t, J=7.5 Hz, 1H), 6.98 (app. d, J=8.4 Hz, 1H), 4.00 (s, 3H). 13C NMR (151 MHz, CDCl3): δ 178.14, 161.90, 137.19, 135.03, 133.87, 132.63, 129.78, 128.53, 120.71, 110.87, 109.48, 91.26, 90.55, 55.94. IR (neat): ν 3065.27 (C—H stretch), 2986.73 (C—H stretch), 2941.26 (C—H stretch), 2835.84 (C—H stretch), 2197.16 (C≡C stretch), 1643.22 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 237.0916, found 237.0908. MP: Tm 73.1-74.2 (literature 74-75).
All data are in accordance with Chen et al. 2009 and Iman et al. 1990 cited above.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-ethynyl-2-methylbenzene (0.47 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 8/2 to 5/5), furnishing the title compound as a brown solid in 52% yield.
1H NMR (400 MHz, CDCl3): δ 8.27-8.21 (m, 2H), 7.68-7.60 (m, 2H), 7.52 (app. t, J=7.6 Hz, 2H), 7.41-7.35 (m, 1H), 7.29 (app. d, J=7.2 Hz, 1H), 7.27-7.20 (m, 1H), 2.59 (s, 3H). 13C NMR (101 MHz, CDCl3): δ 178.08, 142.19, 137.08, 134.05, 133.68, 130.82, 129.90, 129.56, 128.65, 125.96, 120.04, 92.19, 90.78, 20.91. IR (neat): ν 3063.21 (C—H stretch), 2963.99 (C—H stretch), 2918.52 (C—H stretch), 2850.31 (C—H stretch), 2186.82 (C≡C stretch), 1634.95 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 221.0966, found 221.0968. MP: Tm 37.8-39.0.
All data are in accordance with Liu et al. 2008 cited above and Chen et al. 2009 cited above.
General procedure A for the synthesis of propynones was followed using bromobenzene (266 μL, 2.50 mmol, 1.00 equiv.) and 1-chloro-2-ethynylbenzene (0.46 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 7/3), furnishing the title compound as a dark yellow solid in 81% yield.
1H NMR (600 MHz, CDCl3): δ 8.36-8.31 (m, 2H), 7.78-7.72 (m, 1H), 7.67 (app. t, J=7.4 Hz, 1H), 7.59-7.51 (m, 3H), 7.45 (app. dd, J=7.5, 1.4 Hz, 1H), 7.38-7.33 (app. dd, J=7.5, 1.0 Hz, 1H). 13C NMR (151 MHz, CDCl3): δ 177.86, 137.55, 136.83, 135.11, 134.26, 131.79, 129.83, 129.69, 128.69, 126.87, 120.51, 91.06, 89.02. IR (neat): ν 3059.07 (C—H stretch), 2199.23 (C≡C stretch), 1632.88 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 241.0420, found 241.0417. MP: Tm 94.1-94.8 (literature 94).
All data are in accordance with Iman et al. 1990 cited above.
General procedure A for the synthesis of propynones was followed using 1-bromo-3-fluorobenzene (218 μL, 2.50 mmol, 1.00 equiv.) and 1-chloro-2-ethynylbenzene (0.68 mL, 3.75 mmol, 1.50 equiv.) at 80° C. The quantities of the other reagents and solvents were adapted accordingly. Dry, degassed dioxane was used in both chambers. The crude reaction mixture was purified via column chromatography on silica gel (petroleum ether/DCM 9/1 to 7/3), furnishing the title compound as a yellow solid in 78% yield.
1H NMR (600 MHz, CDCl3): δ 8.05 (app. d, J=7.7 Hz, 1H), 7.91 (app. d, J=9.1 Hz, 1H), 7.66 (app. d, J=8.4 Hz, 2H), 7.56-7.50 (m, 1H), 7.48 (app. d, J=8.4 Hz, 2H), 7.35 (app. td, J=8.2, 1.7 Hz, 1H), 1.37 (s, 9H). 13C NMR (151 MHz, CDCl3): δ 176.68 (d, J=2.6 Hz), 162.77 (d, J=248.2 Hz), 154.92, 139.10 (d, J=6.6 Hz), 133.11, 130.30 (d, J=7.7 Hz), 125.85, 125.41 (d, J=2.9 Hz), 121.03 (d, J=21.6 Hz), 116.72, 115.97 (d, J=22.7 Hz), 94.49, 86.48, 35.16, 31.06. IR (neat): ν 3054.94 (C—H stretch), 2957.79 (C—H stretch), 2904.05 (C—H stretch), 2866.85 (C—H stretch), 2211.63 (C≡C stretch), 1639.08 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 281.1342, found 281.1335. MP: Tm 53.7-54.7.
General procedure B for the synthesis of propynones was followed using freshly prepared 2-aminobenzaldehyde (363 mg, 3.00 mmol, 1.00 equiv.) and 5.00 equivalents of 1-chloro-3-ethynylbenzene (1.84 mL, 15.00 mmol, 5.00 equiv.) 2-aminobenzaldehyde was prepared via oxidation of 2-aminobenzyl alcohol. Therefore, a flame-dried round-bottom flask was charged with 2-aminobenzyl alcohol (370 mg, 3.00 mmol, 1.00 equiv.) and manganese dioxide (1.49 g, 21.0 mmol, 7.00 equiv.). The flask was closed with a septum, purged with N2 and dry DCM (30 mL) was added via syringe through the septum. The reaction mixture was stirred for 45 min at ambient temperature. Afterwards, the mixture was filtered over a pad of Celite® 535 and the filtrate was concentrated under reduced pressure. The crude product was used without further purification. The quantities of the other reagents and solvents were adapted accordingly. The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/EtOAc 95/5). Spectroscopic analysis of the product fraction indicated the presence of some residual aldehyde. Therefore, the mixture was dissolved in DMF (10 mL) and saturated aqueous NaHSO3 (25 mL) was added to the solution. The mixture was shaken thoroughly for half a minute. Afterwards, the mixture was diluted with water and extracted three times with a 9/1 mixture of EtOAc/hexane (25 mL). The combined organic layers were washed three times with water, dried over Na2SO4, filtered and concentrated by rotary evaporation. This extraction procedure was repeated three times. The purified product was obtained as an orange solid in 12% overall yield.
1H NMR (400 MHz, CDCl3): δ 8.13 (app. dd, J=8.3, 1.2 Hz, 1H), 7.64 (app. t, 1.6 Hz, 1H), 7.54 (app. dt, J=7.6, 1.3 Hz, 1H), 7.44 (app. ddd, J=8.1, 2.1, 1.2 Hz, 1H), 7.37-7.34 (m, 1H), 7.34-7.30 (m, 1H), 6.73 (app. t, J=7.3 Hz, 1H), 6.67 (app. dt, J=8.3, 0.5 Hz, 1H), 3.67 (br s, 2H). 13C NMR (101 MHz, CDCl3): δ 179.05, 151.18, 135.52, 134.54, 134.39, 132.42, 130.87, 130.62, 129.89, 122.34, 118.74, 116.82, 116.22, 90.21, 87.69. IR (neat): ν 3447.66 (N—H stretch), 3313.31 (N—H stretch), 3065.27 (C—H stretch), 2955.73 (C—H stretch), 2922.65 (C—H stretch), 2852.38 (C—H stretch), 2201.29 (C≡C stretch), 1616.35 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 256.0529, found 256.0529. MP: Tm 225.2-228.9 (decomposition).
General procedure B for the synthesis of propynones was followed using 2-chlorobenzaldehyde (0.11 mL) and 5.00 equivalents of 1-chloro-3-ethynylbenzene (1.84 mL, 15.00 mmol, 5.00 equiv.). The crude reaction mixture after the oxidation step was purified via flash column chromatography on silica gel (heptane/EtOAc 98/2), furnishing the title compound as an off-white solid in 98% yield.
1H NMR (400 MHz, CDCl3): δ 8.06 (app. dt, J=7.6, 0.9 Hz, 1H), 7.62 (app. t, J=1.6 Hz, 1H), 7.53 (app. dt, J=7.6, 1.1 Hz, 1H), 7.49 (app. d, J=4.1 Hz, 2H), 7.48-7.44 (m, 1H), 7.44-7.38 (m, 1H), 7.35 (app. t, J=7.8 Hz, 1H). 13C NMR (101 MHz, CDCl3): δ 176.47, 135.64, 134.66, 133.67, 133.60, 132.67, 132.55, 131.62, 131.19, 131.15, 129.96, 126.88, 121.81, 91.73, 88.71. IR (neat): ν 3061.14 (C—H stretch), 2953.66 (C—H stretch), 2920.59 (C—H stretch), 2852.38 (C—H stretch), 2203.36 (C≡C stretch), 1637.02 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 275.0030, found 275.0027. MP: Tm 70.5-71.8 (literature 77-79).
All data are in accordance with Shao et al. Synthesis 2012, 44 (12), 1798-1805.
Protocol based on Veryser et al., React. Chem. Eng. 2016, 1(2), 142-146, and Ahlburg et al. J. Org. Chem. 2013, 78(20), 10310-10318.
To the right chamber of a flame-dried two-chamber reactor (COware) were added 1-iodo-4-methoxybenzene (117 mg, 0.50 mmol, 1.00 equiv.), phenylboronic acid (73 mg, 0.75 mmol, 1.5 equiv.), potassium carbonate (207 mg, 1.50 mmol, 3.00 equiv.), and palladium(II) chloride (0.9 mg, 0.005 mmol, 1.0 mol %). The reactor was closed with two screw caps and septa and was evacuated and backfilled with argon three times. Dry, degassed toluene (2 mL) was added to the left chamber, followed by formic acid (29 μL, 0.75 mmol, 1.50 equiv.) and mesylchloride (58 μL, 0.75 mmol, 1.50 equiv.). To the right chamber, dry, degassed anisole (2 mL) was added. The reaction was initiated by the addition of triethylamine (0.21 mL, 1.50 mmol, 3.00 equiv.) to the left chamber of the reactor. Immediately after the addition of the triethylamine, the reactor was placed in an oil bath at 80° C. for 18 h. When the reaction was finished, the crude reaction mixture was filtered over a pad of Celite® 535. The filtrate was concentrated in vacuo and purified via flash column chromatography on silica gel (heptane/EtOAc 9/1), furnishing the title compound as pale yellow oil in 66% yield.
1H NMR (300 MHz, CDCl3): δ 7.86-7.79 (m, 2H), 7.78-7.63 (m, 2H), 7.60-7.53 (m, 1H), 7.51-7.43 (m, 2H), 7.00-6.94 (m, 2H), 3.89 (s, 3H). 13C NMR (75 MHz, CDCl3): δ 195.60, 163.23, 138.29, 132.58, 131.91, 130.16, 129.75, 128.20, 113.56, 55.51. IR (neat): ν 3057.01 (C—H stretch), 3003.27 (C—H stretch), 2932.99 (C—H stretch), 2839.98 C—H stretch), 1649.42 (C═O stretch). LR-MS (ESI): m/z calculated for [M+Na]+ 235.0735, found 235.2. MP: Tm 52.5-53.2 (literature 49.7-59).
All data are in accordance with Ahlburg et al. 2013 cited above, Jin et al. Synlett 2011, 2011 (10), 1435-1438, Cho et al. Catal. Commun. 2008, 9 (13), 2261-2263 and Wang et al. Synth. Commun. 2001, 31 (24), 3885-3890.
Protocol based on Bugarin et al. Tetrahedron Lett. 2015, 56 (23), 3285-3287.
To a solution of the 1-(tert-butyl)-4-ethynylbenzene (2.71 mL, 15.00 mmol, 1.00 equiv.) in dry THF (6 mL) was added n-BuLi (2.5 M in hexanes, 6.00 mL, 15.00 mmol, 1.00 equiv.) at −78° C., while stirring under argon. To this solution was added dry DMF (5.81 mL, 75.00 mmol, 5.00 equiv.) dropwise over 10 min at the same temperature. After the addition, the solution was allowed to warm up to room temperature. After 1 h of further stirring, the solution was quenched with saturated aqueous NH4Cl and extracted with DCM. The organic layers were combined, washed sequentially with water and brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure and purified via column chromatography on silica gel (heptane to heptane/Et2O 98/2), yielding 5% of the desired product as a pale yellow oil.
1H NMR (400 MHz, CDCl3): δ 9.41 (s, 1H), 7.55 (app. d, J=8.6 Hz, 2H), 7.43 (app. d, J=8.6 Hz, 2H), 1.33 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 176.96, 155.32, 133.37, 125.96, 116.45, 96.05, 88.56, 35.28, 31.14. IR (neat): ν 3052.87 (C—H stretch), 2953.66 (C—H stretch), 2922.65 (C—H stretch), 2873.05 (C—H stretch), 2854.45 (C—H stretch aldehyde), 2730.43 (C—H stretch aldehyde), 2188.89 (C≡C stretch), 1665.95 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 187.1123, found 187.1101.
All data are in accordance with Murata et al. Chem. Commun. 2020, 56 (43), 5783-5786.
Protocol based on Jeong, et al. J. Org. Chem. 2014, 79 (14), 6444-6455.
To a flame-dried two-neck round-bottom flask, equipped with a septum and a nitrogen balloon, were added 1-(tert-butyl)-4-ethynylbenzene (3.25 mL, 18.00 mmol, 1.20 equiv.) and dry THF (45 mL). The solution was cooled to −78° C., and n-BuLi (2.5 M in hexanes, 7.50 mL, 18.75 mmol, 1.25 equiv.) was added slowly via a syringe through the septum. The mixture was stirred at −78° C. for 1 h, then warmed to 0° C., stirred for 1 h, and then re-cooled to −78° C. A solution of 2-methoxybenzaldehyde (2.042 g, 15.00 mmol, 1.00 equiv.) in dry THF (15 mL) was added dropwise via a syringe and the reaction was stirred at −78° C. for 1 h, warmed to room temperature and stirred for an additional 30 min. Next, the reaction was quenched with saturated aqueous NH4Cl and extracted with Et2O. The combined organic layers were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. The residue was purified via flash column chromatography on silica gel (heptane/EtOAc 9/1), yielding 64% of the title compound as a pale, viscous liquid.
1H NMR (600 MHz, CDCl3): δ 7.65 (app. dd, J=7.5, 1.4 Hz, 1H), 7.42 (app. d, J=8.5 Hz, 2H), 7.33 (app. d, J=8.5 Hz, 2H), 7.35 (app. ddd, J=8.0, 7.5, 1.4 Hz, 1H), 7.00 (app. ddd, J=7.5, 7.5, 0.8 Hz, 1H), 6.94 (app. dd, J=8.0, 0.8 Hz, 1H), 5.93 (d, J=6.2 Hz, 1H), 3.93 (s, 3H), 3.03 (d, J=6.2 Hz, 1H), 1.31 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 157.00, 151.74, 131.60, 129.80, 129.07, 128.19, 125.34, 121.00, 119.84, 111.00, 87.77, 86.38, 61.77, 55.73, 34.86, 31.27. IR (neat): ν 3416.65 (O—H stretch), 3036.34 (C—H stretch), 2959.86 (C—H stretch), 2866.85 (C—H stretch), 2837.91 (C—H stretch), 2195.09 (C≡C stretch). HR-MS (ESI): m/z calculated for [M+Na]+ 317.1517, found 317.1515.
Protocol based on Ma et al. 2016, cited above.
To a solution of 1-(tert-butyl)-4-ethynylbenzene (1.99 mL, 11.00 mmol, 2.20 equiv.) in dry THF (35 mL), n-BuLi (2.5 M in hexanes, 4.20 mL, 10.50 mmol, 2.10 equiv.) was slowly added at −78° C. under nitrogen atmosphere. After 2 h, salicaldehyde (522 μL, 5.00 mmol, 1.00 equiv.) was added dropwise to the solution. The reaction mixture was stirred at −78° C. for another 2 h and was then quenched with saturated aqueous NH4Cl. The mixture was extracted with DCM and the organic layers were combined, washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified via flash column chromatography on silica gel (heptane/EtOAc 8/2). The title compound was obtained as burgundy red solid in 47% yield.
1H NMR (600 MHz, CDCl3): δ 7.47-7.41 (m, 3H), 7.40-7.33 (m, 4H), 7.30-7.23 (m, 2H), 6.95-6.89 (m, 2H), 5.92 (br, s, 1H), 2.77 (d, J=3.6 Hz, 1H), 1.32 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 155.50, 152.45, 131.73, 130.32, 127.92, 125.54, 124.73, 120.38, 119.02, 117.29, 88.67, 85.95, 64.64, 34.97, 31.28. IR (neat): ν 3342.24 (O—H stretch), 3034.27 (C—H stretch), 2959.86 (C—H stretch), 2901.99 (C—H stretch), 2866.85 (C—H stretch), 2195.09 (C≡C stretch). HR-MS (ESI): m/z calculated for [M+H]+ 281.1542, found 281.1496. MP: Tm 131.1-136.7 (decomposition).
Protocol based on Jeong et al. J. Org. Chem. 2014, 79 (14), 6444-6455.
To a flame-dried two-neck round-bottom flask, equipped with a septum and a nitrogen balloon, were added 1-(tert-butyl)-4-ethynylbenzene (11.28 mL, 60.00 mmol, 1.20 equiv.) and dry THF (180 mL). The solution was cooled to −78° C., and n-BuLi (2.5 M in hexanes, 25.00 mL, 62.50 mmol, 1.25 equiv.) was added slowly via a syringe through the septum. The mixture was stirred at −78° C. for 1 h, then warmed to 0° C., stirred for 1 h, and then re-cooled to −78° C. A solution of benzaldehyde (5.31 mL, 50.00 mmol, 1.00 equiv.) in dry THF (60 mL) was added dropwise via a syringe and the reaction was stirred at −78° C. for 1 h, warmed to room temperature and stirred for an additional 30 min. Next, the reaction was quenched with saturated aqueous NH4Cl and extracted with Et2O. The combined organic layers were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. The residue was purified via column chromatography on silica gel (heptane/DCM 5/5 to 2/8), yielding 65% of the title compound as a light yellow solid.
1H NMR (400 MHz, CDCl3): δ 7.65-7.59 (m, 2H), 7.44-7.37 (m, 4H), 7.34-7.31 (m, 3H), 5.65 (d, J=6.5 Hz, 1H), 2.61 (d, J=6.5 Hz, 1H), 1.29 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 151.93, 140.85, 131.58, 128.68, 128.41, 126.85, 125.38, 119.48, 88.22, 86.87, 65.15, 34.84, 31.22. IR (neat): ν 3249.23 (O—H stretch), 3036.34 (C—H stretch), 2963.99 (C—H stretch), 2904.05 (C—H stretch), 2866.85 (C—H stretch), 2197.16 (C≡C stretch). LR-MS (ESI): m/z calculated for [M+Na]+ 287.1412, found 287.2. MP: Tm 55.5-56.4.
All data are in accordance with Oshimoto et al. Org. Biomol. Chem. 2019, 17(17), 4225-4229.
Protocol based on WO2012065963.
At 0° C., EtMgBr (3.0 M in Et2O, 16.67 ml, 50.00 mmol, 5.00 equiv.) was added to a solution of 3-chloro-1-ethynylbenzene (6.16 mL, 50.0 mmol, 5.00 equiv.) in dry THF (100 mL) and stirred for 5 min at 0° C. and then for 30 min at room temperature. This solution was slowly added to a solution of 2-aminonicotinaldehyde (1.22 g, 10.0 mmol, 1.00 equiv.) in dry THF (80 mL) under a nitrogen atmosphere at room temperature. After 2 h, the mixture was quenched with saturated aqueous NH4Cl, extracted with DCM and the organic layers were washed with brine and dried over Na2SO4. The resulting organic solution was filtered and the filtrate was concentrated under reduced pressure. The residue was purified via flash column chromatography on silica gel (heptane/EtOAc 65/35 to 5/5). The title compound was obtained as a yellow solid in 20% yield.
1H NMR (400 MHz, CDCl3): δ 7.99 (app. d, J=4.6 Hz, 1H), 7.71 (app. d, J=7.4 Hz, 1H), 7.45 (app. s, 1H), 7.30-7.27 (m, 3H), 6.67 (app. dd, J=7.3, 5.2 Hz, 1H), 5.64 (s, 1H), 5.19 (br, s, 2H), 3.65 (br, s, 1H). 13C NMR (101 MHz, CDCl3): δ 157.41, 147.68, 135.37, 133.73, 131.27, 131.03, 130.55, 129.31, 124.55, 119.37, 112.72, 91.54, 83.64, 60.73. IR (neat): ν 3358.78 (N—H stretch), 3098.34 (O—H stretch), 2922.65 (C—H stretch), 2850.31 (C—H stretch), 2687.02 (C—H stretch), 2219.89 (C≡C stretch). HR-MS (ESI): m/z calculated for [M+H]+ 259.0638, found 259.0638. MP: Tm 138.8-140.1.
Protocol based on Madu et al. Tetrahedron 2017, 73 (43), 6118-6137.
Triethylsilane (319 μL, 2.00 mmol, 2.00 equiv.) was added at room temperature to a solution of (4-(tert-butyl)phenyl)-1-phenylprop-2-yn-1-ol (3.3) (264 mg, 1.00 mmol, 1.00 equiv.) in dry DCM (2 ml) under nitrogen atmosphere. Then 2,2,2-trifluoroacetic acid (297 μL, 4.00 mmol, 4.00 equiv.) was added and the solution was stirred for 20 min. The reaction mixture was quenched with saturated aqueous NaHCO3 and then extracted with DCM. The organic layers were combined, dried over MgSO4 and filtered. The filtrate was concentrated in vacuo and purified via flash column chromatography on silica gel (heptane to heptane/DCM 95/5). The desired product was obtained as a yellow oil in 10% yield.
1H NMR (400 MHz, CDCl3): δ 7.45 (m, 9H), 3.83 (s, 2H), 1.31 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 151.15, 137.07, 131.49, 128.65, 128.10, 126.72, 125.37, 120.80, 86.85, 82.86, 34.85, 31.33, 25.90. IR (neat): ν 3061.14 (C—H stretch), 2961.93 (C—H stretch), 2904.05 (C—H stretch), 2868.91 (C—H stretch), 2197.16 (C≡C stretch).
A round-bottom flask was charged with 3-(4-(tert-butyl)phenyl)-1-phenylprop-2-yn-1-one (1.16) (262 mg, 1.00 mmol, 1.00 equiv.), K2CO3 (415 mg, 3.00 mmol, 3.00 equiv.) and DMSO (10 mL) and closed with a septum. While stirring at room temperature, ethanethiol (0.74 mL, 10.00 mmol, 10.00 equiv.) was added to the closed reaction flask via a syringe through the septum. The reaction mixture was allowed to stir at room temperature for 18 h. After this time, the mixture was concentrated under reduced pressure and partitioned between DCM and a saturated aqueous solution of NaHCO3. The organic phase was collected, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure and purified via flash column chromatography on silica gel (heptane/DCM 6/4). The desired product was obtained as a bright yellow solid in 89% yield as a mixture of two stereoisomers (E/Z ratio 22/78).
1H NMR (400 MHz, CDCl3): δ 7.98 (app. d, J=7.2 Hz, 2H, Z), 7.79 (app. d, J=7.2 Hz, 2H, E), 7.55-7.22 (m, 7H, E & Z), 7.06 (s, 1H, Z), 6.66 (s, 1H, E), 2.90 (q, J=7.4 Hz, 2H, E), 2.47 (q, J=7.5 Hz, 2H, Z), 1.39 (t, J=7.4 Hz, 3H, E), 1.36 (s, 9H, Z), 1.27 (s, 9H, E), 1.10 (t, J=7.5 Hz, 3H, Z). 13C NMR (101 MHz, CDCl3): δ 189.29 (E), 188.40 (Z), 163.92 (Z), 160.21 (E), 152.15 (E), 152.10 (Z), 139.16 (E), 138.79 (Z), 136.32 (Z), 134.57 (E), 132.12 (Z), 132.00 (E), 128.48 (Z), 128.44 (E), 128.36 (E), 128.19 (E), 128.08 (Z), 127.77 (Z), 125.37 (Z), 125.04 (E), 119.57 (Z), 116.74 (E), 34.76 (Z), 34.67 (E), 31.31 (Z), 31.19 (E), 27.23 (Z), 27.04 (E), 14.20 (Z), 13.02 (E). IR (neat): ν 3065.27 (C═CH stretch), 2955.73 (C—H stretch), 2899.92 (C—H stretch), 2862.71 (C—H stretch), 1624.62 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 325.1626, found 325.1617. MP: Tm 88.4-90.3.
Protocol based on Minders et al. Bioorg. Med. Chem. Lett. 2015, 25 (22), 5270-5276.
In a round bottom flask, 1-(2-aminopyridin-3-yl)ethan-1-one (70 mg, 0.50 mmol, 1.00 equiv.) and 3-chlorobenzaldehyde (72 mg, 0.50 mmol, 1.00 equiv.) were dissolved in absolute ethanol (1.4 mL). Subsequently, aqueous NaOH (40% in H2O, 0.03 mL, 0.25 mmol, 0.50 equiv.) was added dropwise to the solution. After the addition, the mixture was stirred at room temperature for 2 h. Afterwards, the crude reaction mixture was extracted with EtOAc and washed with water. The organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. Crystallization from MeOH, followed by flash column chromatography on silica gel (heptane/EtOAc 6/4), furnished the title compound as a yellow solid in 57% yield.
1H NMR (400 MHz, CDCl3): δ 8.27 (app. d, J=4.4 Hz, 1H), 8.16 (app. d, J=7.8 Hz, 1H), 7.63 (ABq, Δν=55.3 Hz, J=15.6 Hz, 2H), 7.62 (app. s, 1H), 7.52-7.46 (m, 1H), 7.43-7.32 (m, 2H), 7.01 (br, s, 2H), 6.70 (app. dd, J=8.1, 4.8 Hz, 1H). 13C NMR (101 MHz, CDCl3): δ 189.83, 159.62, 154.31, 142.19, 139.47, 136.76, 135.02, 130.28, 130.23, 127.79, 126.77, 122.88, 113.67, 112.36. IR (neat): ν 3381.52 (N—H stretch), 3265.77 (N—H stretch), 3199.63 (C—H stretch), 3150.02 (C—H stretch), 3059.07 (C—H stretch), 2924.72 (C—H stretch), 1649.42 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 259.0638, found 259.0641. MP: Tm 122.4-123.7.
Protocol based on CN106279015.
To a solution of 2-aminonicotinic acid (414 mg, 3.00 mmol, 1.00 equiv.) in chlorobenzene (9 mL) was added dropwise thionyl chloride (1.09 mL, 15.00 mmol, 5.00 equiv.), while stirring at room temperature. After the addition was complete, the flask was heated to 55° C. for 1 h while stirring. After this period, the excess thionyl chloride was distilled off under reduced pressure. Subsequently, 1-(3-chlorophenyl)piperazine (1.48 mL, 9.00 mmol, 3.00 equiv.) was added to the flask and the solution was heated to 80° C. The reaction was stirred for 30 min at this temperature. After this period, the reaction was quenched with saturated aqueous NaHCO3 and extracted with DCM. The organic layers were combined and washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The residue was purified via column chromatography on silica gel (DCM/i-PrOH 95/5), yielding 22% of the title compound as an off-white solid.
1H NMR (600 MHz, CDCl3): δ 8.15 (app. dd, J=5.0, 1.8 Hz, 1H), 7.40 (app. dd, J=7.4, 1.8 Hz, 1H), 7.19 (app. t, J=8.0 Hz, 1H), 6.90-6.87 (m, 1H), 6.87-6.86 (m, 1H), 6.79 (app. ddd, J=8.4, 2.3, 0.7 Hz, 1H), 6.68 (app. dd, J=7.4, 5.0 Hz, 1H), 5.19 (br, s, 2H), 3.77 (br, s, 4H), 3.22 (br, t, J=5.0 Hz, 4H). 13C NMR (151 MHz, CDCl3): δ 202.28, 168.84, 157.13, 154.26, 151.84, 150.18, 136.41, 135.11, 130.20, 120.39, 116.53, 114.56, 113.02, 49.36 (br). IR (neat): ν 3470.39 (N—H stretch), 3119.01 (C—H stretch), 2920.59 (C—H stretch), 2839.98 (C—H stretch), 1626.68 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 317.1169, found 317.1159. MP: Tm 149.7-150.6.
Protocol based on Akerbladh et al. J. Org. Chem. 2015, 80 (3), 1464-1471.
To a 0.5-2 mL microwave vial were added 2-iodoaniline (110 mg, 0.50 mmol, 1.00 equiv.), ethynylbenzene (110 μL, 1.00 mmol, 2.00 equiv.), tris(dibenzylideneacetone)dipalladium(0) (9.2 mg, 0.01 mmol, 2.0 mol %), 1,1′-bis(diphenylphosphino)ferrocene (11.2 mg, 0.02 mmol, 4.0 mol %),
molybdenum hexacarbonyl (132 mg, 0.50 mmol, 1.00 equiv), cesium carbonate (490 mg, 1.50 mmol, 3.00 equiv), and 1.5 mL of diethylamine. The vial was purged with nitrogen, capped with a snap-on cap, and irradiated in a microwave reactor at 120° C. for 20 min while stirring. Afterwards, the reaction mixture was diluted with water and extracted with DCM (3×15 mL). The organic layers were combined, dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified via flash column chromatography on silica gel (DCM/MeOH 99/1 to 95/5), furnishing the title compound as a light brown solid in 38% yield.
1H NMR (400 MHz, DMSO-d6): δ 11.70 (br, s, 1H), 8.10 (app. d, J=8.3 Hz, 1H), 7.87-7.80 (m, 2H), 7.80-7.74 (m, 1H), 7.70-7.63 (m, 1H), 7.63-7.56 (m, 3H), 7.37-7.31 (m, 1H), 6.33 (app. s, 1H). 13C NMR (101 MHz, DMSO-d6): δ 176.93, 149.97, 140.52, 134.20, 131.73, 130.39, 128.95, 128.42, 127.39, 124.68, 123.21, 118.72, 107.31. IR (neat): ν 3061.14 (C═CH stretch), 2961.93 (C—H stretch), 2922.65 (C—H stretch), 1632.88 (C═O stretch), 1502.67 (C═C stretch). LR-MS (ESI): m/z calculated for [M+H]+ 222.0919, found 222.2. MP: Tm 210.2-212.1 (literature 252-254).
All data are in accordance with Huang et al. Org. Lett. 2008, 10 (12), 2609-2612, Kuo et al. J. Med. Chem. 1993, 36 (9), 1146-1156 and Lee & Youn Bull. Korean Chem. Soc. 2008, 29 (9), 1853-1856.
Protocol based on Akerbladh et al. J. Org. Chem. 2015, 80(3), 1464-1471.
To a 0.5-2 mL microwave vial were added 1-(2-aminophenyl)-3-(4-(tert-butyl)phenyl)prop-2-yn-1-one (1.14) (35.2 mg, 0.13 mmol, 1.00 equiv.) and Et2NH (1.5 mL). The vial was purged with nitrogen, capped with a snap-on cap, and irradiated in a microwave reactor at 120° C. for 20 min while stirring. Afterwards, the reaction mixture was concentrated under reduced pressure. The residue was purified via flash column chromatography on silica gel (DCM to DCM/MeOH 9/1), furnishing the desired product as a white solid in 84% yield.
1H NMR (400 MHz, DMSO-d6): δ 11.67 (br, s, 1H), 8.10 (app. ddd, J=8.0, 1.5, 0.4 Hz, 1H), 7.78 (app. d, J=8.4 Hz, 2H), 7.75 (app. d, J=8.0 Hz, 1H), 7.66 (app. ddd, J=8.3, 6.9, 1.5 Hz, 1H), 7.61 (ap. d, J=8.5 Hz, 2H), 7.33 (app. ddd, J=7.2, 6.8, 0.8 Hz, 1H), 6.34 (s, 1H), 1.35 (s, 9H). 13C NMR (151 MHz, DMSO-d6): δ 176.87, 153.21, 149.96, 140.54, 131.68, 131.46, 127.16, 125.77, 124.85, 124.68, 123.15, 118.68, 106.93, 34.59, 30.93. IR (neat): ν 3034.27 (C═CH stretch), 2954.39 (C—H stretch), 2899.92 (C—H stretch), 2866.85 (C—H stretch), 2771.77 (C—H stretch), 1632.88 (C═O stretch), 1496.47 (C═C stretch). HR-MS (ESI): m/z calculated for [M+H]+ 278.1545, found 278.1541. MP: Tm 322.2-322.8.
All data are in accordance with Wang et al. Synthesis 2017, 49 (18), 4309-4320 and Xu et al. Org. Lett. 2018, 20 (7), 1893-1897.
Protocol based on Neumann et al 2014, Veryser et al. 2016 and Akerbladh et al. 2015, cited above.
To the right chamber of a flame-dried two-chamber reactor (COWare) as added 3-bromopyridin-2-amine (86 mg, 0.50 mmol, 1.00 equiv.), palladium(II) chloride (4.4 mg, 0.025 mmol, 5.0 mol %) and Xantphos (14.5 mg, 0.025 mmol, 5.0 mol %). The reactor was closed with two screw caps and septa and was evacuated and backfilled with argon three times. Dry, degassed toluene (3 mL) was added to the left chamber, followed by formic acid (29 μL, 0.75 mmol, 1.50 equiv.) and mesylchloride (58 μL, 0.75 mmol, 1.50 equiv.). To the right chamber, dry, degassed dioxane (3 mL) was added, followed by 1-(tert-butyl)-4-ethynylbenzene (180 μL, 1.00 mmol, 2.00 equiv.), dry triethylamine (0.21 mL, 1.50 mmol, 3.00 equiv.) and dry diethylamine (0.16 mL, 1.50 mmol, 3.00 equiv.). The reaction was initiated by the addition of triethylamine (0.21 mL, 1.50 mmol, 3.00 equiv.) to the left chamber of the reactor. Immediately after the addition of the triethylamine, the reactor was placed in an oil bath at 100° C. for 18 h. When the reaction was finished, the crude reaction mixture was filtered over a pad of Celite® 535. The filtrate was concentrated in vacuo and purified via flash column chromatography on silica gel (DCM/MeOH 95/5 to 9/1), yielding 54% of the desired product as a beige solid.
1H NMR (400 MHz, CDCl3): δ 10.85 (br, 1H), 8.68 (app. dd, J=7.9, 1.9 Hz, 1H), 8.38 (app. dd, J=4.6, 1.9 Hz, 1H), 7.75-7.61 (m, 2H), 7.61-7.53 (m, 2H), 7.28 (app. dd, J=7.9, 4.6 Hz, 1H), 6.61 (s, 1H), 1.39 (s, 9H). 13C NMR (101 MHz, CDCl3): δ 178.89, 154.65, 152.37, 151.54, 151.13, 136.25, 131.52, 127.13, 126.34, 120.36, 119.60, 109.58, 35.00, 31.22. IR (neat): ν 3036.34 (C═CH stretch), 2945.39 (C—H stretch), 2868.91 (C—H stretch), 1610.15 (C═O stretch). HR-MS (ESI): m/z calculated for [M+H]+ 279.1497, found 279.1494. MP: Tm 237.8-239.8.
Protocol based on Hortmann & Harris J. Am. Chem. Soc. 1971, 93 (10), 2471-2481, Corey & Chaykovsky J. Am. Chem. Soc. 1965, 87 (6), 1353-1364 and Hortmann J. Am. Chem. Soc. 1965, 87 (21), 4972-4973.
To a solution of trimethylsulfoxonium iodide (280 mg, 1.27 mmol, 3.18 equiv.) in tert-butanol (3 mL) was added potassium tert-butoxide (145 mg, 1.29 mmol, 3.23 equiv.), while stirring at 50° C. When the addition was complete, the mixture was stirred at the same temperature for another 15 min before a solution of 3-(4-(tert-butyl)phenyl)-1-phenylprop-2-yn-1-one (1.16) (105 mg, 0.40 mmol, 1.00 equiv.) in tert-butanol (2 mL) was added. The resultant mixture was stirred at 50° C. for 18 h. After this period, the reaction solvent was removed under reduced pressure. The residue was partitioned between DCM and water and the aqueous phase was extracted two more times with DCM. The combined organic layers were washed with brine and dried over Na2SO4. The filtrate was concentrated under reduced pressure and the residue was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2 to 6/4). The title compound was obtained as a dark yellow solid in 79% yield.
1H NMR (400 MHz, CDCl3): δ 7.60 (app. d, J=7.0 Hz, 2H), 7.54 (app. d, J=8.3 Hz, 2H), 7.49-7.34 (m, 5H), 6.26 (app. s, 1H), 5.81 (ABq, Δν=7.8 Hz, J=4.2 Hz, 2H), 3.63 (s, 3H), 1.36 (s, 9H). 13C NMR (151 MHz, CDCl3): δ 151.73, 145.97, 141.01, 137.97, 128.68, 128.48, 127.46, 127.14, 125.65, 102.01, 83.61, 83.38, 50.22, 34.66, 31.34. IR (neat): ν 3054.94 (C—H stretch), 2957.79 (C—H stretch), 2924.72 (C—H stretch), 2866.85 (C—H stretch), 1696.96 (C═S═O stretch, presumed). HR-MS (ESI): m/z calculated for [M+H]+ 337.1626, found 337.1613. MP: Tm 93.2-94.4.
Protocol based on Yadav et al., Chem. Eur. J. 2014, 20 (23), 7122-7127.
A flame-dried pressure tube was charged with 3-iodo-1H-pyrazolo[3-4-b]pyridine (250 mg, 1.00 mmol, 1.00 equiv.), copper(I) iodide (8 mg, 0.04 mmol, 4.0 mol %) and bis(triphenylphosphine)palladium(II) dichloride (28 mg, 0.04 mmol, 4.0 mol %). The tube was sealed with a screw cap and septum and evacuated and backfilled with N2 three times. Next, dry triethylamine (4.4 mL) and 1-ethynyl-3-chlorobenzene (148 μL, 1.20 mmol, 1.20 equiv.) were added via a syringe through the septum. The reaction mixture was stirred at 90° C. for 18 h. Afterwards, the mixture was filtered over a pad of Celite® 535 and the filtrate was concentrated by rotary evaporation. The residue was purified via column chromatography on silica gel (heptane/EtOAc 8/2 to 6/4), followed by recrystallization from n-hexane at −78° C. The title compound was obtained as an off-white solid in 42% yield.
1H NMR (600 MHz, CDCl3): δ 12.80 (br, s, 1H), 8.70 (app. d, J=4.0 Hz, 1H), 8.28 (app. d, J=7.7 Hz, 1H), 7.64 (app. s, 1H), 7.53 (app. d, J=7.6 Hz, 1H), 7.40-7.36 (m, 1H), 7.35-7.33 (m, 1H), 7.33-7.29 (m, 1H). 13C NMR (101 MHz, DMSO-d6): δ 151.93, 150.36, 133.90, 131.48, 131.19, 130.69, 129.84, 129.82, 127.36, 124.09, 118.47, 116.46, 91.40, 82.79. IR (neat): ν 3133.48 (C—H stretch), 3085.94 (C—H stretch), 2875.12 (C—H stretch), 2823.44 (C—H stretch), 2761.43 (C—H stretch), 2221.96 (C≡C stretch). HR-MS (ESI): m/z calculated for [M+H]+ 254.0485, found 254.0482. MP: Tm 210.1-211.0.
Protocol based on WO2012151158.
To a solution of 3-((3-chlorophenyl)ethynyl)-1H-pyrazolo[3,4-b]pyridine (10.1) (146 mg, 0.58 mmol, 1.00 equiv.) in dry DMF (11.5 mL) was added potassium carbonate (159 mg, 1.16 mmol, 2.00 equiv.) and methyliodide (39 μL, 0.63 mmol, 1.10 equiv.) at 0° C. The reaction mixture was slowly warmed to room temperature and stirred for an additional 3 h. Afterwards, the reaction mixture was diluted with water (50 mL) and extracted three times with EtOAc (50 mL). The combined organic layers were washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified via crystallization from MeOH. The desired product was obtained as an off-white solid in 98% yield.
1H NMR (400 MHz, DMSO-d6): δ 8.67 (app. dd, J=4.5, 1.5 Hz, 1H), 8.43 (app. dd, J=8.1, 1.5 Hz, 1H), 7.57-7.48 (m, 2H), 7.38 (app. dd, J=8.1, 4.5 Hz, 1H), 4.13 (s, 3H). 13C NMR (101 MHz, DMSO-d6): δ 150.36, 150.12, 133.90, 131.43, 131.21, 130.67, 130.23, 129.86, 125.76, 124.02, 118.67, 117.02, 91.81, 82.37, 34.65. IR (neat): ν 2932.99 (C—H stretch), 2215.76 (C≡C stretch). HR-MS (ESI): m/z calculated for [M+H]+ 268.0641, found 268.0634. MP: Tm 136.0-136.3.
Protocol based on Yadav et al. Chem. Eur. J. 2014, 20(23), 7122-7127.
A flame-dried pressure tube was charged with 3-iodo-1H-indazole (244 mg, 1.00 mmol, 1.00 equiv.), copper(I) iodide (8 mg, 0.04 mmol, 4.0 mol %) and bis(triphenylphosphine)palladium(II) dichloride (28 mg, 0.04 mmol, 4.0 mol %). The tube was sealed with a screw cap and septum and evacuated and backfilled with N2 three times. Next, dry triethylamine (4.4 mL) and 1-ethynyl-3-chlorobenzene (148 μL, 1.20 mmol, 1.20 equiv.) were added via a syringe through the septum. The reaction mixture was stirred at 90° C. for 18 h. Afterwards, the mixture was filtered over a pad of Celite® 535 and the filtrate was concentrated by rotary evaporation. The residue was purified via flash column chromatography on silica gel (heptane/EtOAc 8/2), followed by recrystallization from n-heptane. The title compound was obtained as a white solid in 21% yield.
1H NMR (400 MHz, DMSO-d6): δ 13.58 (s, 1H), 7.90 (app. d, J=8.1 Hz, 1H), 7.78 (app. s, 1H), 7.67-7.60 (m, 2H), 7.57-7.42 (m, 3H), 7.27 (app. t, J=7.5 Hz). 13C NMR (101 MHz, DMSO-d6): δ 140.00, 133.41, 130.83, 130.68, 130.12, 129.09, 127.29, 126.96, 124.19, 123.97, 121.69, 119.68, 110.88, 90.83, 83.03. IR (neat): ν 3154.15 (C—H stretch), 3125.22 (C—H stretch), 2910.25 (C—H stretch), 2221.96 (C≡C stretch), 1593.61 (C═C stretch). HR-MS (ESI): m/z calculated for [M+H]+ 253.0533, found 253.0519. MP: Tm 198.1-198.8.
Male NMRI mice (average body weight 30 g) were maintained as described in example 3. For each time period (i.e., 2 min, 15 min, 30 min, 1 h, 2-2.5 h, 4 h, 8 h, and 24 h), 1-5 mice were i.p. injected with 200 μL (injection volume was adjusted to the individual weight) of VHC (8% solutol/12% PEG200/80% water) or 300 mg/kg test compound dissolved in VHC. After the treatment period, blood samples were drawn from the tail veins, collected in Greiner MiniCollect K2EDTA tubes, and centrifuged twice for 5 min at 15,000 g to obtain plasma samples. Three volumes of acetonitrile were added to one volume of plasma to precipitate out the proteins. The samples were vortexed for 20 s and placed on ice. Immediately after, they were centrifuged at 5,000 g for 10 min and again at 10,000 g for 2 min. The resulting supernatants were transferred to Eppendorf tubes and centrifuged for 2 min at 10,000 g. Finally, the supernatants were collected for analysis by LC-MS/MS to determine the targeted compound concentration. Percentage recovery was determined as follows: known concentrations of the compound were spiked into the blank plasma and acetonitrile was added (3:1 ratio) to precipitate out the proteins. Samples were vortexed for 20 s and placed on ice. The resulting supernatants were isolated via centrifugation as described above. The targeted compounds were identified using LC-MS/MS to detect their characteristic ions. Plasma concentrations at each point were plotted as a function of time.
Brain samples were collected at the same time points as described for blood samples. Male NMRI mice were overdosed with dolethal via i.p. administration. Mice were perfused with 0.9% saline and brains were collected in Eppendorf tubes. Brains were weighed, whereafter 400 μL of acetonitrile was added to the samples. Brains were homogenized with a pellet mixer (VWR, EU article no. 431-0100) and immediately placed on ice. Homogenized samples were centrifuged at 5,000 g for 10 min and again at 10,000 g for 2 min. The resulting supernatants were transferred to Eppendorf tubes and centrifuged for 2 min at 10,000 g. Finally, the supernatants were collected for analysis by LC-MS/MS to determine the targeted compound concentration in the brain tissue.
Pharmacokinetic analysis of compounds 3.3 and 10.1 was performed at 2 min, 15 min, 30 min, 1 h, 2-2.5 h, 4 h, 8 h, and 24 h after i.p. administration in mice at a dose of 300 mg/kg (
| Number | Date | Country | Kind |
|---|---|---|---|
| 21171769.9 | May 2021 | EP | regional |
This application is a national-stage application under 35 U.S.C. § 371 of International Application No. PCT/EP2022/061860, filed May 3, 2022, which International Application claims benefit of priority to European Patent Application No. 21171769.9, filed May 3, 2021.
| Filing Document | Filing Date | Country | Kind |
|---|---|---|---|
| PCT/EP2022/061860 | 5/3/2022 | WO |