Next Article in Journal
Coordination Complexes Built from a Ditopic Triazole-Pyrazole Ligand with Antibacterial and Antifungal Performances
Next Article in Special Issue
Biological Activity of Hexaazaisowurtzitane Derivatives
Previous Article in Journal
Experimental and Computational Study on Inhibitory Effect and Adsorption Properties of N-Acetylcysteine Amino Acid in Acid Environment
Previous Article in Special Issue
Structural Optimization of BIPPO Analogs as Potent Antimalarials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Lateral Metalation of Isoxazolo[3,4-d]pyridazinones towards Hit-to-Lead Development of Selective Positive Modulators of Metabotropic Glutamate Receptors

1
Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
2
Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(19), 6800; https://doi.org/10.3390/molecules28196800
Submission received: 14 July 2023 / Revised: 22 September 2023 / Accepted: 22 September 2023 / Published: 25 September 2023

Abstract

:
Isoxazolo[3,4-d] pyridazinones ([3,4-d]s) were previously shown to have selective positive modulation at the metabotropic glutamate receptor (mGluR) Subtypes 2 and 4, with no functional cross-reactivity at mGluR1a, mGluR5, or mGluR8. Additional analogs were prepared to access more of the allosteric pocket and achieve higher binding affinity, as suggested by homology modeling. Two different sets of analogs were generated. One uses the fully formed [3,4-d] with an N6-aryl with and without halogens. These underwent successful selective lateral metalation and electrophilic quenching (LM&EQ) at the C3 of the isoxazole. In a second set of analogs, a phenyl group was introduced at the C4 position of the [3,4-d] ring via a condensation of 4-phenylacetyl-3-ethoxcarbonyl-5-methyl isoxazole with the corresponding hydrazine to generate the 3,4-ds 2b and 2j to 2n.

1. Introduction

The mGluRs are members in good standing of class C of G-protein-coupled receptors (GPCR). The mGluRs consist of a venus flytrap domain (VFD), which contains the orthosteric glutamate binding site, and the seven transmembrane (7TM) domain, which contains the allosteric site. The mGluRs are located on both post- and pre-synaptic neurons and are involved with signal regulation. Compounds that target mGluRs are important for the treatment of a variety of central nervous system (CNS) disorders, as well as cancer [1,2,3,4,5]. The class is further broken down into three subgroups by sequence homology. Subgroup 1 has the excitatory receptors mGluR1 and mGluR5. Subgroup 2 (mGluR2–3) and Subgroup 3 (mGluR4,6–8) are inhibitory [6]. Each has a unique potential therapeutic application associated with it, and therefore ligands with sub-type selectivity are of paramount importance. In particular, mGluR2 is a target for treatment of anxiety and schizophrenia, while activation of mGluR4 has been postulated to ease the symptoms of Parkinson’s disease and may even slow the progress of the disease [1,2,7,8]. Although there is high sequence homology between the subgroups, the allosteric sites are less conserved and present the most logical opportunity to develop novel small molecules to selectively target them. Using synthetic methods largely developed by Renzi and Dal Piaz [9,10], we reported previously initial hits with [3,4-d]s, which exhibited selective activity at mGluR2 and mGluR4 [11,12]. We next desired to explore the hit-to-lead development of this template to further elaborate this series with liphophilic groups and test the practicality of increasing efficacy and selectivity.

2. The Working Hypothesis, Application of Structure-Based Drug Design

Recent crystallographic advances in GPCR structures have allowed for increasing confidence in homology modeling for hypothesis-driven, structure-based drug design.
The allosteric binding site crystal coordinates were used for mGluR1, PDB accession number 4OR2 [13]. Ten homology models were compared using the Discovery Studio protocol, and the highest score for goodness of fit was used for the homology comparison. The calculations were performed using Autodock Vina for mGluR2 and Discovery Studio for mGluR4 [12].
For Discovery Studio, the protein structures were typed with the CHARMm forcefield [14], and energy was minimized with the smart minimizer protocol within Discovery Studio [15] using the Generalized-Born with simple switching implicit solvent model to a root mean square gradient (RMS) convergence of <0.001 kcal/mol prior to use in the docking studies. Docking was performed using the flexible docking protocol [16], which allows for conformational degrees of freedom in both the ligand and the binding site residues. The amino acid residues within the allosteric site were allowed to attain an optimum conformation using flexible docking. For example, the essential unique amino acid binding residues in mGluR4 were selected based on Feng’s model of allosteric activation [17] and with numbering of the relative position in the GPCR 7TM based on the protocol established by Isberg [18]: Leu6593.36, Met6633.40, Leu 7535.40, Leu7565.43, Leu7575.44, Thr7946.46, Trp7986.50, Phe8016.53, Phe8056.57, Leu 8227.32, and Val8267.36.

2.1. Computational Prediction Based on the Working Hypothesis for mGluR2

Our initial studies indicated that experimentally, the selectivity centered on one particular variable for mGluR2, that is, each of the N-Aryl groups contained fluorine substituents (Figure 1) [12]. On closer examination of an expanded ligand set, the most reasonable interactions for the unique non-conserved residues associated with mGluR2 were the sulfur-containing residues at the hinge between the Venus flytrap domain and the transmembrane domain. The observation that the program assigned the interaction of the fluorine-containing aryl group with Met728 as a pi-sulfur interaction was especially suggestive. The far intracellular end of the allosteric pocket is formed by Phe776 and Trp 773, which allow just sufficient space to accommodate the geminal di-benzyl substitution.

2.2. Computational Prediction Based on the Working Hypothesis for mGLuR4

The computational prediction that the unique residue for mGluR4, Leu756, is involved directly in binding is encouraging (Figure 2). The fact that the calculation predicts a reasonably strong pi-stacked amide interaction would bode well for subsequent selectivity and affinity. The geminal benzyl groups of 4f both add important interactions at the VFD and transmembrane domains, with a predicted strong pi-cation interaction with Arg655.
A series of analogs containing a 4-phenyl group were prepared that corresponded to our previous hits at mGluR2 and mGluR4 [12] from 2j to 2n. These contain a rigidly connected lipophilic group and represent a control concerning the role of conformational flexibility at the allosteric site. Compared to our initial analogs in this series [12], the di-substituted analogs had superior predicted CDocker scores to both the mono- and un-substituted analogs. The more rigid 4-phenyl analogs generally showed slightly lower binding energies compared to their 4-methyl analogs.
Caution against confirmation bias should always be accorded to predictions with theoretical calculations, and testing by experiment is always warranted [19,20]. The computational predictions described above, however, (1) suggest a plausible basis for selectivity as the unique sub-type residues are predicted to be involved in the drug receptor interaction and (2) allow for testable hypotheses. Our next task was to prepare the small molecules to test each of these hypotheses.

3. Results and Discussion

The synthesis of selectively substituted examples of the [3,4-d] scaffold was accomplished in the present study using lateral metalation and electrophilic quenching with non-nucleophilic bases, as summarized in Scheme 1 and Table 1 [21,22,23,24,25,26,27].
The Dal Piaz group has pioneered the use of isoxazolyl[3,4-d] analogs as a useful scaffold [9,10], both for their intrinsic biological activity [29,30,31,32] and their utility as precursors for medicinally relevant exploration [33,34,35,36,37,38,39,40]. Our rationale for exploring lateral metalation with non-nucleophilic bases lies in previous studies by the Dal Piaz group, in which they reported that upon use of nucleophilic bases for deprotonation, they observed three different routes of rearrangement: isolated compounds 10, 14, and 15 (Scheme 2) [41,42,43], all predicated on the Kemp elimination [44,45,46,47]. In our hands, we found that extended reflux time evidenced one of these rearrangements, which requires a Kemp elimination, and subsequent rearrangement arising from attack of the nucleophilic base at the pyridazinone carbonyl, loss of carbon dioxide to 12, followed by protonation and tautomerization to 13. In our study, we observed the 3-amino-pyrazole 15, but not its 4-cyano-analog 14 (Scheme 2), most probably since we used thermodynamic conditions, whereas the other two documented rearrangements could be kinetic processes.
We therefore turned to lateral metalation and electrophilic quenching (LM&EQ) using non-nucleophilic bases. Originally reported by Micetich [21], we have expanded the application of LM&EQ to functionally complex isoxazoles for a variety of target types [22,23,24,25,26,27].
Method A involved direct metalation of the [3,4-d] scaffold, conducted in THF, at low temperatures as listed in Table 1. Method A generally accords with a mixture of mono-3 and di-substitution 4. These were usually readily separable by chromatography, and the di-substitution product has been confirmed by single crystal X-ray diffractometry of 4a [11]. In contrast with our previous experience with lateral metalation, [3,4-d] proceeds very quickly through mono-electrophile incorporation products 3, and under almost all conditions studied, it produced a predominant double substitution 4. Di-substitution can be optimized by using excess bases and electrophiles and is the major product found when the 6-N-aryl group is electron withdrawing, especially in the case of the N-6-3,5-difluorormethyl phenyl, 4f.
We had previously reported the LM&EQ of isoxazolyl acetals 5, for which mono-electrophile incorporation was facile, but subsequent di-substitution was found to be very difficult [26,27]. This was found to be the method of choice if predominant mono-substitution was desired. In the case of larger electrophiles (i.e., 1-bromo-methyl-naphthylene 6g), subsequent hydrazine reactions stopped at the open hydrazone 7g, and only extended reflux produced the desired closed [3,4-d] pyridazinone 3g. This protection first, mono-LM&EQ, deprotection sequence represents Method B. In the 4-phenyl series [3,4-d]s 2b and 2j to 2n, it was found that Hunig’s base catalysis served to complete the ring closure efficiently. The characterization of compounds in the 4-phenyl series 2j to 2n is given after Table S1 in the Supplementary Materials; compound 2b is previously known [10].
We found that the 3,5-diCl-phenyl example proceeded to produce di-substitution 4f as the major product by method A. We anticipated that metalation between the 3,5-dichloro groups was a distinct alternative pathway (Scheme 3), which could result in a benzyne intermediate and subsequent cine-substitution [48,49,50,51], followed by addition of the conjugate acid to 18 or 19, and also potential electrophile incorporation to 20 and 21. Therefore, we carefully examined the reaction mixture by HPLC-MS. We did not find evidence of expected products of benzyne intermediates, and the low yield was found to be oligomeric or polymeric baseline material.
The addition of lipophilic groups for compounds 3 and 4 was designed to enhance affinity; however, binding is not pharmacology. The conformational flexibility of the lipophilic additions is intended to enhance lipophilic surface area while simultaneously allowing for the swing of TM6 essential to GPCR activation [3,4,5]. In contrast, series 2b and 2j to 2n contain additional lipophilic character, but in contrast, they are rigid, which would be plausibly expected to alter the pharmacology to inhibit the receptor due to the strut-like lipophilic group acting to prevent the essential movement of the receptor.

4. Materials and Methods

Tetrahydrofuran (THF) was dried over activated sieves, then distilled under argon from sodium and benzophenone. Argon gas was passed through tubes with the indicator Drierite for reactions that required an inert atmosphere. NMR spectra were recorded at 400 MHz, unless otherwise specified, in CDCl3 solution, and chemical shifts are reported in ppm. The mass spectra were obtained using chemical ionization unless otherwise noted and are reported as m/z (relative intensity). Starting materials for the lateral metalation were prepared via Dal Piaz’s method for 4-phenyl and 4-methyl 3,4-ds [9,10].
All steps were performed in an inert atmosphere, unless otherwise noted. To the pre-dried round bottom cooled under argon, 3,4-d was added. After which, dry THF was added in sufficient amounts to reach a concentration of 50 mM. The reaction was then placed and stirred in a cooling bath at the desired reaction temperature or based on solubility for 5 min. Then 1 or 2 eq of the amine base was added via syringe dropwise over 5 min. The reaction mixture was then allowed to react for 30 min, during which time the solution is usually observed to darken. Furthermore, 1 or 2 eq of a cooled 1.7M solution of the electrophile in dry THF (kept at 0 °C or −78 °C) was slowly added dropwise. The solution was allowed to reach room temperature, and a saturated ammonium chloride solution was added at about −20 °C until warming to room temperature. The solvent was evaporated under reduced pressure using a rotary evaporator. The aqueous phase was extracted with dichloromethane (DCM). Moreover, the organic phase was washed with water and brine and finally dried over sodium sulfate overnight. After filtration of the solid, the solution was concentrated using a rotary evaporator and purified by PTLC or column with a mixture of hexanes, ethylacetate, and DCM in a ratio of 6:1:1. The characterization of compounds in the 4-phenyl series 2j to 2n is given after Table S1 in the Supplementary Materials; compound 2b is previously known [10].
  • 4-Methyl-3-phenethyl-6-phenylisoxazolo[3,4-d]pyridazin-7(6H)-one, 3a. 1H NMR (500 MHz, CDCl3): δ 7.57 (d, 1H); 7.46 (t, 2H); 7.32 (t, 1H); 7.30 (t, 2H), 7.28 (t, 1H); 7.12 (d, 2H); 3.47 (t, J = 7.5 Hz, 2H); 3.18 (t, J = 7.5 Hz, 2H), 2.29 (s, 3H). 13C NMR: 172.6, 152.9, 152.4, 140.8, 140.0, 138.9, 128.9, 128.9, 128.4, 127.9, 127.1, 125.8, 112.5, 34.3, 29.6, 19.4. C20H17N3O2 MW 331.13; ESI-MS m/z 332.0971 [(M + H)+ 90% rel. I.]. HRMS calc’d for C20H18N3O2 (M + H+): 332.1399, found: 332.1396. −0.9 ppm.
  • 3-(1,3-Diphenylpropan-2-yl)-4-methyl-6-phenylisoxazolo[3,4-d]pyridazin-7(6H)-one, 4a. 1H NMR (500 MHz, CDCl3): δ 7.51 (d, 2H); 7.43 (t, 2H); 7.36 (t, 1H); 7.21 (m, 8H); 7.01 (d, 2H); 3.796 (pentet, J = 7.5 Hz, 1H); 3.268 (d, J = 8 Hz, 4H); 1.86 (s, 3H). 13C NMR: 174.5, 152.8, 151.9, 140.6, 139.8, 138.0, 128.8, 128.5, 127.8, 127.1, 125.6, 113.4, 45.0, 40.8, 18.9. C27H23N3O2 MW 421.18; ESI-MS m/z 422.1432 [(M + H+), 61% rel. I.]. HRMS calc’d for C27H24N3O2 (M + H+): 422.1869, found: 422.1871. 0.5 ppm.
  • 3-(2-(4-Chlorophenyl)-2-hydroxyethyl)-4-methyl-6-(p-tolyl)isoxazolo[3,4-d]pyridazin-7(6H)-one, 3h. TLC (SiO2 4:4:1 hexane-EtOAc-DCM) Rf 0.16. 1H NMR (500 MHz, CDCl3): δ 7.42 (d, J = 8.5 Hz, 2H); 7.259 (m, 4H); 7.08 (d, J = 8.5 Hz, 2H); 5.1 (br. m., 1H); 4.04 (dd, J = 7 Hz); 3.69 (dd, J = 15, 7 Hz, 1H); 3.62 (dd, J = 7, 15 Hz); 2.40 (s, 3H); 2.386 (s, 3H). C21H18ClN3O3 MW 395.1; ESI-MS m/z 378.0079 [(35Cl, M-OH+) 47% rel. I.], 379.9991 [(37Cl, M-OH+) 15% rel. I.].
  • 4-Methyl-3-phenethyl-6-(p-tolyl)isoxazolo[3,4-d]pyridazin-7(6H)-one, 3c. 1H NMR (400 MHz, CDCl3): δ 7.29 (d, J = 8 Hz, 2H); 7.12 (m, 5H); 6.97 (d, J = 8 Hz, 2H); 3.32 (t, J = 8 Hz, 2H); 3.03 (t, J = 8 Hz, 2H); 2.24 (s, 3H); 2.14 (s, 3H). 13C NMR: 172.4, 152.9, 152.4, 139.7, 138.8, 138.2, 137.9, 129.5, 128.9, 128.3, 127.0, 125.6, 112.5, 34.3, 29.6, 21.1, 19.3. C21H19N3O2 MW 345.39; ESI-MS m/z 346.1176 [(M + H)+ 100% rel. I.]. HRMS: calc’d for C21H20N3O2 (M + H+): 346.1556, found: 346.1558. 0.6 ppm.
  • 6-(p-Methoxyphenyl)-4-methyl-3-phenethyl-isoxazolo[3,4-d]pyridazin-7(6H)-one, 3d. 1H NMR (400 MHz, CDCl3): δ 7.385 (d, J = 8 Hz, 2H); 7.16–7.22 (m, 3H); 7.7.65 (d, J + 8 Hz, d); 6.89 (d, J = 8 Hz, 2H); 3.73 (s, 3H); 3.38 (t, J = 8 Hz, 2H); 3.11 (t, J = 8 Hz, 2H); 2.20 (s, 3H). 13C NMR: 172.6, 159.0, 153.0, 152.4, 139.8, 138.9, 133.8, 128.9, 127.1,114.1, 55.6, 34.3, 29.6, 19.4. C21H19N3O3 MW: 361.3. HRMS calc’d for C21H20N3O3 (M + H+): 362.1505, found: 362.1506. 0.3 ppm.
  • 6-(p-Methoxyphenyl)-3-(1,3-diphenylpropan-2-yl)-4-methyl-isoxazolo[3,4-d]pyridazin-7(6H)-one, 4d. 1H NMR (400 MHz, CDCl3): δ 7.41 (d, J = 8 Hz, 2H); 7.19–7.25 (m, 6H); 7.19 (d, J = 8 Hz, 4H); 6.94 (d, J = 8 Hz, 2H); 3.83 (s, 3H); 3.81 (pentet, J = 8 Hz, 1H); 3.28 (d, J = 8 Hz, 4H); 1.85 (s, 3H). 13C NMR: 174.5, 159.0, 152.9, 151.9, 139.6, 138.0, 133.7, 128.8, 128.6, 126.9, 114.1, 113.4, 55.6, 45.0, 40.8, 19.0. C28H25N3O3 MW: 451.5. HRMS calc’d for C28H26N3O3 (M + H+): 452.1974, Found: 452.1975. 0.2 ppm.
  • 6-(3,5-Dichlorophenyl)-4-methyl-3-phenethylisoxazolo[3,4-d]pyridazin-7(6H)-one, 3f. 1H NMR (400 MHz, CDCl3): δ 7.515 (d, J = 4 Hz, 2H); 7.18–7.27 (m, 5H); 7.04 (d, 1H); 3.41 (t, 3 J = 8 Hz, 2H); 3.12 (t, 3 J = 8 Hz, 2H); 2.2 (s, 3H). 13C NMR: 173.0, 152.7, 152.0, 142.1, 140.9, 138.6, 134.8, 128.9, 128.3, 127.2, 124.2, 112.3, 34.2, 29.6, 19.3. C20H15Cl2N3O2 MW: 400.26; ESI-MS m/z 400 [(M + H+), 100% rel. I.]; 402 [(M + H) + 2, 67.7]; 404 [(M + H) + 4, 12.2]. HRMS Calc’d for C20H16Cl2N3O2 400.0620, Found: 400.0622. 0.5 ppm.
  • 6-(3,5-Dichlorophenyl)-4-methyl-3-(1,3-diphenylpropan-2-yl)-isoxazolo[3,4-d]pyridazin-7(6H)-one, 4f. 1H NMR (400 MHz, CDCl3): δ 7.532 (d, J = 1.7 Hz, 2H); 7.32 (t, J = 1.7,1H); 7.25–7.19 (m, 6H); 7.01–6.99 (d, J = 6.4 Hz, 4H); 3.81–3.77 (pentet, J = 8 Hz, 1H); 3.30 (s, 2H); 3.28 (s,2H); 1.85 (s, 3H);1.59 (s, 1H). 13C NMR: 175.0, 140.7, 137.9, 134.8, 128.8, 128.5, 127.2, 124.1; 45.2, 40.8, 18.9. C27H21Cl2N3O2 MW: 490.39; HRMS 489 [(M − H)+ 100% rel. I.]; 491 (M + H). Calc’d for C27H21Cl2N3O2 490.3805, Found: 490.1226.
  • 6-(3,5-Bistrifluoromethylphenyl)-4-methyl-3-(1,3-diphenylpropan-2-yl)-isoxazolo[3,4-d]pyridazin-7(6H)-one, 4e. 1H NMR (400 MHz, CDCl3): δ 8.13 (s, 2H);7.82(s, 1H); 7.04 (d, 1H); 7.25–7.18 (m, J = 8 Hz, 6H); 7.02–7.00 (d, J = 8 Hz, 4H); 3.82–3.79 (m,1H); 3.31 (s 2H); 3.29 (s, 2H); 1.88(s, 3H); 1.6 (s, 1H). C29H21F6N3O2 MW: 557.49; HRMS m/z 558 [(M + H)+ 100% rel. I.]; 559 [(M + H) + 2]; Calc’d for C29H21F6N3O2 557.49, Found: 557.1153.
  • 6-(3,5-Dichlorophenyl)-3-methyl-4-phenylisoxazolo[3,4-d]pyridazin-7(6H)-one, 2i. 1H NMR (400 MHz, CDCl3): δ 7. 68 (2H); 7.57 (5H); 7.38 (1H); 5.59 (s, 3H). 13C NMR: δ 208.2, 194.3, 181.6, 170.9, 164.9, 152.7, 152.0, 142.1, 140.7, 134.8, 134.6, 131.2; 129.3, 127.0, 126.7, 125.6, 124.2, 122.6, 112.5, 31.4, 28.7, 18.9. HRMS Calc’d for C18H1135Cl2N3O2 + H 372.0307, Found: 372.0309. 0.5 ppm.
  • 6-(3,5-Dichlorophenyl)-3-methyl-4-phenyl-6H,7H-[1,2]oxazolo [3,4-d]pyridazin-7-one, 2j. 1H NMR (400 MHz, CDCl3): δ 7.57–7.56 (d, 5H);7.37(t, 1H); 2.58 (s, 3H).13C NMR 171.0, 152.6, 143.7, 142.2, 134.9, 133.2, 130.5, 129.0, 128.4, 128.0, 124.3, 111.2, 14.0. C18H11Cl2N3O2 MW: 372.205; HRMS m/z 372.0309 [(M + H)+ 100% rel. I.]; 374.0289 (M + H + 2); Calc’d for C18H11Cl2N3O2 372.20538, Found: 372.0309.
  • 6-(4-Methoxyphenyl)-3-methyl-4-phenyl-6H,7H-[1,2]oxazolo [3,4-d]pyridazin-7-one, 2k. 1H NMR (400 MHz, CDCl3): δ 7.58–7.53 (t, J = 8, 7H); 7.00–6.98 (d, J = 8, 2H); 3.85(s, 3H); 2.58 (s, 3H).13 C NMR 170.4, 159.0, 152.8, 152.7, 142.6, 133.8, 130.1, 28.8, 128.4, 127.0, 114.0, 111.4, 55.5, 13.9. C19H15N3O3 MW: 333.35; HRMS m/z: 333.1113 [100% rel. I], 334.1147 (20.5%), 335.1181 (2.0%), 334.1084 (1.1%); Calc’d for C19H15N3O3 333.3407, Found: 333.1113.
  • 3-Methyl-6-(4-methylphenyl)-4-phenyl-6H,7H-[1,2]oxazolo [3,4-d]pyridazin-7-one, 2l. 1H NMR (400 MHz, CDCl3): δ 7.60–7.58 (m, J = 4, 2H); 7.55–7.53(t, J = 4, 5H); 2.59 (s, 3H); 2.40 (s, 3H).13C NMR 170.6, 152.8, 133.5, 130.3, 128.9, 128.4, 127.7, 115.8, 115.6, 111.4, 14.0. C19H15N3O2 MW: 317.35; HRMS Calc’d for C19H15N3O2 317.12, Found: 317.1164.
  • (E)-Ethyl-4-(1-(2-(3,5-dichlorophenyl)hydrazono)ethyl)-5-phenethylisoxazole-3-carboxylate, 7f. 1H NMR (400 MHz, CDCl3): δ 4.34 (q, J = 8 Hz, 2H); 3.17 (t, 2H); 2.98 (t, 2H); 1.80 (s, 3H); 1.32 (t, J = 8 Hz, 3H). C22H21Cl2N3O3 MW: 446.3; ESI-MS m/z 446 [(M + H)+, 100% rel. I.], 448 (M + 3+, 67.4).
  • 6-(3,5-Dichlorophenyl)-4-methyl-3-phenethylisoxazolo[3,4-d]pyridazin-7(6H)-one, 3f. 1H NMR (400 MHz, CDCl3): δ 7.515 (d, J = 4 Hz, 2H); 7.18–7.27 (m, 5H); 7.04 (d, 1H); 3.41 (t, 3 J = 8 Hz, 2H); 3.12 (t, 3 J = 8 Hz, 2H); 2.2 (s, 3H). 13C NMR: 173.0, 152.7, 152.0, 142.1, 140.9, 138.6, 134.8, 128.9, 128.3, 127.2, 124.2, 112.3, 34.2, 29.6, 19.3. C20H15Cl2N3O2 MW: 400.26; ESI-MS m/z 400 [(M + H)+, 100% rel. I.); 402 (M + H+2, 67.7); 404 (M + H + 4, 12.2). HRMS Calc’d for C20H16Cl2N3O2 400.0620, Found: 400.0622. 0.5 ppm.
  • 6-(3,5-Dichlorophenyl)-4-methyl-3-(2-(naphthalen-1-yl)ethyl)isoxazolo[3,4-d]pyridazin-7(6H)-one, 3g. 1H NMR (400 MHz, CDCl3): δ 7.96 (d, J = 8 Hz, 1H); 7.90 (d, J = 8 Hz, 1H); 7.79 (d, J = 8 Hz, 1H); 7.37–7.61 (m, H); 7.56 (d, J = 2 Hz, 2H); 7.35 (t, 1H); 7.35 (d, J = 2 Hz, 1H); 7.21 (d, J = 8 Hz, 1H), 3.675 (dd, 2H): 3.64 (dd, 2H); 2.00 (s, 3H). 13C NMR: δ 173.0, 152.7, 152.0, 142.1, 140.7, 134.8, 134.6, 131.2; 129.3, 127.0, 126.7, 125.6. 124.2, 122.6, 112.5, 31.4, 28.7, 18.9. C24H17Cl2N3O2 MW: 450.32; ESI-MS m/z 450 [(M + H)+,100% rel. I.]; 452 (M + H+2, 68.9); 452 (M + H+4, 13.1). HRMS Calc’d for C24H18Cl2N3O2 450.0776, Found: 450.0775. −0.2 ppm.
Kemp Elimination/Rearrangement products, General Experimental: To 25 mL of anhydrous ethanol was added sodium metal (82 mg, 3.5 mol). After the reaction to the alkoxide was complete, 1 mmol of isoxazolo[3,4-d] pyridazinone, followed by aldehyde (in a slight excess of 1 mmol), was added in one portion, and the reaction was heated to reflux overnight. The reaction was cooled, the ethanol concentrated by a rotary evaporator, and the residue chromatographed on silica gel using a 4:1:1 hexane-DCM-ethylacetate mixture.
  • 1-p-Tolyl-3-methyl-(4-p-Chlorocinnamyl)-5-amino-pyrazole, 15c. Ar1 = p-ClC6H4; Ar2 = p-CH3C6H4; C20H19ClN3O MW 352.8, ESI-MS: m/z 352.1 (100, M+)); 354.1 (M + 2+, 35).
  • 1-p-Anisyl-3- methyl -(4-p-Chlorocinnamyl)-5-amino-pyrazole, 15d. Ar1 = p-ClC6H4; Ar2 = p- CH3OC6H4; C20H19ClN3O2 MW 368.8, m/z 368.1 (100, M+); 370.1 (M + 2+, 35).
  • 1-[3,5-Dichlorophenyl]-3-methyl-(4-p-Methoxycinnamyl)-5-amino-pyrazole, 15f Ar1 = p-CH3OC6H4; Ar2 = 3,5-Cl2C6H3; C20H18Cl2N3O2 MW: 403.28; ESI-MS m/z 402 (100% rel. I.), 404 (M + 2, 65.5); 406 (M + 4, 11).

5. Conclusions

Intense interest in the development of ligands that bind metabotropic glutamate receptors continues [52,53,54,55,56,57,58,59,60], often employing the principles of structure-based hypothesis-driven drug design. The present report is an illustration of how new chemical synthetic methodologies can drive the discovery and exploration of novel biology. In our hands, Method A produced di-substituted alkyaltion products directly in the most efficient manner, and Method B, while requiring more steps, could selectively give rise to mono-subsitution. With mono- and di-substituted products in hand, we can proceed to further expand the series with double-substituted analogs at the C3 position, as well as explore asymmetric synthesis [61]. We are actively pursuing the synthesis of ligands for glutamate receptors and transporters, as well as their pharmacological evaluation, and will report on our progress in due course.

6. Patents

“Isoxazolyl[3,4-d] pyridazinones selectively bind metabotropic glutamate receptors”, N.R. Natale, Y.R. Mirzaei, C. Gates, and C. Koerner, Invention Disclosure UM Docket No. UMT-2013-00x, filed on 4 March 2013.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28196800/s1, representative spectra and characterization.

Author Contributions

Conceptualization, N.R.N. and C.A.G.; methodology, C.A.G.; software, D.S.B. and P.R.; validation, N.R.N. and P.R.; formal analysis, N.R.N.; investigation, all; resources, all; data curation, N.R.N. and P.R.; writing—original draft preparation, N.R.N. and C.A.G.; writing—review and editing, all; visualization, C.A.G., D.S.B. and P.R.; supervision, N.R.N.; project administration, N.R.N. and P.R.; funding acquisition, N.R.N., D.S.B. and P.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the ALSAM Foundation via grants to N.R.N., D.S.B., and P.R. In addition, N.R.N. thanks the University of Montana for Small Grants MRA796 and 325490, and C.A.G. acknowledges the support of the ASUM Research and Creative Scholarship and the Besancon Scholarship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Computational files are available from the authors.

Acknowledgments

The authors thank David Burkhart, Jared Nelson, and Trideep Rajale for the preparation of intermediates in sufficient quantity and purity to serve in the present study and thank Scott Steiger for scaling up the starting materials. C.A.G. thanks David Burkhart for helpful suggestions.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of most of the compounds are available from the authors.

List of Abbreviations

ArAryl
CDCl3Deuterated chloroform
CHARMmChemistry at Harvard Macromolecular Mechanics
CNSCentral nervous system
DCMDichloromethane or methylene chloride
ESIElectrospray ionization
GPCRG-protein-Coupled Receptor
mGluRMetabotropic glutamate receptor
HPLCHigh-performance liquid chromatography
HRMSHigh-resolution mass spectrometry
IPIsolated and purified yield
LDALithium diisopropyl amide
LiHMDSLithium hexamethyl disilazide
LM&EQLateral metalation and electrophilic quenching
MHzMega Hertz
MSMass spectrometry
NMRNuclear magnetic resonance
ONOvernight
PDBProtein data bank
PTSAp-Toluenesuflonic acid
RMSRoot mean squared gradient
RSMYield based on recovered starting material
7TMSeven-transmembrane receptor
THFtetrahydro furan
[3,4-d] isoxazolo[3,4-d] pyridazinone
VFDVenus flytrap domain or glutamate binding domain of mGluR2 and 4

References

  1. Conn, P.J.; Christopoulos, A.; Lindsley, C.W. Allosteric Modulators of GPCRs: A Novel Approach for the Treatment of CNS Disorders. Nat. Rev. Drug Discov. 2010, 8, 41–54. [Google Scholar] [CrossRef]
  2. Conn, P.J.; Lindsley, C.W.; Meiler, J.; Niswender, C.M. Opportunities and Challenges in the Discovery of Allosteric Modulators of GPCRs for Treating CNS Disorders. Nat. Rev. Drug Discov. 2014, 13, 692–708. [Google Scholar] [CrossRef] [PubMed]
  3. Engers, J.L.; Rodriguez, A.L.; Konkol, L.C.; Morrison, R.D.; Thompson, A.D.; Byers, F.W.; Blobaum, A.L.; Chang, S.; Venable, D.F.; Loch, M.T.; et al. Discovery of a Selective and CNS Penetrant Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 3 with Antidepressant and Anxiolytic Activity in Rodents. J. Med. Chem. 2015, 58, 7485–7500. [Google Scholar] [CrossRef]
  4. Volpi, C.; Fallarino, F.; Mondanelli, G.; Macchiarulo, A.; Grohmann, U. Opportunities and Challenges in Drug Discovery Targeting Metabotropic Glutamate Receptor 4. Expert Opin. Drug Discov. 2018, 13, 411–423. [Google Scholar] [CrossRef] [PubMed]
  5. Crupi, R.; Impellizzeri, D.; Cuzzocrea, S. Role of Metabotropic Glutamate Receptors in Neurological Disorders. Front. Mol. Neurosci. 2019, 12, 20. [Google Scholar] [CrossRef]
  6. Bai, M. Dimerization of G-Protein-Coupled Receptors: Roles in Signal Transduction. Cell. Signal. 2004, 16, 175–186. [Google Scholar] [CrossRef] [PubMed]
  7. De Filippis, B.; Lyon, L.; Taylor, A.; Lane, T.; Burnet, P.W.J.; Harrison, P.J.; Bannerman, D.M. The Role of Group II Metabotropic Glutamate Receptors in Cognition and Anxiety: Comparative Studies in GRM2-/-, GRM3-/- and GRM2/3-/- Knockout Mice. Neuropharmacology 2015, 89, 19–32. [Google Scholar] [CrossRef]
  8. Niswender, C.M.; Conn, P.J. Metabotropic Glutamate Receptors: Physiology, Pharmacology, and Disease. Ann. Rev. Pharmacol. Toxicol. 2010, 50, 295–322. [Google Scholar] [CrossRef]
  9. Renzi, G.; Dal Piaz, V. Ricerche Su Alcuni 3-Carbetossi-Isossazoli-4,5-Disostituiti. Gazz. Chim. It. 1965, 95, 1478–1491. [Google Scholar]
  10. Renzi, G.; Pinzauti, S. Nuovi Derivati Del Systema Isossazolo-[3,4-d]Piridazin-7-One. Farm. Ed. Sci. 1969, 24, 885–892. [Google Scholar]
  11. Campana, C.; Mirzaei, J.; Koerner, C.; Gates, C.; Natale, N.R. 3-(1,3-Diphenylpropan-2-yl)-4-methyl-6-phenylisoxazolo[3,4-d] pyridazin-7(6H)-one. Acta Cryst. 2013, E69, o1680. [Google Scholar] [CrossRef] [PubMed]
  12. Gates, C.; Backos, D.S.; Reigan, P.; Kang, H.J.; Koerner, C.; Mirzaei, J.; Natale, N.R. Isoxazolo[3,4-d]Pyridazinones Positively Modulate the Metabotropic Glutamate Subtypes 2 and 4. Bioorg. Med. Chem. 2018, 26, 4797–4803. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, H.; Wang, C.; Gregory, K.J.; Han, G.W.; Cho, H.P.; Xia, Y.; Niswender, C.M.; Katritch, V.; Meiler, J.; Cherezov, V.; et al. Structure of a Class C GPCR Metabotropic Glutamate Receptor 1 Bound to an Allosteric Modulator. Science 2014, 344, 58–64. [Google Scholar] [CrossRef] [PubMed]
  14. Brooks, B.R.; Brooks, C.L.; Mackerell, A.D.; Nilsson, L.; Petrella, R.J.; Roux, B.; Won, Y.; Archontis, G.; Bartels, C.; Boresch, S.; et al. CHARMM: The Biomolecular Simulation Program. J. Comput. Chem. 2009, 30, 1545–1614. [Google Scholar] [CrossRef]
  15. Onufriev, A.; Case, D.A.; Bashford, D. Effective Born Radii in the Generalized Born Approximation: The Importance of Being Perfect. J. Comput. Chem. 2002, 23, 1297–1304. [Google Scholar] [CrossRef]
  16. Koska, J.; Spassov, V.Z.; Maynard, A.J.; Yan, L.; Austin, N.; Flook, P.K.; Venkatachalam, C.M. Fully Automated Molecular Mechanics Based Induced Fit Protein—Ligand Docking Method. J. Chem. Inf. Model. 2008, 48, 1965–1973. [Google Scholar] [CrossRef]
  17. Feng, Z.; Ma, S.; Hu, G.; Xie, X. Allosteric Binding Site and Activation Mechanism of Class C G-Protein Coupled Receptors: Metabotropic Glutamate Receptor Family. AAPS J. 2015, 17, 737–753. [Google Scholar] [CrossRef]
  18. Isberg, V.; de Graaf, C.; Bortolato, A. Generic GPCR residue numbers—Aligning topology maps while minding the gaps. Trends Pharmacol. Sci. 2015, 36, 22–31. [Google Scholar] [CrossRef]
  19. Clark, R.D.; Waldman, M. Lions and tigers and bears, oh my! Three barriers to progress in computer-aided molecular design. J. Comput. Aided Mol. Des. 2012, 26, 29–34. [Google Scholar] [CrossRef]
  20. Jain, A.N.; Cleves, A.C. Does Your Model Weigh the Same as a Duck? J. Comput. Aided Mol. Des. 2012, 26, 57–67. [Google Scholar] [CrossRef]
  21. Micetich, R.G.; Chin, C.G. Studies in Isoxazole Chemistry. 111. The Preparation and Lithiation of 3,5-Disubstituted Isoxazoles. Can. J. Chem. 1970, 48, 1371. [Google Scholar] [CrossRef]
  22. Natale, N.R.; Niou, C.-S. A Facile Synthesis of Functionally Complex Isoxazole Derivatives. Tetrahedron Lett. 1984, 25, 3943–3946. [Google Scholar] [CrossRef]
  23. Natale, N.R.; McKenna, J.I.; Niou, C.S.; Borth, M.; Hope, H. Metalation of Isoxazolyloxazolines, a Facile Route to Functionally Complex Isoxazoles: Utility, Scope, and Comparison to Dianion Methodology. J. Org. Chem. 1985, 50, 5660–5666. [Google Scholar] [CrossRef]
  24. Natale, N.R.; Niou, C.-S. Synthesis, Metalation and Electrophilic Quenching of Alkyl-Isoxazole-4-Tertiary Carboxamides. A Critical Comparison of Three Isoxazole Lateral Metalation Methods. Heterocycles 1986, 24, 401. [Google Scholar] [CrossRef]
  25. Natale, N.R.; Mirzaei, Y.R. The Lateral Metalation of Isoxazoles. A Review. Org. Prep. Proced. Int. 1993, 25, 515–556. [Google Scholar] [CrossRef]
  26. Zhou, P.; Natale, N.R. Lateral Lithiation of Ethyl 4-Acetyl-5-Methyl-3-Isoxazolyl Carboxylate with 5,5-Dimethyl-l,3-Dioxanyi as a Directing Group. Tetrahedron Lett. 1998, 39, 8249–8252. [Google Scholar] [CrossRef]
  27. Burkhart, D.J.; Zhou, P.; Blumenfeld, A.; Twamley, B.; Natale, N.R. An Improved Procedure for the Lateral Lithiation of Ethyl 4-Acetyl-5-Methyl-3-Isoxazolyl Carboxylate. Tetrahedron 2001, 57, 8039–8046. [Google Scholar] [CrossRef]
  28. Burkhart, D.J.; McKenzie, A.R.; Nelson, J.K.; Myers, K.I.; Zhao, X.; Magnusson, K.R.; Natale, N.R. The Catalytic Asymmetric Synthesis of Glutamate Analogues. Org. Lett. 2004, 6, 1285–1288. [Google Scholar] [CrossRef]
  29. Montesano, F.; Barlocco, D.; Piaz, V.D.; Leonardi, A.; Poggesi, E.; Fanelli, F.; De Benedetti, P.G. Isoxazolo-[3,4-d]-Pyridazin-7-(6H)-Ones and Their Corresponding 4,5-Disubstituted-3-(2H)-Pyridazinone Analogues as New Substrates for A1-Adrenoceptor Selective Antagonists: Synthesis, Modeling, and Binding Studies. Bioorg. Med. Chem. 1998, 6, 925–935. [Google Scholar] [CrossRef]
  30. Costantino, L.; Rastelli, G.; Gamberini, M.C.; Giovannoni, M.P.; Piaz, V.D.; Vianello, P.; Barlocco, D. Isoxazolo-[3,4-d]-Pyridazin-7-(6H)-One as a Potential Substrate for New Aldose Reductase Inhibitors. J. Med. Chem. 1999, 42, 1894–1900. [Google Scholar] [CrossRef]
  31. Dal Piaz, V.; Rascón, A.; Dubra, M.E.; Giovannoni, M.P.; Vergelli, C.; Castellana, M.C. Isoxazolo[3,4-d]Pyridazinones and Analogues as Leishmania Mexicana PDE Inhibitors. Il Farmaco 2002, 57, 89–96. [Google Scholar] [CrossRef] [PubMed]
  32. Dal Piaz, V.; Castellana, M.C.; Vergelli, C.; Giovannoni, M.P.; Gavaldà, A.; Segarra, V.; Beleta, J.; Ryder, H.; Palacios, J.M. Synthesis and Evaluation of Some Pyrazolo[3,4-d]Pyridazinones and Analogues as PDE 5 Inhibitors Potentially Useful as Peripheral Vasodilator Agents. J. Enzyme Inh. Med. Chem. 2002, 17, 227–233. [Google Scholar] [CrossRef] [PubMed]
  33. Dal Piaz, V.; Giovannoni, M.P.; Ciciani, G.; Barlocco, D.; Giardina, G.; Petrone, G.; Clarke, G.D. 4,5-Functionalized 6-Phenyl-3(2H)-Pyridazinones: Synthesis and Evaluation of Antinociceptive Activity. Eur. J. Med. Chem. 1996, 31, 65–70. [Google Scholar] [CrossRef]
  34. Barlocco, D.; Cignarella, G.; Dal Piaz, V.; Giovannoni, M.P.; De Benedetti, P.G.; Fanelli, F.; Montesano, F.; Poggesi, E.; Leonardi, A. Phenylpiperazinylalkylamino Substituted Pyridazinones as Potent A1 Adrenoceptor Antagonists. J. Med. Chem. 2001, 44, 2403–2410. [Google Scholar] [CrossRef] [PubMed]
  35. Giovannoni, M.P.; Vergelli, C.; Ghelardini, C.; Galeotti, N.; Bartolini, A.; Dal Piaz, V. [(3-Chlorophenyl)Piperazinylpropyl]Pyridazinones and Analogues as Potent Antinociceptive Agents. J. Med. Chem. 2003, 46, 1055–1059. [Google Scholar] [CrossRef]
  36. Dal Piaz, V.; Vergelli, C.; Giovannoni, M.P.; Scheideler, M.A.; Petrone, G.; Zaratin, P. 4-Amino-3(2H)-Pyridazinones Bearing Arylpiperazinylalkyl Groups and Related Compounds: Synthesis and Antinociceptive Activity. Farmaco 2003, 58, 1063–1071. [Google Scholar] [CrossRef]
  37. Giovannoni, M.P.; Vergelli, C.; Biancalani, C.; Cesari, N.; Graziano, A.; Biagini, P.; Gracia, J.; Gavaldà, A.; Dal Piaz, V. Novel Pyrazolopyrimidopyridazinones with Potent and Selective Phosphodiesterase 5 (PDE5) Inhibitory Activity as Potential Agents for Treatment of Erectile Dysfunction. J. Med. Chem. 2006, 49, 5363–5371. [Google Scholar] [CrossRef]
  38. Giovannoni, M.P.; Cesari, N.; Vergelli, C.; Graziano, A.; Biancalani, C.; Biagini, P.; Ghelardini, C.; Vivoli, E.; Dal Piaz, V. 4-Amino-5-Substituted-3(2H)-Pyridazinones as Orally Active Antinociceptive Agents: Synthesis and Studies on the Mechanism of Action. J. Med. Chem. 2007, 50, 3945–3953. [Google Scholar] [CrossRef]
  39. Biancalani, C.; Giovannoni, M.P.; Pieretti, S.; Cesari, N.; Graziano, A.; Vergelli, C.; Cilibrizzi, A.; Di Gianuario, A.; Colucci, M.; Mangano, G.; et al. Further Studies on Arylpiperazinyl Alkyl Pyridazinones: Discovery of an Exceptionally Potent, Orally Active, Antinociceptive Agent in Thermally Induced Pain. J. Med. Chem. 2009, 52, 7397–7409. [Google Scholar] [CrossRef]
  40. Chimichi, S.; Ciciani, G.; Piaz, V.D.; De Sio, F.; Sarti-Fantoni, P.; Torroba, T. Solid State Photodimerization Reaction of Some 3-Styrylisoxazolo-[3,4-d]Pyridazin-7(6h)-Ones. Heterocycles 1986, 24, 3467–3471. [Google Scholar] [CrossRef]
  41. Dal Piaz, V.; Pinzauti, S.; Lacrimini, P.J. Condensation of Some 3-Methylisoxazolo[3,4-d]Pyridazin-7(6H)Ones with Aromatic Aldehydes. J. Heterocycl. Chem. 1976, 13, 409–410. [Google Scholar] [CrossRef]
  42. Dal Piaz, V.; Ciciani, G.; Costanzo, A.; Auzzi, G.; Chimichi, S. 1,2,3,4-Tetrahydro-1,2-diazepine derivatives from isoxazolo[3,4-d]pyridazin-7(6H)-ones. Heterocycles 1984, 22, 1741–1746. [Google Scholar] [CrossRef]
  43. Dal Piaz, V.; Ciciani, G.; Chimichi, S. New Functionalized Pyrazoles from Isoxazolopyridazinones. Heterocycles 1985, 23, 365–369. [Google Scholar] [CrossRef]
  44. Casey, M.L.; Kemp, D.S.; Paul, K.G.; Cox, D.D. The Physical Organic Chemistry of Benzisoxazoles. I. The Mechanism of the Base-Catalyzed Decomposition of Benzisoxazoles. J. Org. Chem. 1973, 38, 2294–2301. [Google Scholar] [CrossRef]
  45. Kemp, D.S.; Casey, M.L. Physical organic chemistry of benzisoxazoles II. Linearity of the brønsted free energy relationship for the base-catalyzed decomposition of benzisoxazoles. J. Am. Chem. Soc. 1973, 95, 6670–6680. [Google Scholar] [CrossRef]
  46. Röthlisberger, D.; Khersonsky, O.; Wollacott, A.M.; Jiang, L.; DeChancie, J.; Betker, J.; Gallaher, J.L.; Althoff, E.A.; Zanghellini, A.; Dym, O.; et al. Kemp elimination catalysts by computational enzyme design. Nature 2008, 453, 190–197. [Google Scholar] [CrossRef]
  47. Khersonsky, O.; Röthlisberger, D.; Dym, O.; Albeck, S.; Jackson, C.J.; Baker, D.; Tawfik, D.S. Evolutionary Optimization of Computationally Designed Enzymes: Kemp Eliminases of the KE07 Series. J. Mol. Biol. 2010, 396, 1025–1042. [Google Scholar] [CrossRef]
  48. Roberts, J.D.; Simmons, H.E., Jr.; Carlsmith, L.A.; Vaughan, C.W. Rearrangement in the reaction of Chlorobenzene-1-C14 with Potassium Amide. J. Am. Chem. Soc. 1953, 75, 3290–3291. [Google Scholar] [CrossRef]
  49. Goetz, A.E.; Shah, T.K.; Garg, N.K. Pyridynes and indolynes as building blocks for functionalized heterocycles and natural products. Chem. Comm. 2015, 51, 34–45. [Google Scholar] [CrossRef]
  50. Vitale, P.; Di Nunno, L.; Scilimati, A. Synthesis of N,N-dialkylaminobenzonitriles and halo-(N,N-dialkyl)benzamidines by reaction of halobenzonitriles with lithium amides. Tetrahedron 2011, 67, 6944–6952. [Google Scholar] [CrossRef]
  51. Di Nunno, L.; Vitale, P.; Scilimati, A. Effect of the aryl group substituent in the dimerization of 3-arylisoxazoles to syn 2,6-diaryl-3,7-diazatricyclo[4.2.0.02,5]octan-4,8-diones induced by LDA. Tetrahedron 2008, 64, 11198–11204. [Google Scholar] [CrossRef]
  52. Amalric, M.; Lopez, S.; Goudet, C.; Fisone, G.; Battaglia, G.; Nicoletti, F.; Pin, J.P.; Acher, F.C. Group III and Subtype 4 Metabotropic Glutamate Receptor Agonists: Discovery and Pathophysiological Applications in Parkinson’s Disease. Neuropharmacology 2013, 66, 53–64. [Google Scholar] [CrossRef] [PubMed]
  53. Leach, K.; Gregory, K.J. Molecular Insights into Allosteric Modulation of Class C G Protein-Coupled Receptors. Pharmacol. Res. 2017, 116, 105–118. [Google Scholar] [CrossRef] [PubMed]
  54. Panarese, J.D.; Engers, W.; Wu, D.; Bronson, Y.-J.J.; Macor, J.E.; Chun, J.; Rodriguez, A.L.; Felts, A.S.; Engers, A.L.; Loch, J.T.; et al. Discovery of VU2957 (Valiglurax): An MGlu4 Positive Allosteric Modulator Evaluated as a Preclinical Candidate for the Treatment of Parkinson’s Disease. ACS Med. Chem. Lett. 2018, 10, 255–260. [Google Scholar] [CrossRef]
  55. Yang, D.; Zhou, Q.; Labroska, V.; Qin, S.; Darbalaei, S.; Wu, Y.; Yuliantie, E.; Xie, L.; Tao, H.; Cheng, J.; et al. G Protein-Coupled Receptors: Structure- and Function-Based Drug Discovery. Signal Transduct. Target. Ther. 2021, 6, 1–27. [Google Scholar] [CrossRef] [PubMed]
  56. Harpsøe, K.; Boesgaard, M.W.; Munk, C.; Bräuner-Osborne, H.; Gloriam, D.E. Structural Insight to Mutation Effects Uncover a Common Allosteric Site in Class C GPCRs. Bioinformatics 2017, 33, 1116–1120. [Google Scholar] [CrossRef] [PubMed]
  57. Robichaud, A.J.; Engers, D.W.; Lindsley, C.W.; Hopkins, C.R. Recent Progress on the Identification of Metabotropic Glutamate 4 Receptor Ligands and Their Potential Utility as CNS Therapeutics. ACS Chem. Neurosci. 2011, 2, 433–449. [Google Scholar] [CrossRef]
  58. Lindsley, C.W.; Emmitte, K.A.; Hopkins, C.R.; Bridges, T.M.; Gregory, K.J.; Niswender, C.M.; Conn, P.J. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem. Rev. 2016, 116, 6707–6741. [Google Scholar] [CrossRef]
  59. Rovira, X.; Malhaire, F.; Scholler, P.; Rodrigo, J.; Gonzalez-Bulnes, P.; Llebaria, A.; Pin, J.-P.P.; Giraldo, J.; Goudet, C. Overlapping Binding Sites Drive Allosteric Agonism and Positive Cooperativity in Type 4 Metabotropic Glutamate Receptors. FASEB J. 2015, 29, 116–130. [Google Scholar] [CrossRef]
  60. Ferreira, L.G.; Lamba, D.; Consiglio, N.; Delle, R.; Dharmendra, I.; Yadav, K.; Kang, S.; Choi, S.; Basith, S.; Cui, M.; et al. Exploring G Protein-Coupled Receptors (GPCRs) Ligand Space via Cheminformatics Approaches: Impact on Rational Drug Design. Front. Pharmacol. 2018, 9, 128. [Google Scholar]
  61. Gates, C.A. Development of the Novel Isoxazolo[3,4-d]pyridazinone scaffold, Which Positively Modulates the Metabotropic Glutamate Receptor Subtypes 2 and 4. Graduate Student Theses, Dissertations, & Professional Papers 11830. 2022. Available online: https://scholarworks.umt.edu/etd/11830 (accessed on 21 September 2023).
Figure 1. For mGluR2, the unique, non-conserved residues have the potential to associate with the fluorinated N-aryl group of 4e, according to computation.
Figure 1. For mGluR2, the unique, non-conserved residues have the potential to associate with the fluorinated N-aryl group of 4e, according to computation.
Molecules 28 06800 g001
Figure 2. Model for plausible selectivity of ligand 4f at mGluR4. The unique residue Leu756 interacts with the 3,5-dichloro aryl. The unique residue Leu756 (magenta) and allosteric pocket residues Trp798 and Phe801 (gold) are highlighted.
Figure 2. Model for plausible selectivity of ligand 4f at mGluR4. The unique residue Leu756 interacts with the 3,5-dichloro aryl. The unique residue Leu756 (magenta) and allosteric pocket residues Trp798 and Phe801 (gold) are highlighted.
Molecules 28 06800 g002
Scheme 1. Synthesis of substituted isoxazolo[3,4-d]pyridazinones. (A). Method A, direct metalation of 2af conducted in THF. (B). Method B, mono-substitution of acetal 5, followed by deprotection and cyclization of hydrazone 7. Reagents: a. corresponding ArNHNH2 (Aryl hydrazine), b. non-nucleophilic base, THF, and low temperature, as noted in Table 1, allow to warm to ambient temperature, c. 2,2-dimethyl-propane-1,3- diol, p-Toluenesulfonic acid (PTSA), reflux, d. non-nucleophilic base, low temperature, THF, followed by electrophile, e. addition of corresponding ArNHNH2, reflux.
Scheme 1. Synthesis of substituted isoxazolo[3,4-d]pyridazinones. (A). Method A, direct metalation of 2af conducted in THF. (B). Method B, mono-substitution of acetal 5, followed by deprotection and cyclization of hydrazone 7. Reagents: a. corresponding ArNHNH2 (Aryl hydrazine), b. non-nucleophilic base, THF, and low temperature, as noted in Table 1, allow to warm to ambient temperature, c. 2,2-dimethyl-propane-1,3- diol, p-Toluenesulfonic acid (PTSA), reflux, d. non-nucleophilic base, low temperature, THF, followed by electrophile, e. addition of corresponding ArNHNH2, reflux.
Molecules 28 06800 sch001
Scheme 2. Rearrangement of the [3,4-d] scaffold has been observed in previous studies [41,42,43] and is predicated on the Kemp elimination [44,45,46,47].
Scheme 2. Rearrangement of the [3,4-d] scaffold has been observed in previous studies [41,42,43] and is predicated on the Kemp elimination [44,45,46,47].
Molecules 28 06800 sch002
Scheme 3. The 3,5-dichloro presents the possibility of benzyne formation [48,49,50,51]. However, the cine-substitution products were not observed by HPLC-MS.
Scheme 3. The 3,5-dichloro presents the possibility of benzyne formation [48,49,50,51]. However, the cine-substitution products were not observed by HPLC-MS.
Molecules 28 06800 sch003
Table 1. Representative reaction conditions and yields for LM&EQ.
Table 1. Representative reaction conditions and yields for LM&EQ.
EntrySM aMono-Yield b (%) cDi-Yield b (%) cMethodBaseT (°C)
12a3a
(36)
4a
(23)
ALiHMDS−78
22a3a
trace
4a
38
A2 LiHMDS−78
32c3c
29(45)
ALiHMDS−20
42c3h
8 (15)
ALDA−25
52d3d
18 (22)
4d
5
ALiHMDS−40
62d3d
(33)
4d
27
(67)
ALiHMDS−80
75a3f
(34)
Bd
85a3g (45) Be
a, starting material (SM). b, yield in percent after isolation and purification (IP). c, RSM = percent yield adjusted for recovered starting material. d, yield is for the final step; metalation is described in reference [27]. e, yield is for the final step; metalation is described in reference [28].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gates, C.A.; Backos, D.S.; Reigan, P.; Natale, N.R. The Lateral Metalation of Isoxazolo[3,4-d]pyridazinones towards Hit-to-Lead Development of Selective Positive Modulators of Metabotropic Glutamate Receptors. Molecules 2023, 28, 6800. https://doi.org/10.3390/molecules28196800

AMA Style

Gates CA, Backos DS, Reigan P, Natale NR. The Lateral Metalation of Isoxazolo[3,4-d]pyridazinones towards Hit-to-Lead Development of Selective Positive Modulators of Metabotropic Glutamate Receptors. Molecules. 2023; 28(19):6800. https://doi.org/10.3390/molecules28196800

Chicago/Turabian Style

Gates, Christina A., Donald S. Backos, Philip Reigan, and Nicholas R. Natale. 2023. "The Lateral Metalation of Isoxazolo[3,4-d]pyridazinones towards Hit-to-Lead Development of Selective Positive Modulators of Metabotropic Glutamate Receptors" Molecules 28, no. 19: 6800. https://doi.org/10.3390/molecules28196800

Article Metrics

Back to TopTop