Synthesis and Evaluation of a Series of ... - ACS Publications

May 27, 2009 - Synthesis and Evaluation of a Series of Heterobiarylamides That Are Centrally Penetrant Metabotropic Glutamate Receptor 4 (mGluR4) ...
1 downloads 0 Views 779KB Size
J. Med. Chem. 2009, 52, 4115–4118 4115 DOI: 10.1021/jm9005065

*To whom correspondence should be addressed. Phone: 615-9366892. Fax: 615-322-8577. E-mail: [email protected]. a Abbreviations: mGluR, metabotropic glutamate receptor; PAM, positive allosteric modulator; HTS, high-throughput screening; tPSA, total polar surface area; LE, ligand efficiency; HLM, human liver microsomes; RLM, rat liver microsomes; PPB, protein binding.

allosteric modulation of mGluR4 (Figure 1).12-16 Unfortunately, all of the disclosed mGluR4 PAMs are deficient in their penetration into the CNS and their effects have only been demonstrated via intracerebroventricular (icv) injection or with the use of toxic vehicles such as a 50% DMSO solution. Herein, we report a class of centrally penetrant mGluR4 PAMs which can be administered in a nontoxic vehicle. A high-throughput screening (HTS) campaign was initiated at Vanderbilt to identify novel mGluR4 PAMs.12-14 In addition to the ligands shown in Figure 1, there were a number of small arylamide compounds identified as having mGluR4 PAM activity (Figure 2). These were attractive hits because of their favorable calculated properties (MW < 300, cLogP < 3.50, total polar surface area (tPSA) < 40, ligand efficiency (LE)17 > 0.30). From these lead compounds we initiated an optimization program in order to further profile this chemical series as novel mGluR4 PAMs. Initial effort was directed at the left-side heteroarylamide portion for which the synthesis is outlined in Scheme 1. A small library utilizing the furylamide revealed that the 3,4-dichloroaniline displayed good potency and efficacy at the hmGluR4 receptor. Next, a more detailed SAR evaluation of this structural class was undertaken. Keeping the right-hand aniline constant as the 3,4-dichlorophenyl, we first evaluated different aryl, cycloalkyl, and heteroarylamide replacements (Table 1). One issue that has been observed for many allosteric ligands is the noted intractable SAR;14,19,20 this property was also observed for the amide modifications. While the furylamide, 8a, was active in the micromolar range with good efficacy (>5 μM, 135% GluMax) and bromo substitution was tolerated (8b, 4.1 μM, 62% GluMax), further modifications led to loss of activity (see 8c,d). An additional survey of five-membered heterocycles was undertaken, but all compounds were inactive as mGluR4 PAMs (8e-g). Substituting a cyclohexyl, 8h, or phenyl, 8i, amide for the furyl also led to inactive compounds. However, upon introduction of the 2-pyridyl group, good potency and GluMax values were restored (8j, 1.4 μM, 80% GluMax). The 2-pyrazine compound, 8k, lost activity, while the 4-pyrimidine, 8l, retained potency, again highlighting this feature of intractable SAR around allosteric modulators. On the basis of these results, the 2-pyridylamide was chosen to further analyze for SAR.21 Starting from the 3,4-dichloroaniline derivative 8j, a number of halogenated and/or oxygenated compounds were synthesized and evaluated (Table 2). Compounds 9a-p were synthesized and tested, and a clear improvement on the HTS lead was identified. Compounds 9a-c were the best compounds evaluated in this series and presently are some of the best compounds for mGluR4 PAM activity that have been disclosed to date. These compounds have excellent potency at hmGluR4 (240-780 nM) and rmGluR4 (80-370 nM) receptors and exhibit excellent efficacy (hGluMax, 182-235%). The SAR shows that oxygen in the 3-position is favored; however, a 3-Cl substituent also imparts activity. As has been seen with other allosteric modulators, the SAR is very narrow with very minor modifications, leading to compounds with reduced or no activity. A number of modifications highlight this fact; see 9c (340 nM) vs 9d (>5 μM), where a simple

r 2009 American Chemical Society

Published on Web 05/27/2009

Synthesis and Evaluation of a Series of Heterobiarylamides That Are Centrally Penetrant Metabotropic Glutamate Receptor 4 (mGluR4) Positive Allosteric Modulators (PAMs)

)

Darren W. Engers,†,§ Colleen M. Niswender,†,§ C. David Weaver,†,§,‡ Satyawan Jadhav,†,§ Usha N. Menon,†,§ Rocio Zamorano,†,§ P. Jeffrey Conn,†,§,‡ Craig W. Lindsley,†,§,‡, and Corey R. Hopkins*,†,§ )

† Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, and Department of Chemistry, § Vanderbilt Program in Drug Discovery, and ‡Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232

Received April 21, 2009 Abstract: We report the synthesis and evaluation of a series of heterobiaryl amides as positive allosteric modulators of mGluR4. Compounds 9b and 9c showed submicromolar potency at both human and rat mGluR4. In addition, both 9b and 9c were shown to be centrally penetrant in rats using nontoxic vehicles, a major advance for the mGluR4 field.

The metabotropic glutamate receptors (mGluRsa) are members of the GPCR family C, characterized by a large extracellular amino-terminal binding domain (agonist) along with a seven-transmembrane spanning (7TM) domain which is the binding site for most known mGluR allosteric modulators.1-3 The eight cloned mGluRs have been assigned to three groups (groups I, II, III) based on their structural similarity, ligand specificity, and preferred coupling mechanisms.4 The group I subfamily is composed of mGluR1 and mGluR5; group II receptors include mGluR2 and mGluR3; and the group III receptors are represented by mGluR4, mGluR6, mGluR7, and mGluR8. Among the mGluRs, the group III receptors have thus far received less attention in terms of their therapeutic potential because of the paucity of selective ligands. However, recently there have been numerous reports detailing the potential benefits of mGluR4 activation in several disease models, most notably rodent models of Parkinson’s disease.5,6 It has been shown that activation of mGluR4 decreases GABAergic transmission at the inhibitory striato-pallidal synapse with the basal ganglia, a mechanism that is expected to provide palliative benefit for the treatment of Parkinson’s disease.7 In addition, there have been recent reports detailing the neuroprotective effects of an mGluR4 PAM in cultured neurons and in vivo.8,9 The most well characterized mGluR4 PAM for many years has been the compound PHCCC, 1, a partially selective mGluR4 potentiator.8,10,11 More recently, our laboratory and others have expanded the list of novel probes for positive

pubs.acs.org/jmc

4116 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 14

Engers et al. Table 1. Amide SAR

Figure 1. Chemical structure of (-)-PHCCC, 1, the mGluR4 ago-potentiator VU0155041, 2, and mGluR4 PAMs VU0001171, 3, VU0080241, 4, and VU0092145, 5.

Figure 2. Initial HTS hits.

Scheme 1. Synthesis of Amides 8 and 9a

a EC50 and GluMax are the average of at least three independent determinations performed in triplicate. a Reagents and conditions: (a) for acid chloride, DIEA, DMF, 12 h; (b) for carboxylic acids, EDCI, HOBt, 1,4-dioxane, 50 °C, 12 h. All library compounds were purified by mass-directed HPLC when required.18

modification of a difluoromethoxy to a trifluoromethoxy imparts a >10-fold loss of activity. Substituting the phenyl ring of the aniline with a pyridine (9n,o) or pyrimidine (9p) also led to compounds with much reduced activity (9b (240 nM) vs 9n (>5 μM)) or inactive compounds (9o,p). A number of compounds were synthesized to examine the 3methoxy substitution by making longer alkyl chain substitutions or by cyclization (data not shown). However, each of these modifications led to inactive or weakly active compounds. A number of the more active compounds were next evaluated for their ability to shift the glutamate response curve to the left. The fold shift for both the human and rat receptors is shown in Table 2. Many of the compounds that were analyzed showed a robust shift of the glutamate response (Figure 3, 9b). The SAR for this series of compounds was also mirrored in the fold shift data. As was observed in the potency data (Table 2), 9a-c were the most potent compounds and they also produced the largest fold shift in the human and rat cell lines. In addition to these compounds, 9f and 9j also produced robust fold shifts, giving this series several compounds with fold shifts of >15.

In addition, the selectivity of a few compounds (9a-c) was determined among the various mGluR subtypes. Although these compounds are very active at the human and rat mGluR4 receptor, they showed little activity against the other mGluRs, with the exceptions being weak PAM activity at mGluR5 and mGluR8. For example, 9b and 9c showed weak PAM activity at mGluR5 and -8 (Supporting Information, Table 1); however, there is more than a 20-fold separation in potencies compared to mGluR4. On the basis of the potency and efficacy of 9a-e, and the very favorable calculated properties, we next looked at metabolic stability and protein binding (PPB) (Table 3). These compounds were not stable in human or rat liver microsomes (HLM, RLM), with three compounds (9a-c) having less than 10% of the parent remaining after the incubation period and two compounds (9d,e) having less than 25% remaining. However, it is worth noting that 9d and 9e are significantly more stable; neither compound possesses the metabolically unstable methoxy group. All of these compounds (except 9d) possess favorable % free fraction in human and rat protein binding experiments. Because of these findings and the potency of these compounds, 9b and 9c were advanced further for in vivo DMPK analysis. The hydrochloride salts of these compounds (9b,c) were synthesized and dosed intraperitoneally (10 mg/kg) as an aqueous microsuspension containing 10% Tween-80, and

Letter

Journal of Medicinal Chemistry, 2009, Vol. 52, No. 14

4117

Table 2. SAR of the Aniline

Figure 3. Potency and efficacy of the novel mGluR4 PAM, 9b. (a) Compound 9b was added in progressively higher concentrations to cell coexpressing human mGluR4 and the chimeric G protein Gqi5 (white boxes). After a 2.5 min incubation period, an EC20 concentration of glutamate was added (black boxes) and the change in fluorescence was monitored using a kinetic imaging plate reader. Data are three independent determinations performed in triplicate. (b) DMSO vehicle or 30 μM 9b was incubated with human mGluR4/ Gqi5 cells, and 2.5 min later increasing concentrations of glutamate were added. In this experiment, performed in triplicate and representative of three independent determinations, 9b shifted the glutamate concentration-response curve 33-fold to the left. Table 3. In Vitro Pharmacokinetic Evaluation of 9a-c

a EC50, GluMax, and FS are the average of at least three independent determinations performed in triplicate.

compd

HLM (% remaining)

RLM (% remaining)

PPB (h) (% free)

PPB (r) (% free)

9a 9b 9c 9d 9e

3.0 5.3 2.4 22 26

6.3 1.8 1.4 20 19

5.1 2.2 4.0 0.8 1.3

6.5 2.4 5.8 1.6 2.9

Table 4. Rat Pharmacokinetic Data for Compounds 9b and 9c

the amount of compound present in brain and plasma was determined at 0.5, 1, and 8 h after administration (Table 4). Consistent with the poor in vitro microsomal stability, these compounds showed high clearance and low plasma exposure in rats. However, the brain levels for 9b and 9c were significant when compared to the total amount of compound, indicative of good brain penetration for these amides. In summary, we report a series of small molecule mGluR4 positive allosteric modulators. These compounds represent a series of 2-pyridylamide compounds that possess excellent calculated properties, making them ideal candidates for tool compounds. In addition, a number of compounds have excellent in vitro potency and efficacy at both the human and rat mGluR4 receptor, and many possess the ability to robustly shift the glutamate response to the left (>15). Selected compounds were further profiled for selectivity, in vitro PK, and ultimately in vivo PK. Two compounds, 9b and 9c, although possessing less than ideal in vitro PK parameters, show sufficient brain penetration to enable further evaluation

9b

9c

parameters

plasma

brain

plasma

brain

Cmax (ng/mL or g) Tmax (h) AUC (ng 3 h/mL or g) T1/2 (h) clearance/F ((mL/min)/kg) AUC0-8h, rain/AUC0-8h,plasma

154 0.5 173 1.9 894

1584 0.5 705 0.15

188 0.5 99 0.26 1419

1679 0.5 976 0.39

4.1

9.9

in anti-Parkinsonian in vivo rodent models, which will be reported in due course. Acknowledgment. The authors thank the assistance of members of the Vanderbilt HTS facility, Miranda Nolan for the in vitro PK, and Matt Mulder, Chris Denicola, and Sichen Chang for the purification of compounds utilizing the mass-directed HPLC system. This work was supported by the National Institute of Mental Health, the Michael J. Fox Foundation, the Vanderbilt Department of Pharmacology and the Vanderbilt Institute of Chemical Biology.

4118 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 14

Supporting Information Available: Experimental procedures, spectroscopic data, and NMR data for selected compounds and biological procedures. This material is available free of charge via the Internet at http://pubs.acs.org.

References (1) Conn, P. J.; Pin, J.-P. Pharmacology and functions of metabotropic glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 1997, 37, 205–237. (2) Marino, M. J.; Conn, P. J. Glutamate-based therapeutic approaches: allosteric modulators of metabotropic glutamate receptors. Curr. Opin. Pharmacol. 2006, 6 (1), 98–102. (3) Conn, P. J.; Christopoulos, A.; Lindsley, C. W. Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nat. Rev. Drug Discovery 2009, 8 (1), 41–54. (4) Schoepp, D. D.; Jane, D. E.; Monn, J. A. Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology 1999, 38 (10), 1431–1476. (5) Hopkins, C. R.; Lindsley, C. W.; Niswender, C. M. mGluR4 positive allosteric modulation as potential treatment of Parkinson’s disease. Future Med. Chem., in press. (6) Hefti, F. F. Parkinson’s Disease. Drug Discovery for Nervous System Diseases; Wiley-Interscience: Hoboken, NJ, 2005; pp 183204. (7) Valenti, O.; Marino, M. J.; Wittmann, M.; Lis, E.; DiLella, A. G.; Kinney, G. G.; Conn, P. J. Group III metabotropic glutamate receptor-mediated modulation of the striatopallidal synapse. J. Neurosci. 2003, 23 (18), 7218–7226. (8) Maj, M.; Bruno, V.; Dragic, Z.; Yamamoto, R.; Battaglia, G.; Inderbitzin, W.; Stoehr, N.; Stein, T.; Gasparini, F.; Vranesic, I.; Kuhn, R.; Nicoletti, F.; Flor, P. J. (-)-PHCCC, a positive allosteric modulator of mGluR4: characterization, mechanism of action, and neuroprotection. Neuropharmacology 2003, 45 (7), 895–906. (9) Battaglia, G.; Busceti, C. L.; Molinaro, G.; Giagioni, F.; Traficante, A.; Nicoletti, F.; Bruno, V. Pharmacological activation of mGluR4 metabotropic glutamate receptors reduces nigrostriatal degeneration in mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. J. Neurosci. 2006, 26 (27), 7222–7229. (10) Annoura, H.; Fukunaga, A.; Uesugi, M. A novel class of antagonists for metabotropic glutamate receptors, 7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxylates. Bioorg. Med. Chem. Lett. 1996, 6 (7), 763–766. (11) Marino, M. J.; Williams, D. L., Jr.; O’Brien, J. A.; Valenti, O.; McDonald, T. P.; Clements, M. K.; Wang, R.; DiLella, A. G.; Kinney, G. G.; Conn, P. J. Allosteric modulation of group III metabotropic glutamate receptor 4: a potential approach to Parkinson’s disease treatment. Proc. Natl. Acad. Sci. U.S.A. 2003, 100 (23), 13668-13673.

Engers et al. (12) Niswender, C. M.; Johnson, K. A.; Weaver, C. D.; Jones, C. K.; Xiang, Z.; Luo, Q.; Rodriguez, A. L.; Marlo, J. E.; de Paulis, T.; Thompson, A. D.; Days, E. L.; Nalywajko, T.; Austin, C. A.; Williams, M. B.; Ayala, J. E.; Williams, R.; Lindsley, C. W.; Conn, P. J. Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4. Mol. Pharmacol. 2008, 74 (5), 1345–1358. (13) Niswender, C. M.; Lebois, E. P.; Luo, Q.; Kim, K.; Muchalski, H.; Yin, H.; Conn, P. J.; Lindsley, C. W. Positive allosteric modulators of the metabotropic glutamate receptor subtype 4 (mGluR4): Part I. Discovery of pyrazolo[3,4-d]pyrimidines as novel mGluR4 positive allosteric modulators. Bioorg. Med. Chem. Lett. 2008, 18 (20), 5626–5630. (14) Williams, R.; Niswender, C. M.; Luo, Q.; Le, U.; Conn, P. J.; Lindsley, C. W. Positive allosteric modulators of the metabotropic glutamate receptor subtype 4 (mGluR4). Part II: Challenges in hitto-lead. Bioorg. Med. Chem. Lett. 2009, 19 (3), 962–966. (15) Reynolds, I. J. Metabotropic Glutamate Receptors As Therapeutic Targets in Parkinson’s Disease. Presented at the 6th International Meeting on Metabotropic Glutamate Receptors, Taormina, Sicily, Italy, 14 September 14-19, 2008. (16) Ortuno, D.; Cheng, C.; Weiss, M.; Bergeron, M.; Shanker, Y. Identification and Characterization of a Potent and Selective Positive Allosteric Modulator of mGluR4. Presented at the Society for Neuroscience, Washington, DC, November 15-19, 2008. (17) Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful metric for lead selection. Drug Discovery Today 2004, 9 (10), 430–431. (18) Leister, W.; Strauss, K.; Wisnoski, D.; Zhao, Z.; Lindsley, C. Development of a custom high-throughput preparative liquid chromatography/mass spectrometer platform for the preparative purification and analytical analysis of compound libraries. J. Comb. Chem. 2003, 5 (3), 322–329. (19) Zhao, Z.; Wisnoski, D. D.; O’Brien, J. A.; Lemaire, W.; Williams, D. L. Jr.; Jacobsen, M. A.; Wittman, M.; Ha, S. N.; Schaffhauser, H.; Sur, C.; Pettibone, D. J.; Duggan, M. E.; Conn, P. J.; Hartmann, G. D.; Lindsley, C. W. Challenges in the development of mGluR5 positive allosteric modulators: the discovery of CPPHA. Bioorg. Med. Chem. Lett. 2007, 17 (5), 1386–1391. (20) Lindsley, C. W.; Wisnoski, D. D.; Leister, W. H.; O’Brien, J. A.; Lemaire, W.; Williams, D. L. Jr.; Burno, M.; Sur, C.; Kinney, G. G.; Pettibone, D. J.; Tiller, P. R.; Smith, S.; Duggan, M. E.; Hartman, G. D.; Conn, P. J.; Huff, J. R. Discovery of positive allosteric modulators for the metabotropic glutamate subtype 5 from a series of N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamides that potentiate function in vivo. J. Med. Chem. 2004, 47 (24), 5825–5828. (21) Bolea, C. Amido Derivatives and Their Use as Positive Allosteric Modulators of Metabotropic Glutamate Receptors. PCT Int. Appl. WO2009/010454, 2009; 67 pp.