Skip to main content
ACS Medicinal Chemistry Letters logoLink to ACS Medicinal Chemistry Letters
. 2012 Sep 4;3(11):931–935. doi: 10.1021/ml300209g

Synthesis and SAR Studies of Fused Oxadiazines as γ-Secretase Modulators for Treatment of Alzheimer's Disease

Xianhai Huang †,*, Wei Zhou , Xiaoxiang Liu , Hongmei Li , George Sun , Mihirbaran Mandal , Monica Vicarel , Xiaohong Zhu , Chad Bennett , Troy McCraken , Dmitri Pissarnitski , Zhiqiang Zhao , David Cole , Gioconda Gallo , Zhaoning Zhu , Anandan Palani , Robert Aslanian , John Clader , Michael Czarniecki , William Greenlee , Duane Burnett , Mary Cohen-Williams , Lynn Hyde , Lixin Song §, Lili Zhang §, Inhou Chu , Alexei Buevich
PMCID: PMC4025817  PMID: 24900409

Abstract

graphic file with name ml-2012-00209g_0009.jpg

Fused oxadiazines (3) were discovered as selective and orally bioavailable γ-secretase modulators (GSMs) based on the structural framework of oxadiazoline GSMs. Although structurally related, initial modifications showed that structure–activity relationships (SARs) did not translate from the oxadiazoline to the oxadiazine series. Subsequent SAR studies on modifications at the C3 and C4 positions of the fused oxadiazine core helped to identify GSMs such as compounds 8r and 8s that were highly efficacious in vitro and in vivo in a number of animal models with highly desirable physical and pharmacological properties. Further improvements of in vitro activity and selectivity were achieved by the preparation of fused morpholine oxadiazines. The shift in specificity of APP cleavage rather than a reduction in overall γ-secretase activity and the lack of changes in substrate accumulation and Notch processing as observed in the animal studies of compound 8s confirm that the oxadiazine series of compounds are potent GSMs.

Keywords: Alzheimer's disease, oxadiazine, amyloid precursor protein, γ-secreatase modulator, Notch processing, morpholine


Alzheimer's disease (AD) is an age-related neurodegenerative disorder that affects millions of elderly people in the United States. It is estimated that more than 35 million people suffer from AD worldwide, with an annual cost of over $600 billion, and the population may increase to more than 115 million by 2050.1 Because of clear unmet medical need, both academic and industrial laboratories are working very aggressively to develop therapies to halt or even reverse AD. It is believed that the accumulation of amyloid-β (Aβ) peptide and hyperphosphorylated protein τ contributes to AD progression.26 Aβ peptide is formed from a larger amyloid precursor protein (APP) via sequential proteolytic cleavage by β- and γ-secretases (Figure 1).7 γ-Secretase cleaves the APP C-terminal fragment at multiple sites leading to Aβ peptides of 37–42 amino acids of which Aβ42, the more hydrophobic form, is the most amyloidogenic and neurotoxic. Although the molecular mechanism of action remains largely unknown, γ-secretase modulators (GSMs) are believed to act at an allosteric site to shift the predominant site of γ-secretase cleavage toward shorter, nonamyloidogenic peptides (e.g., Aβ38) by selectively inhibiting Aβ42 formation without blocking overall γ-secretase function. This potentially offers a better selectivity window over γ-secretase inhibitors (GSIs)810 versus, for example, notch processing.11 From an in vitro point of view, the Aβtotal/Aβ42 ratio can be used to distinguish GSMs from GSIs with a ratio of >10 for GSMs and <3 for GSIs. There are two major classes of GSMs in clinical trials: one is the nonsteroidal anti-inflammatory acids (NSAIDs),12,13 and the other is the non-NSAIDs class such as lactam 1 from Eisai. In our effort in the AD area, we have recently identified multiple structural classes1418 of GSMs with good in vitro potency and selectivity. Herein, we report our continued structure–activity relationship (SAR) effort focusing on novel fused oxadiazine core structures, which resulted in the identification of compounds with potent in vitro and in vivo activity and highly desirable physicological and physical properties.

Figure 1.

Figure 1

Sequential cleavage of APP by β- and γ-secretases.

Focusing on the non-NSAID GSM class represented by compound 1 to further optimize its overall pharmacological and pharmacokinetic (PK) profile and address its lenticular toxicity issue,7 we have recently identified cyclic hydroxyamidines such as oxadiazolines (2, Figure 2) as highly efficacious GSMs in both in vitro studies and in vivo animal models. These oxadiazolines were designed as novel isosteric replacements of amides with a consideration of hydrogen-bonding characteristics and lack of strong basicity. They were found to not only be chemically stable but also possess highly desirable PK and toxicological profiles.17 With the validation of five-membered cyclic hydroxyamidines as lactam carbonyl isosteric replacements and to further improve properties of this series of compounds, we became interested in fused oxadiazines (3), which are modestly basic and could offer opportunities to improve activity and enable salt formation for ease of formulation for rapid absorption.

Figure 2.

Figure 2

Design rationale of oxadiazine GSMs.

Equipped with SAR information from the oxadiazoline series, we believed that it would be possible to identify a lead compound from the oxadiazine series considering the similarity of the two core structures. Because the left-hand imidazolylphenyl unit is necessary for potency,17 we chose to focus our SAR studies on the modification of the right-hand side. Because of better activity of C3-disubstituted compound to monosubstituted oxadiazolines,17 we first prepared C4-disubstituted oxadiazine compounds 46 (racemic, Figure 3). To our surprise, it turned out that the SAR clearly did not translate from the oxadiazoline to the oxadiazine series. C4 methyl 4-fluorophenyl-substituted compound 4 lost in vitro potency by 4-fold vs 4′, and the spiro compound 6 was about 7-fold less active as compared to 6′. More dramatically, hydroxylmethyl oxadiazine 5 was much less active than oxadiazoline compound 5′. This SAR trend prompted us to consider that the two series might have different conformational interactions with the enzyme such that substitution at C3 of the oxadiazine core was preferred. Therefore, we decided to turn our effort to C3 modifications and carry out focused SAR studies (third degree SAR studies).19

Figure 3.

Figure 3

Comparison between oxadiazoline/oxadiazine of initial SAR studies. Each IC50 value is an average of at least two determinations.

Indeed, more tractable SAR was realized with the modifications at C3 (Table 1). We first introduced a hydroxymethyl group for ease of functionalization. Although this compound (7a) was not very potent, its TBDPS protected form (7b) caught our attention due to its much improved Aβ42 inhibition even though the Aβtotal/Aβ42 selectivity was not desirable. We then explored the benzyl (7c) and phenyl (7d) ethers, which displayed moderated activity, with 7d having improved selectivity as compared to compound 7b. This suggested that the aromatic ring might be playing a role in the activity. By moving the aryl group closer to the oxadiazine ring (7e), we saw much improved binding activity and selectivity as compared to the primary alcohol 7a. The hydroxyl group did not appear to be crucial for activity. Compound 7f retained similar potency and selectivity. When the methylene group of 7f was eliminated, a slightly decreased activity was observed (7g). Aryl modifications such as naphthyl (7h) were tolerated and retained good activity and selectivity. Further improvement of potency was achieved by introduction of the trifluorophenyl group, and compound 7i showed very good Aβ42 activity and good selectivity. With this improved in vitro profile, 7i was further studied in in vivo, PK, and ancillary profile evaluations. Compound 7i displayed 45% inhibition of Aβ42 in the rat cerebrospinal fluid (CSF) at the 3 h time point following acute oral administration at the 10 mg/kg dose. An oral rat PK study at a dose of 10 mpk showed high exposure with a plasma AUC of 24300 nM h, substantial brain concentration (1885 ng/g) at 6 h, and a brain/plasma ratio of 1.0. It did not inhibit CYP P450s or induce PXR in in vitro studies; however, 83% inhibition of hERG was observed in a function assay at a concentration of 10 μM. In an attempt to improve the hERG profile, we introduced polar groups such as heteroaryl substitutions at the C3 position. Compounds 7j and 7k showed much reduced Aβ42 activity. A carbonyl substitution at C4 (7l) also resulted in decreased activity.

Table 1. SAR Studies of C3-Monosubstituted Oxadiazinesa.

graphic file with name ml-2012-00209g_0005.jpg

graphic file with name ml-2012-00209g_0006.jpg

a

Each IC50 value is an average of at least two determinations. Racemic at C3 except 7i, which was the S-isomer as prepared from chiral starting material.

To further improve the in vivo activity and overall profile of 7i, we decided to revisit substitutions at the C4 position by systematically focusing on monosubstitution.19 As summarized in Table 2, the absence of substitution at C3 and C4 (8a) resulted in loss of activity. The carbonyl group at C4 did not improve activity (8b). However, when small alkyl groups such as isopropyl (8c) were introduced, improved activity was observed. Steric bulk was tolerated as both isobutyl and tert-butyl substitutions gave compounds 8d and 8e with good activity and reasonable selectivity. Tetrahydropyran-substituted compound 8f showed decreased activity, which might be due to the introduction of the polar oxygen atom. A similar SAR trend was observed with C3 modification. The hydroxyl group (8g) did not improve activity, but benzyl ether (8h), aryl-substituted secondary alcohol (8i), and simple benzyl substitution (8j) showed improved activity. To modify the physical properties of these compounds, polar groups were introduced at C4 position to decrease the clog D value. However, all of these changes resulted in much decreased activity [thiozole 8k (6400 nM), sulfone 8l (19000 nM), amide 8m (820 nM), morpholine 8n (3700 nM), and reverse amide 8o (1500 nM)]. On the other hand, when aryl substitutions were introduced, consistently good activity and selectivity (8pt) were achieved. In particular, the introduction of halogens to the aryl groups further improved the binding activity and Aβtotal to Aβ42 selectity (8qt). To differentiate these compounds for further profiling, they were characterized in PK and in vivo studies. As summarized in Table 3, compounds 8r and 8s showed highly potent in vivo activity with both compounds resulting in 62% reduction of CSF Aβ42 following oral dosing of 10 mg/kg. On the basis of the correlation between the decreased in vivo activity of compounds 8p and 8t and their lower brain exposure at the 6 h time point in rapid rat PK studies and at the 3 h time point in the PK/PD studies (for 8p), sufficient brain exposure seemed important for improved CSF activity. Because of its potent in vitro and in vivo activity and excellent rat PK profile (bioavailability, 100%; T1/2, 3.5 h; Vdss, 0.4 L/kg; and CL, 1.4 mL min–1 kg–1), compound 8s was further profiled. This compound was found to dose dependently reduce Aβ42 preferentially over Aβtotal and Aβ40 upon acute oral administration to rats,17 thus confirming modulator activity in vivo. Compound 8s displayed pKa values of 7.4 and 2.7, indicating that the oxadiazine ring was modestly basic, which conferred to a highly desirable aqueous solubility of the HCl salt of >100 mM at pH 3.5. Compound 8s was highly permeable in a Caco-2 membrane permeability assay and did not appear to be a substrate for PGP efflux. Even though a 76% inhibition of hERG activity was observed at 10 μM, no affect on QTc intervals in dogs at sufficient exposure multiples over exposures required for pharmacological effects were observed (data not shown). Compound 8s did not affect Notch processing in HEK293 cells expressing the human Notch1 protein at concentrations up to 50 μM. There was no evidence of Notch-related side effects or biomarker changes following repeated administration of the highest tolerable doses of 8s to rats.17

Table 2. SAR Studies of C4-Monosubstituted Oxadiazinesa.

graphic file with name ml-2012-00209g_0007.jpg

graphic file with name ml-2012-00209g_0008.jpg

a

Each IC50 value is an average of at least two determinations. The stereochemistry of the more active enantiomer is as shown in the table. The absolute stereochemistry was confirmed by synthesis from chiral starting material.

Table 3. Rat PK Profile20 and in Vivo Activity of Compounds 8pt.

compd AUC0–6ha brain concnb B/P ratioc CSF Aβ42 reductiond (%) brain levele plasma levele
8p 17620 539 0.9 39 2.8 6.2
8q 86460 2846 0.8 53 7.7 6.7
8r 24580 1340 1.1 62 NDf 7.2
8s 87410 2907 0.4 62 NDf 10.7
8t 22680 268 1.1 26 NDf 6.3
a

nM h with 10 mpk, po dosing.

b

ng/g at 6 h from rapid rat PK studies.

c

At 6 h from rapid rat PK studies.

d

Average of multiple testings at the 3 h time point with 10 mpk acute oral dosing in PD studies.

e

μM, average of multiple testings at the 3 h time point with 10 mpk acute oral dosing in PD studies.

f

Not determined.

To further improve the overall profile of this series of compounds, we also prepared fused morpholine oxadiazines (912) by replacing the C8 carbon with an oxygen atom. This modification could block potential metabolism at C8 and, by modifying the overall electronic proterties of the molecule, might also offer advantageous properties. On the basis of SAR studies of the morpholine oxadiazoline series,21 the C7 methyl group was installed to further improve activity. As shown in Figure 4, difluorophenyl derivatives (9 and 10) showed comparable in vitro activity to the trifluorophenyl analogs (11 and 12). In both cases, the (4S,7S)-isomers had better Aβ42 inhibition and Aβtotal/Aβ42 selectivity than the (4S,7R)-isomers.22 Compound 12 represented one of the most potent and selective GSMs that we identified. This compound displayed excellent in vivo activity, reducing Aβ42 in rat (60% reduction of CSF Aβ42 at 3 h with 10 mpk acute oral dosing), and provided opportunities for future SAR development.

Figure 4.

Figure 4

Identification of highly potent and selective fused morpholine oxadiazine GSMs. Each IC50 value is an average of at least two determinations.

In summary, in an effort to identify GSMs to treat AD, we discovered a series of fused oxadiazines (3) as selective and orally bioavailable GSMs based on the structural framework of oxadiazolines GSMs. Although structurally related, initial studies showed that SAR did not translate from the oxadiazoline to the oxadiazine series. Subsequently, we focused our SAR studies on modifications at the C3 and C4 positions of the fused oxadiazine core and identified GSMs such as compounds 8r and 8s that were highly efficacious in vitro and in vivo in a number of animal models with highly desirable physical and physicological proterties. Further improvement of in vitro activity and selectivity was achieved by the preparation of fused morpholine oxadiazines, which provided opportunities for future SAR development. The shift in specificity of APP cleavage rather than a reduction in overall γ-secretase activity and the resulting lack of changes in substrate accumulation and Notch processing as observed in the animal studies of compound 8s confirm that the oxadiazine series of compounds are potent GSMs. Further exploration of these compounds as potential drugs to treat AD is underway and will be the subject of future communications.

Acknowledgments

We thank Dr. Andrew Stamford for comments on the preparation of the manuscript. We thank Dr. Eric Parker for his strong support of the program. We thank Dr. Li-Kang Zhang for technical support.

Supporting Information Available

Biological assay protocols, general experimental descriptions and procedures, and characterization of final compounds. This material is available free of charge via the Internet at http://pubs.acs.org.

The authors declare no competing financial interest.

Supplementary Material

ml300209g_si_001.pdf (162.6KB, pdf)

References

  1. Wimo A.; Prince M.. World Alzheimer Report 2010: The Global Economic Impact of Dementia; Alzheimer's Disease International (ADI): London, United Kingdom, September, 2010. [Google Scholar]
  2. Avila J. Tau phosphorylation and aggregation in Alzheimer's disease pathology. FEBS Lett. 2006, 580, 2922–2927. [DOI] [PubMed] [Google Scholar]
  3. Hardy J. A.; Selkoe D. J. The amyloid hypothesis of Alzheimer's disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [DOI] [PubMed] [Google Scholar]
  4. Goate A.; Hardy J. Twenty years of Alzheimer's diseasecausing mutations. J. Neurochem. 2012, 120, 3–8. [DOI] [PubMed] [Google Scholar]
  5. Kung H. F. The β-amyloid hypothesis in Alzheimer's disease: Seeing is believing. ACS Med. Chem. Lett. 2012, 3, 265–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Zhang H.; Ma Q.; Zhang Y.-w.; Xu H. Proteolytic processing of Alzheimer's β-amyloid precursor protein. J. Neurochem. 2012, 120, 9–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Oehlrich D.; Berthelot D. J.-C.; Gijsen H. J. M. γ-Secretase modulators as potential disease modifying anti-Alzheimer's drugs. J. Med. Chem. 2011, 54, 669–698. [DOI] [PubMed] [Google Scholar]
  8. Josien H. Recent advances in the development of γ-secretase inhibitors. Curr. Opin. Drug Discovery Dev. 2002, 5, 513–525. [PubMed] [Google Scholar]
  9. Harrison T.; Churcher I.; Beher D. γ-Secretase as a target for drug intervention in Alzheimer's disease. Curr. Opin. Drug Discovery Dev. 2004, 7, 709–719. [PubMed] [Google Scholar]
  10. Wu W.; Zhang L. γ-Secretase inhibitors for the treatment of Alzheimer's disease. Drug Dev. Res. 2009, 70, 94–100. [Google Scholar]
  11. Artavanis-Tsakonas S.; Rand M. D.; Lake R. J. Notch signaling: cell fate control and signal integration in development. Science 1999, 284, 770–776. [DOI] [PubMed] [Google Scholar]
  12. Joo Y.; Kim H.-S.; Woo R.-S.; Park C. H.; Shin K.-Y.; Lee J.-P.; Chang K.-A.; Kim S.; Suh Y.-H. Mefenamic acid shows neuroprotective effects and improves cognitive impairment in in vitro and in vivo Alzheimer's disease models. Mol. Pharmacol. 2006, 69, 76–84. [DOI] [PubMed] [Google Scholar]
  13. Davis K. L. NSAID and Alzheimer's disease; possible answers and new questions. Mol. Psychiatry 2002, 7, 925–926. [DOI] [PubMed] [Google Scholar]
  14. Huang X.; Aslanian R.; Zhou W.; Zhu X.; Qin J.; Greenlee W.; Zhu Z.; Zhang L.; Hyde L.; Chu I.; Cohen-Williams M.; Palani A. The discovery of pyridone and pyridazone heterocycles as γ-secretase modulators. ACS Med. Chem. Lett. 2010, 1, 184–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Qin J.; Dhondi P.; Huang X.; Mandal M.; Zhao Z.; Pissarnitski D.; Zhou W.; Aslanian R.; Zhu Z.; Greenlee W.; Clader J.; Zhang L.; Cohen-Williams M.; Jones N.; Hyde L.; Palani A. Discovery of fused 5,6-bicyclic heterocycles as γ-secretase modulators. Bioorg. Med. Chem. Lett. 2011, 21, 664–669. [DOI] [PubMed] [Google Scholar]
  16. Qin J.; Zhou W.; Huang X.; Dhondi P.; Palani A.; Aslanian R.; Zhu Z.; Greenlee W.; Cohen-Williams M.; Jones N.; Hyde L.; Zhang L. Discovery of a potent pyrazolopyridine series of γ-secretase modulators. ACS Med. Chem. Lett. 2011, 2, 471–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Sun Z.-Y.; Asberom T.; Bara T.; Bennett C.; Burnett D.; Chu I.; Clader J.; Cohen-Williams M.; Cole D.; Czarniecki M.; Durkin J.; Gallo G.; Greenlee W.; Josien H.; Huang X.; Hyde L.; Jones N.; Kazakevich I.; Li H.; Liu X.; Lee J.; MacCoss M.; Mandal M. B.; McCracken T.; Nomeir A.; Mazzola R.; Palani A.; Parker E. M.; Pissarnitski D. A.; Qin J.; Song L.; Terracina G.; Vicarel M.; Voigt J.; Xu R.; Zhang L.; Zhang Q.; Zhao Z.; Zhu X.; Zhu Z. Cyclic hydroxyamidines as amide isosteres: discovery of oxadiazolines and oxadiazines as potent and highly efficacious γ-secretase modulators in vivo. J. Med. Chem. 2012, 55, 489–502. [DOI] [PubMed] [Google Scholar]
  18. Caldwell J. P.; Bennett C. E.; McCracken T. M.; Mazzola R. D.; Bara T.; Buevich A. V.; Burnett D. A.; Chu I.; Cohen-Williams M.; Jones N. T.; Josien H.; Hyde L. A.; Lee J.; McKittrick B.; Song L.; Terracina G.; Voigt J. H.; Zhang L.; Zhu Z. Iminoheterocycles as gamma-secretase modulators. Bioorg. Med. Chem. Lett. 2010, 20, 5380–5384. [DOI] [PubMed] [Google Scholar]
  19. Huang X.; Zhu X.; Chen X.; Zhou W.; Xiao D.; Degrado S.; Aslanian R.; Fossetta J.; Lundell D.; Tian F.; Trivedi P.; Palani A. A three-step protocol for lead optimization: Quick identification of key conformational features and functional groups in the SAR studies of non-ATP competitive MK2 (MAPKAPK2) inhibitors. Bioorg. Med. Chem. Lett. 2012, 22, 65–70. [DOI] [PubMed] [Google Scholar]
  20. Korfmacher W. A.; Cox K. A.; Ng K. J.; Veals J.; Hsieh Y.; Wainhaus S.; Broske L.; Prelusky D.; Nomeir A.; White R. E. Cassette-accelerated rapid rat screen: a systematic procedure for the dosing and liquid chromatography/atmospheric pressure ionization tandem mass spectrometric analysis of new chemical entities as part of new drug discovery. Rapid Commun. Mass Spectrom. 2001, 15, 335–340. [DOI] [PubMed] [Google Scholar]
  21. Huang X.; Pissarnitski D.; Li H.; Asberom T.; Josien H.; Zhu X.; Vicarel M.; Zhao Z.; Rajagopalan M.; Palani A.; Aslanian R.; Zhu Z.; Greenlee W.; Buevich A. Efficient synthesis and reaction pathway studies of novel fused morpholine oxadiazolines for use as gamma secretase modulators. Tetrahedron Lett. 2012, 10.1016/j.tetlet.2012.09.070. [DOI] [Google Scholar]
  22. Stereochemistry was assigned by NMR studies (chemical shift) and in vitro activity comparisons to the oxadiazoline analogues.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ml300209g_si_001.pdf (162.6KB, pdf)

Articles from ACS Medicinal Chemistry Letters are provided here courtesy of American Chemical Society

RESOURCES