Accepted Manuscript Benzoxazoles and Oxazolopyridines in Medicinal Chemistry Studies Charles S. Demmer , Lennart Bunch PII:

S0223-5234(14)01106-4

DOI:

10.1016/j.ejmech.2014.11.064

Reference:

EJMECH 7557

To appear in:

European Journal of Medicinal Chemistry

Received Date: 22 September 2014 Revised Date:

14 November 2014

Accepted Date: 30 November 2014

Please cite this article as: C.S. Demmer, L. Bunch, Benzoxazoles and Oxazolopyridines in Medicinal Chemistry Studies, European Journal of Medicinal Chemistry (2015), doi: 10.1016/ j.ejmech.2014.11.064. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Graphical Abstract (for review) Click here to download high resolution image

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

*Highlights (for review)

ACCEPTED MANUSCRIPT Highlights

EP

TE D

M AN U

SC

RI PT

Crystal structure data and interaction points of benzoxazoles Metabolism of benzoxazoles Medicinal chemistry studies of oxazolopyridines

AC C

  

ACCEPTED MANUSCRIPT

Benzoxazoles and Oxazolopyridines in Medicinal

RI PT

Chemistry Studies

Charles S. Demmer and Lennart Bunch*

SC

Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences,

*

TE D

M AN U

University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark

To whom correspondence should be addressed. (LB), Phone: +45 35336244. Fax: +45

AC C

EP

35336041. E-mail: [email protected]

1

ACCEPTED MANUSCRIPT

Abstract The benzoxazole heterocycle is often found in ligands targeting a plethora of receptors and enzymes. By analysis of published X-ray structures, this review aims at highlighting key

RI PT

interactions which the benzoxazole may engage in with its host protein. Furthermore, bioavailability, metabolism and the use of benzoxazole as a bioisostere are discussed. The review is extended to cover structure-activity relationship studies of 2-substituted benzoxazoles, 2-

SC

substituted oxazolopyridines, and in perspective, application of the recently published novel

M AN U

heterocycle oxazolopyrazine in medicinal chemistry studies.

Keywords Drug Design Heterocycle

Oxazolopyrazine

1. Introduction

EP

Oxazolopyridine

TE D

Benzoxazole

AC C

Planar heterocycles define an important class of chemical entities in life science research.

They are often incorporated as building blocks in medicinal chemistry studies with the purpose of modulating a ligand’s affinity and/or selectivity towards a biological target. Furthermore, they are applied in several other fields of research such as material science and catalysis.

2

ACCEPTED MANUSCRIPT

Amongst the class of planar heterocycles, the benzoxazole parental skeleton is found. The scaffold is a constituent of several natural products (Figure 1) and often incorporated in drug design. In this review, we are will focus on 2-substituted benzoxazoles and their corresponding

RI PT

nitrogen analogs (oxazolopyridines) in medicinal chemistry studies. As represented in Figure 2, an increasing number of 2-substituted benzoxazoles have been characterized pharmacologically from 1990 until today. In total, 6418 2-substituted benzoxazoles have been investigated as

SC

ligands for one or more biological targets, while only 1211 of its nitrogen analogs (the four oxazolopyridines, entry 2-5, Table 1) have been explored. Examples of the use of 2-substituted

M AN U

benzoxazoles cover a wide range of research fields, such as antibacterial[1]–[3], antimicrobial[4], antiviral[5], antifungal[3], [6] or antiproliferative[7], [8] activities.

AC C

EP

Boxazomycins A[11]

TE D

Figure 1. Examples of natural products comprising a benzoxazole heterocycle: Calcimycin[9], Nakijinol[10] and

3

ACCEPTED MANUSCRIPT

Figure 2. Total number of instances per year of 2substituted benzoxazoles characterized on biological targets

RI PT

from 1960 to 2013.

1000 900 800 700

SC

600 500

M AN U

400 300 200 100

TE D

1962 1965 1968 1971 1974 1977 1980 1983 1986 1989 1992 1995 1998 2001 2004 2007 2010 2013

0

Table 1. Number of biologically active 2-substituted

including hydrogena

1 2 3

a

Structure

AC C

Entry

EP

benzoxazoles and its nitrogen analogs. R= any group

Number of Appearance 6418 733 275

4

125

5

78

Reaxys search performed the 2nd of August 2014.

4

ACCEPTED MANUSCRIPT

2. Binding mode of ligands comprising a benzoxazole The benzoxazole scaffold may engage in a number of distinct energetically favorable interactions with its host protein. Both the 1-oxygen as well as the 3-nitrogen atoms can act as

RI PT

hydrogen bond acceptors, and due to its aromatic planar nature both pi-pi stacking and pi-cation interactions are possible. Finally, due to its lipophilic character, hydrophobic interactions with its host protein are possible. We commenced the study by investigating all available x-ray structures

SC

of 2-substituted benzoxazoles in their respective host proteins. In all five analogs, compounds 1-

M AN U

5 (Figure 3) are available from the protein data bank (Figure 4-8).

Figure 3. Biologically active molecules which comprises a

2-substituted benzoxazole or a 2-substituted oxazolo[4,5-

AC C

EP

TE D

b]pyridine moiety crystallized in receptors or enzymes

5

ACCEPTED MANUSCRIPT

The protein tyrosine phosphatase (PTP) belongs to a family of enzymes that catalyzes the hydrolysis of phosphorylated tyrosine residues. Amongst them, protein tyrosine phosphatase 1B (PTP1B) is of profound biological importance as it is considered as a negative regulator for

RI PT

insulin and leptin.[12] Hence, an important class of drugs for the treatment of type II diabetes and obesity targets this enzyme.[12] Compound 1 is an example of a PTP1B-inhibitor, which comprises a benzoxazole (Figure 3) and inhibits the PTP1B enzyme in the mid-micromolar

SC

range. A 2D-representation of its binding mode and key interactions with its host protein is depicted in Figure 4.[13] Although the acidic functional groups of 1 engage in extensive

M AN U

hydrogen bonding interactions with the polar side chains and backbone of PTP1B, the benzoxazole moiety is surrounded by non-polar amino acids (Figure 4). Not surprisingly, hydrophobic interactions are predominant interactions between the phenyl ring of the benzoxazole and Gly259. Moreover, the crystal structure also reveals that the benzoxazole C4-

AC C

EP

TE D

carbon engaged in a weak hydrogen bonding interaction with the backbone carbonyl of Val49.

6

ACCEPTED MANUSCRIPT

Figure 4. Crystal structure of compound 1 with PTP1B

TE D

M AN U

SC

RI PT

(PDB code: 4I8N)

A second example of a crystallized protein-ligand complex is compound 2, which is an inhibitor of the channel-activating protease prostasin (CAP1/PRSS8).[14] This enzyme is a key

EP

player in the severe kidney disease Liddle’s syndrome, which is characterized by malfunction of an epithelial sodium channel (ENaC) due to a genetic mutation. In humans, prostasin is thought

AC C

to be a potential regulator of ENaC function. Compound 2 was successfully crystallized in CAP1/PRSS8 (Figure 5): The nitrogen atom of the oxazole moiety engages in two hydrogen bonds with His57B and Ser195B. Due to the polar receptor residues located in the vicinity of the benzoxazole, no hydrophobic interactions are observed. Moreover, the crystal structure also reveals the presence of explicit water molecules, which would make it interesting to investigate the use of nitrogen analogs of the benzoxazole (oxazolopyridines). On another hand, disruption

7

ACCEPTED MANUSCRIPT

of the water matrix may result in loss of affinity due to the actual hydrogen bonds between the

AC C

EP

TE D

M AN U

SC

Figure 5. Crystal structure of compound 2 with CAP1/PRSS8 (pdb code: 3E0P)

RI PT

water molecules (in particular HOH453) and the ligand (Figure 5).

8

ACCEPTED MANUSCRIPT

Transthyretin (TTR) is a protein composed of four distinct subunits and functions as a serum carrier of retinol-binding protein and thyroxine.[15] It is secreted by the liver into the blood stream and therefrom further transported into the cerebrospinal fluid. Protein malfunction or

RI PT

mutations within the TTR gene can lead to life-threatening diseases such as transthyretin familial amyloid neuropathy (TTR FAP).[16] A family of 2-substituted benzoxazoles was found to be potent and selective inhibitors of TTR.[17] A hydrogen bond is observed between the phenolic

SC

alcohol and the SerA117 side chain. Besides from this hydrophobic interactions are predominant,

M AN U

involving LysA15, and AlaA108 residues.

Figure 6. X-Ray crystal structure of compound 3 with

AC C

EP

TE D

homotetrameric WT-TTR (pdb code: 2QGC)

9

ACCEPTED MANUSCRIPT

Tuberculosis (TB) is a life-threatening disease caused by the bacteria Mycobacterium tuberculosis. Most commonly affected are the lungs. Hence TB is transmitted person to person via air posing a serious health problem. The prevalence is high in poor regions of Africa and

RI PT

death numbers are high, due to the lack of treatments.[18] In the year 2010, 1.20–1.45 million people died from TB – the majority from developing countries.[18], [19] Moreover, due to multidrug-resistant tuberculosis (MDR-TB) and extensively-drug-resistant TB (XDR-TB), the

SC

development of new treatments is essential.[20] Compound 4, a low-nanomolar affinity peptide deformylase inhibitor of Mycobacterium tuberculosis, was reported in 2008 and its crystal

M AN U

structure depicted in Figure 7. The crystal structure is a trimer in which the benzoxazole moiety is directed out of the active site. As shown in Figure 7, hydrophobic interactions are observed within a range of 4Ǻ in particular with Leu141 (3.6-3.7 Ǻ) and Val50 (3.6 Ǻ). A hydrogen bond between the nitrogen atom of the benzoxazole and HOH208 (3.0 Ǻ) is also present. Moreover,

TE D

the presence of other water molecules around the phenyl ring might make this inhibitor an interesting target compound for the exploration of oxazolopyridines as isosteres for

AC C

EP

benzoxazoles.

10

ACCEPTED MANUSCRIPT

Figure 7. X-Ray crystal structure of 4 bound to Ni–PDF of

M AN U

SC

RI PT

M. tuberculosis (pdb code: 3E3U)

TE D

The human immunodeficiency virus (HIV) is a retrovirus which infiltrates the immune system eventually leading to its collapse. Todays’ standard treatment is to suppress virus replication by administration of a combination of at least three drugs often referred to “highly active

EP

antiretroviral therapy” (HAART). A newly synthesized HIV-1 reverse transcriptase inhibitor comprises a benzoxazole heterocycle (compound 5).[21] The inhibitor was co-crystallized with

AC C

two HIV-1 reverse transcriptase mutants (Figure 8). Noteworthy is, that the mutated residues do not engage in direct interactions with the inhibitor (>5 Ǻ). This is an important feature as to avoid loss of drug efficacy as the HIV virus mutates. The HIV inhibitor 5 is buried inside the HIV-1 reverse transcriptase mutants and adopts a butterfly-like conformation.[21] The benzoxazole moiety engages in π-π stacking interactions with both Tyr188 and Tyr181 (dist. 3.63.9 Ǻ, not shown) and moreover, the crystal structure reveals H-π bond interactions with Trp229

11

ACCEPTED MANUSCRIPT

(dist. 3.8 Ǻ, not shown). Classic hydrophobic interactions are present between the side chains of Leu234, Leu100 and Pro95 and the phenyl ring of the heterocycle (dist. 3.7-3.8 Ǻ, not shown). A

oxazolo ring component of the benzoxazole (3.0 Ǻ).

AC C

EP

TE D

M AN U

SC

Figure 8. X-Ray crystal structure of 5 (pdb code: 2YKN)

RI PT

water molecule (HOH2162) is making a hydrogen interaction with the oxygen atom of the

Hepatitis is a serious medical condition defined by the inflammation of the liver tissues.

Three different types exist, named hepatitis A, B and C. While vaccines exist for hepatitis A and B, one against the hepatitis C virus (HCV) remains to be developed.[22] Approximately 3% of the world’s population (170 million people) is infected by the HCV.[23] Non-structural proteins 5 (NS5) are viral proteins which are found in the hepatitis C virus[24] and are involved in the replication of the virus RNA. They are divided into two subtype families, NS5A and NS5B. With 12

ACCEPTED MANUSCRIPT

the objective of finding new treatments, NS5A and NS5B were pointed out as potential therapeutic targets.[23] Recently, a new quinazolinone-based compound 6 that comprises a oxazolo[4,5-b]pyridine moiety was found to be an inhibitor with low nanomolar efficacy.[25]

following confirmed by X-ray structure (Figure 9).

RI PT

The binding mode of compound 6 was first predicted by molecular dynamic protocols and

The crystal structure of the inhibitor 6 with NS5B reveals that the oxazolopyridine

SC

heterocycle is surrounded by the hydrophobic amino acids Val494, Pro495 and Leu497. Favorable non-conventional hydrogen bonds are observed between the backbone carbonyl of

M AN U

Val494 (not shown) and the hydrogen in position 6 of the pyridine ring, and between the side chain of Pro495 and the hydrogen of position 7 (not shown). The two interactions are favorable due to the electronegative character of the nitrogen and oxygen atoms of the oxazolo[4,5-

TE D

b]pyridine heterocycle, which results in a polarization of the C-H bonds of the pyridine ring.

AC C

EP

Figure 9. X-Ray crystal structure of 6 (pdb code: 4JUI)[25]

13

ACCEPTED MANUSCRIPT

3. Bioavailability and metabolism of the benzoxazole heterocycle Looking beyond a ligand’s target affinity, efficacy and selectivity profile, its pharmacokinetic properties are parameters also to be studied and mastered in a successful drug

RI PT

discovery program. A detailed study of the metabolism of benzoxazole, 2-methyl benzoxazole and 2-phenyl benzoxazole in rabbits has been conducted (Figure 10).[26] Two groups of metabolites from the benzoxazole and/or the 2-methyl benzoxazole were identified in the rabbits’

SC

urine. The first originates from the hydrolysis of the oxazolo ring (compounds 7-12). Not surprisingly, phenylglucuronides (compounds 13-15) are also observed as metabolites and

M AN U

originate from the glucuronidation of the o-aminophenol. This process assists in the excretion of toxic substances in animals. The second group stems from the oxidation of the benzoxazole (compounds 16 and 17).

On the other hand, when the rabbits were fed with 2-phenyl benzoxazole, this heterocycle was

TE D

not subject to hydrolysis. Only a hydroxy-2-phenylbenzoxazole metabolite could be extracted from the urine, which was thought to be 3-(benzo[d]oxazol-2-yl)phenol (not shown).

AC C

EP

Figure 10. Observed metabolites benzoxazole and/or 2-methyl benzoxazole administered per oral in rabbit.[26]

14

ACCEPTED MANUSCRIPT

In another study, a set of heterocycles were investigated as potential substrates of rabbit liver aldehyde oxidase (Table 2).[27] Amongst the pool of heterocycles, the quinazoline was found to be a good substrate with a Km value in the low micromolar range. The benzoxazole and

RI PT

1,2-benzisoxazole were both oxidized at much lower rates. Due to the fact that 2-chloro and 2-

oxidation takes place at the 2-position.

Table 2. Comparison of kinetic data between heterocycles

1

Quinozaline (18)

2

Benzoxazole (19)

3

1,2-Benzisoxazole (20)

5

2-Cl-benzoxazole (21) 2-Me-benzoxazole (22)

(mM)

oxidation rate

0.017

100

2.0

1.5

--

0.3

b

--

b

--

NA NA

EP

4 a

Compound

Relative

TE D

Entry

Km

M AN U

as substrates of aldehyde oxidasea

SC

methyl benzoxazoles are not substrates for the enzyme, it is reasonable to assume that the

Initial rates were measured at 30°C and pH 6-8. Relative

oxidation rates were measured with a concentration of 0.2

AC C

mM test compound. b No activity.

4. Serving as a bioisostere

The benzoxazole can be regarded as a conformationally restricted N-aryl amide - a functionality which is often incorporated in drugs. Substitution for a benzoxazole provides a conformationally restricted analog which is a well-known strategy to improve ligand affinity

15

ACCEPTED MANUSCRIPT

(reduction in conformational entropic loss). The pharmacokinetic properties of the ligand are also modified and hence this is also a strategy for overcoming problems encountered with bioavailability and/or metabolism.[28] It is indeed true that the benzoxazole cannot engage in

RI PT

hydrogen donor activities with the host protein. Thus the strategy of substituting an N-aryl amide for a benzoxazole should only be attempted if it is the hydrogen bond accepting features of the N-aryl amide which is in play. Following this concept the benzoxazole scaffold also offers

SC

metabolically stable, conformationally restricted analogs of N/O-aryl carbamates, N-aryl urea, and O-aryl esters. The latter functionality is rarely incorporated in drugs due to the fast

M AN U

hydrolysis of the aryl ester functionality. Noteworthy is therefore, that the 2-ethoxybenzoxazole proved to be a successful bioisostere of an ethyl benzoate and thus represents an approach to overcome the hydrolysis issue.[29]

chemistry studies

TE D

5. Incorporation of a nitrogen atom into the benzoxazole core structure in medicinal

The replacement with a nitrogen atom(s) for a carbon atom(s) in a heterocycle is an

EP

attractive strategy in drug discovery for a number of reasons. Firstly, the enhanced polarity will affect the adsorption of the compound and this is at the center of attention for the medicinal

AC C

chemists during the design phase. Secondly, the introduced nitrogen atoms can engage in new hydrogen bond with either a polar receptor residue or a water molecule if present. This may again lead to improved target selectivity. In the following examples of structure-activity relationship studies, the effects of the replacement of the benzoxazole heterocycle by oxazolopyridines are discussed.

16

ACCEPTED MANUSCRIPT

5.1 Example of a positive effect: The fatty acid amide hydrolase (FAAH) inhibitors The endocannabinoid system is defined by a group of neuromodulatory lipids and their receptors, which are found in the brain. It is involved in a wide range of physiological processes

RI PT

such as appetite, mood and memory.[30] A breakthrough in its understanding was the isolation of the endogenous fatty acid amides (FAA) named Anamandine.[31] Since then, the enzyme fatty acid amide hydrolase (FAAH) has been a potential therapeutic target for inflammatory pain

SC

and anxiety.[32], [33] With the objective of discovering new FAAH inhibitors, a set of compounds which comprises the benzoxazole heterocycle and all possible oxazolopyridines were

M AN U

synthesized (Table 3). In summary, it was shown that the incorporation of a nitrogen into the benzoxazole core structure resulted in a significant increase of affinity irrespective of the nitrogen atom position (50–200 fold). These results are intriguing as the position of the nitrogen

TE D

does not seem to be critically important for the improvement in affinity.

oxazolopyridines[34]

Compound

1

23

2 3 4 5

R

AC C

Entry

EP

Table 3. Inhibitors of FAAH comprising benzoxazole and

Ki, nM 370 ± 130

24

2.3 ± 0.1

25

7.2 ± 1.6

26

3.7 ± 1.0

27

11 ± 4.0

17

ACCEPTED MANUSCRIPT

A previous study showed that 3-substituted phenyl propylcarbamates are also FAAH inhibitors. As a part of the structure-activity relationship study, the affinity of a benzoxazole derivate and its nitrogen analogs were compared (Table 4). Accordingly, the use of an

nitrogen atoms resulted in no significant change (Table 4).

SC

Table 4. 3-Substitued phenyl propylcarbamate as FAAH

FAAH IC50a (µM)

Compd

R

1

28

3.0 ± 0.6

2

29

0.68 ± 0.09

3

30

MGL, % of inhibitionb 26 29

TE D

Entry

M AN U

inhibitors[35]

a

RI PT

oxazolopyridine increased the inhibitory activity by 4-5 fold, while the incorporation of two

4.5 ± 0.6

18c

Values represent the mean of three independent

EP

experiments (n=3) performed in duplicate; b Inhibition of enzymatic activity (% of control) at 100 µM (n=2);

c

Lipase.

AC C

Inhibition of human recombinant. MGL: Monoglyceride

5.2 Negative effect

The disease, human African trypanosomiasis (HAT), also referred as sleeping sickness, is

caused by a parasite transported by the Glossina insect, the tsetse fly. The disease is most commonly observed in Equatorial Africa and especially in remote rural areas.[36] In the light of

18

ACCEPTED MANUSCRIPT

finding an irreversible inhibitor of a critical enzyme in the parasite, a preliminary study showed that chloronitrobenzamides exhibit low micromolar inhibitory activity against T. brucei brucei (EC50 =1.5 μM).[37], [38] A further structure-activity relationship led to the synthesis of

RI PT

benzoxazole 31, which displays nanomolar inhibitory activity. Interestingly, the use of the

10).[39]

Figure 10. Anti-trypanosomal Activity of 31 and 32 on T.

TE D

M AN U

brucei brucei

SC

oxazolo[4,5-b]pyridine heterocycle instead of the benzoxazole led to a loss of affinity (Figure

5.3 No effect

Leukotrienes are a family of inflammatory mediators biosynthesized in leukocytes from

EP

arachidonic acid by action of the enzyme 5-lipoxygenase (5-LO).[40] A series of aryldienylbenzoxazoles proved to be potent inhibitors of 5-LO (Figure 11, compound 33).[41]

AC C

Interestingly, the use of the oxazolopyridine heterocycle (compound 34) did not lead to an increase in affinity but was equipotent to compound 33. This suggests that the nitrogen atom from the pyridine does not engage in favorable or unfavorable interactions with the enzyme.

Figure 11. Inhibition of 5-lipoxygenase (5-LO) by benzoxazole derivates 33 and 34

19

RI PT

ACCEPTED MANUSCRIPT

6. Perspectives

SC

The benzoxazole and its four nitrogen analogs – the oxazolopyridines – find extensive use in drug discovery as they allow the medicinal chemist to fine-tune guest-host interactions and

M AN U

furthermore optimize the pharmacokinetic properties of the ligand. It is therefore of particular interest that a synthetic protocol for the oxazolopyrazine scaffold 35 was recently published by us.[42][22] The methodology is based on a robust palladium-catalyzed domino reaction between an amide and 2,3-dichloropyrazine (Scheme 1). An even more detailed fine-tuning of ligand

TE D

properties can now be explored by medicinal chemists.

Scheme 1. Newly developed synthetic route of the

AC C

EP

oxazolopyrazine scaffold (R= aryl)

7. Conclusion

The benzoxazole heterocycle is often incorporated in medicinal chemistry studies but

also found in natural products. By analysis of X-ray structures we have highlighted its potential interactions points with host proteins and discussed the substitution for its nitrogen analogs oxazolopyridines. Furthermore, we have addressed bioavailability/metabolism which highlights 20

ACCEPTED MANUSCRIPT

the 2-position as a hot spot, and finally its application as a biosisostere in medicinal chemistry studies.

RI PT

8. Acknowledgements

AC C

EP

TE D

M AN U

SC

The authors would like to thank the Lundbeck foundation for financial support.

21

ACCEPTED MANUSCRIPT

9. References [1] K. Wolfart, A. Molnar, M. Kawase, N. Motohashi, and J. Molnar, “Effects of trifluoromethyl ketones on the motility of Proteus vulgaris.,” Biol. & Pharm. Bull., vol. 27, no. 9,

[2]

RI PT

pp. 1462–1464, 2004. J. Alvesalo, H. Vuorela, P. Tammela, M. Leinonen, P. Saikku, and P. Vuorela,

“Inhibitory effect of dietary phenolic compounds on Chlamydia pneumoniae in cell cultures.,”

[3]

SC

Biochem. Pharmacol., vol. 71, no. 6, pp. 735–741, 2006.

I. Yildiz-Oren, I. Yalcin, E. Aki-Sener, and N. Ucarturk, “Synthesis and structure-activity

Chem., vol. 39, pp. 291–298, 2004. [4]

M AN U

relationships of new antimicrobial active multisubstituted benzazole derivatives.,” Eur. J. Med.

İ. Yalçin, İ. Ören, E. Şener, A. Akin, and N. Uçartürk, “The synthesis and the structure-

activity relationships of some substituted benzoxazoles, oxazolo(4,5-b)pyridines, benzothiazoles and benzimidazoles as antimicrobial agents,” Eur. J. Med. Chem., vol. 27, pp. 401–406, 1992. A. Akbay, I. Oren, O. Temiz-Arpaci, E. Aki-Sener, and I. Yalçin, “Synthesis and HIV-1

TE D

[5]

reverse transcriptase inhibitor activity of some 2,5,6-substituted benzoxazole, benzimidazole,

pp. 266–271, 2003.

C. Ramalingan, S. Balasubramanian, S. Kabilan, and M. Vasudevan, “Synthesis and

AC C

[6]

EP

benzothiazole and oxazolo(4,5-b)pyridine derivatives.,” Arzneimittel-Forschung, vol. 53, no. 4,

study of antibacterial and antifungal activities of novel 1-[2-(benzoxazol-2-yl)ethoxy]- 2,6diarylpiperidin-4-ones.,” Eur. J. Med. Chem., vol. 39, no. 6, pp. 527–533, 2004. [7]

G. Wells, J. M. Berry, T. D. Bradshaw, A. M. Burger, A. Seaton, B. Wang, A. D.

Westwell, and M. F. G. Stevens, “4-Substituted 4-hydroxycyclohexa-2,5-dien-1-ones with selective activities against colon and renal cancer cell lines.,” J. Med. Chem., vol. 46, no. 4, pp. 532–541, 2003. 22

ACCEPTED MANUSCRIPT

[8]

S. M. Courtney, P. A. Hay, R. T. Buck, C. S. Colville, D. W. Porter, D. I. C. Scopes, F.

C. Pollard, M. J. Page, J. M. Bennett, M. L. Hircock, E. A. McKenzie, C. R. Stubberfield, and P. R. Turner, “2,3-Dihydro-1,3-dioxo-1H-isoindole-5-carboxylic acid derivatives: a novel class of

RI PT

small molecule heparanase inhibitors.,” Bioorganic & Med. Chem. Lett., vol. 14, no. 12, pp. 3269–3273, 2004. [9]

M. O. Chaney, P. V. Demarco, N. D. Jones, and J. L. Occolowitz, “Letter: The structure

SC

of A23187, a divalent cation ionophore.,” J. Am. Chem. Soc., vol. 96, no. 6, pp. 1932–1933, 1974.

S. P. B. Ovenden, J. L. Nielson, C. H. Liptrot, R. H. Willis, D. M. Tapiolas, A. D.

M AN U

[10]

Wright, and C. A. Motti, “Sesquiterpene benzoxazoles and sesquiterpene quinones from the marine sponge Dactylospongia elegans.,” J. Nat. Prod., vol. 74, no. 1, pp. 65–68, 2011. [11]

T. Kusumi, T. Ooi, M. R. Walchli, and H. Kakisawa, “Structure of the novel antibiotics

[12]

TE D

boxazomycins A, B, and C,” J. Am. Chem. Soc., vol. 110, no. 9, pp. 2954–2958, Apr. 1988. C. D. Rochester and O. Akiyode, “Novel and emerging diabetes mellitus drug therapies

for the type 2 diabetes patient.,” World J. Diabetes, vol. 5, no. 3, pp. 305–315, 2014. A. P. Chandrasekharappa, S. E. Badiger, P. K. Dubey, S. K. Panigrahi, and S. R. V. V. V.

EP

[13]

Manukonda, “Design and synthesis of 2-substituted benzoxazoles as novel PTP1B inhibitors.,”

[14]

AC C

Bioorganic & Med. Chem. Lett., vol. 23, no. 9, pp. 2579–2584, 2013. D. C. Tully, A. Vidal, A. K. Chatterjee, J. A. Williams, M. J. Roberts, H. M. Petrassi, G.

Spraggon, B. Bursulaya, R. Pacoma, A. Shipway, A. M. Schumacher, H. Danahay, and J. L. Harris, “Discovery of inhibitors of the channel-activating protease prostasin (CAP1/PRSS8) utilizing structure-based design.,” Bioorganic & Med. Chem. Lett., vol. 18, no. 22, pp. 5895– 5899, 2008.

23

ACCEPTED MANUSCRIPT

[15]

J. A. Palha, “Transthyretin as a thyroid hormone carrier: function revisited.,” Clin. Chem.

Lab. Med. : CCLM / FESCC, vol. 40, no. 12, pp. 1292–1300, 2002. [16]

V.

and

Planté-Bordeneuve

P.

Kerschen,

“Transthyretin

familial

amyloid

[17]

S. M. Johnson, S. Connelly, I. A. Wilson, and J. W. Kelly, “Biochemical and structural

evaluation

of

highly

selective

2-arylbenzoxazole-based

transthyretin

C.

Williams,

“Global

Tuberculosis

Control:

SC

inhibitors.,” J. Med. Chem., vol. 51, no. 2, pp. 260–270, 2008. [18]

RI PT

polyneuropathy.,” Handb. Clin. Neurol., vol. 115, pp. 643–658, 2013.

WHO

Report

amyloidogenesis

2011.”

WILEY-

[19]

M AN U

BLACKWELL, 111 RIVER ST, HOBOKEN 07030-5774, NJ USA.

R. Lozano, M. Naghavi, K. Foreman, S. Lim, K. Shibuya, V. Aboyans, J. Abraham, T.

Adair, R. Aggarwal, S. Y. Ahn, M. Alvarado, H. R. Anderson, L. M. Anderson, K. G. Andrews, C. Atkinson, L. M. Baddour, S. Barker-Collo, D. H. Bartels, M. L. Bell, E. J. Benjamin, D.

TE D

Bennett, K. Bhalla, B. Bikbov, A. Bin Abdulhak, G. Birbeck, F. Blyth, I. Bolliger, S. Boufous, C. Bucello, M. Burch, P. Burney, J. Carapetis, H. Chen, D. Chou, S. S. Chugh, L. E. Coffeng, S. D. Colan, S. Colquhoun, K. E. Colson, J. Condon, M. D. Connor, L. T. Cooper, M. Corriere, M.

EP

Cortinovis, K. C. de Vaccaro, W. Couser, B. C. Cowie, M. H. Criqui, M. Cross, K. C. Dabhadkar, N. Dahodwala, D. De Leo, L. Degenhardt, A. Delossantos, J. Denenberg, D. C. Des

AC C

Jarlais, S. D. Dharmaratne, E. R. Dorsey, T. Driscoll, H. Duber, B. Ebel, P. J. Erwin, P. Espindola, M. Ezzati, V. Feigin, A. D. Flaxman, M. H. Forouzanfar, F. G. R. Fowkes, R. Franklin, M. Fransen, M. K. Freeman, S. E. Gabriel, E. Gakidou, F. Gaspari, R. F. Gillum, D. Gonzalez-Medina, Y. A. Halasa, D. Haring, J. E. Harrison, R. Havmoeller, R. J. Hay, B. Hoen, P. J. Hotez, D. Hoy, K. H. Jacobsen, S. L. James, R. Jasrasaria, S. Jayaraman, N. Johns, G. Karthikeyan, N. Kassebaum, A. Keren, J.-P. Khoo, L. M. Knowlton, O. Kobusingye, A.

24

ACCEPTED MANUSCRIPT

Koranteng, R. Krishnamurthi, M. Lipnick, S. E. Lipshultz, S. L. Ohno, J. Mabweijano, M. F. MacIntyre, L. Mallinger, L. March, G. B. Marks, R. Marks, A. Matsumori, R. Matzopoulos, B. M. Mayosi, J. H. McAnulty, M. M. McDermott, J. McGrath, G. A. Mensah, T. R. Merriman, C.

RI PT

Michaud, M. Miller, T. R. Miller, C. Mock, A. O. Mocumbi, A. A. Mokdad, A. Moran, K. Mulholland, M. N. Nair, L. Naldi, K. M. V. Narayan, K. Nasseri, P. Norman, M. O’Donnell, S. B. Omer, K. Ortblad, R. Osborne, D. Ozgediz, B. Pahari, J. D. Pandian, A. P. Rivero, R. P.

SC

Padilla, F. Perez-Ruiz, N. Perico, D. Phillips, K. Pierce, C. A. Pope, E. Porrini, F. Pourmalek, M. Raju, D. Ranganathan, J. T. Rehm, D. B. Rein, G. Remuzzi, F. P. Rivara, T. Roberts, F. R. De

M AN U

León, L. C. Rosenfeld, L. Rushton, R. L. Sacco, J. A. Salomon, U. Sampson, E. Sanman, D. C. Schwebel, M. Segui-Gomez, D. S. Shepard, D. Singh, J. Singleton, K. Sliwa, E. Smith, A. Steer, J. A. Taylor, B. Thomas, I. M. Tleyjeh, J. A. Towbin, T. Truelsen, E. A. Undurraga, N. Venketasubramanian, L. Vijayakumar, T. Vos, G. R. Wagner, M. Wang, W. Wang, K. Watt, M.

TE D

A. Weinstock, R. Weintraub, J. D. Wilkinson, A. D. Woolf, S. Wulf, P.-H. Yeh, P. Yip, A. Zabetian, Z.-J. Zheng, A. D. Lopez, C. J. L. Murray, M. A. AlMazroa, and Z. A. Memish, “Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a

EP

systematic analysis for the Global Burden of Disease Study 2010.,” Lancet, vol. 380, no. 9859, pp. 2095–2128, 2012.

G. L. Calligaro, L. Moodley, G. Symons, and K. Dheda, “The medical and surgical

AC C

[20]

treatment of drug-resistant tuberculosis.,” J. Thorac. Dis., vol. 6, no. 3, pp. 186–195, 2014. [21]

J. Boyer, E. Arnoult, M. Médebielle, J. Guillemont, J. Unge, and D. Jochmans,

“Difluoromethylbenzoxazole Pyrimidine Thioether Derivatives: A Novel Class of Potent NonNucleoside HIV-1 Reverse Transcriptase Inhibitors,” J. Med. Chem., vol. 54, no. 23, pp. 7974– 7985, Oct. 2011.

25

ACCEPTED MANUSCRIPT

[22]

C. I. Yu and B.-L. Chiang, “A new insight into hepatitis C vaccine development.,” J.

Biomed. & Biotechnol., vol. 2010, Jun. 2010. [23]

J. J. Kohler, J. H. Nettles, F. Amblard, S. J. Hurwitz, L. Bassit, R. A. Stanton, M.

inhibitors.,” Infect. drug Resist., vol. 7, pp. 41–56, Mar. 2014. [24]

RI PT

Ehteshami, and R. F. Schinazi, “Approaches to hepatitis C treatment and cure using NS5A

S. Gehring, S. H. Gregory, P. Wintermeyer, C. Aloman, and J. R. Wands, “Generation of

SC

immune responses against hepatitis C virus by dendritic cells containing NS5 protein-coated microparticles.,” Clin. vaccine Immunol. : CVI, vol. 16, no. 2, pp. 163–171, 2009. O. Hucke, R. Coulombe, P. Bonneau, M. Bertrand-Laperle, C. Brochu, J. Gillard, M.-A.

M AN U

[25]

Joly, S. Landry, O. Lepage, M. Llinàs-Brunet, M. Pesant, M. Poirier, M. Poirier, G. McKercher, M. Marquis, G. Kukolj, P. L. Beaulieu, and T. A. Stammers, “Molecular dynamics simulations and structure-based rational design lead to allosteric HCV NS5B polymerase thumb pocket 2

1943, 2014. [26]

TE D

inhibitor with picomolar cellular replicon potency.,” J. Med. Chem., vol. 57, no. 5, pp. 1932–

H. G. Bray, R. C. Clowes, and W. V. Thorpe, “The metabolism of aminophenols,

EP

oformamidophenol, benzoxazole, 2-methyl- and 2-phenyl-benzoxazoles and benzoxazolone in the rabbit.,” Biochem. J., vol. 51, pp. 70–78, Apr. 1952. W. Gristwood and K. Wilson, “Kinetics of some benzothiazoles, benzoxazoles, and

AC C

[27]

quinolines as substrates and inhibitors of rabbit liver aldehyde oxidase.,” Xenobiotica; fate Foreign Compd. Biol. Syst., vol. 18, no. 8, pp. 949–954, 1988. [28]

L. S. Lin, T. J. Lanza, L. A. Castonguay, T. Kamenecka, E. McCauley, G. Van Riper, L.

A. Egger, R. A. Mumford, X. Tong, M. MacCoss, J. A. Schmidt, and W. K. Hagmann, “Bioisosteric replacement of anilide with benzoxazole: potent and orally bioavailable antagonists

26

ACCEPTED MANUSCRIPT

of VLA-4.,” Bioorganic & Med. Chem. Lett., vol. 14, no. 9, pp. 2331–2334, 2004. [29]

R. N. Brown, R. Cameron, D. K. Chalmers, S. Hamilton, A. Luttick, G. Y. Krippner, D.

B. McConnell, R. Nearn, P. C. Stanislawski, S. P. Tucker, and K. G. Watson, “2-

RI PT

Ethoxybenzoxazole as a bioisosteric replacement of an ethyl benzoate group in a human rhinovirus (HRV) capsid binder.,” Bioorganic & Med. Chem. Lett., vol. 15, no. 8, pp. 2051– 2055, 2005.

E. Fride, “Endocannabinoids in the central nervous system: from neuronal networks to

SC

[30]

behavior.,” Curr. drug targets. CNS Neurol. Disord., vol. 4, no. 6, pp. 633–642, 2005. W. A. Devane, L. Hanus, A. Breuer, R. G. Pertwee, L. A. Stevenson, G. Griffin, D.

M AN U

[31]

Gibson, A. Mandelbaum, A. Etinger, and R. Mechoulam, “Isolation and structure of a brain constituent that binds to the cannabinoid receptor.,” Science , vol. 258, no. 5090, pp. 1946–1949, 1992.

B. F. Cravatt and A. H. Lichtman, “Fatty acid amide hydrolase: an emerging therapeutic

TE D

[32]

target in the endocannabinoid system.,” Curr. Opin. Chem. Biol., vol. 7, no. 4, pp. 469–475,

[33]

T. Bisogno and M. Maccarrone, “Latest advances in the discovery of fatty acid amide

EP

2003.

hydrolase inhibitors.,” Expert Opin. drug Discov., vol. 8, no. 5, pp. 509–522, 2013. D. L. Boger, H. Sato, A. E. Lerner, M. P. Hedrick, R. A. Fecik, H. Miyauchi, G. D.

AC C

[34]

Wilkie, B. J. Austin, M. P. Patricelli, and B. F. Cravatt, “Exceptionally potent inhibitors of fatty acid amide hydrolase: the enzyme responsible for degradation of endogenous oleamide and anandamide.,” Proc. Natl. Acad. Sci. United States Am., vol. 97, no. 10, pp. 5044–5049, 2000. [35]

M. J. Myllymäki, H. Käsnänen, A. O. Kataja, M. Lahtela-Kakkonen, S. M. Saario, A.

Poso, and A. M. P. Koskinen, “Chiral 3-(4,5-dihydrooxazol-2-yl)phenyl alkylcarbamates as

27

ACCEPTED MANUSCRIPT

novel FAAH inhibitors: Insight into FAAH enantioselectivity by molecular docking and interaction fields.,” Eur. J. Med. Chem., vol. 44, no. 10, pp. 4179–4191, 2009. [36]

J. R. Franco, P. P. Simarro, A. Diarra, and J. G. Jannin, “Epidemiology of human African

[37]

RI PT

trypanosomiasis.,” Clin. Epidemiol., vol. 6, pp. 257–275, Aug. 2014.

J. Y. Hwang, R. R. Attia, A. K. Carrillo, M. C. Connelly, and R. K. Guy, “Synthesis and

evaluation of methylsulfonylnitrobenzamides (MSNBAs) as inhibitors of the thyroid hormone

SC

receptor-coactivator interaction.,” Bioorganic & Med. Chem. Lett., vol. 23, no. 6, pp. 1891–1895, 2013.

J. Y. Hwang, R. R. Attia, F. Zhu, L. Yang, A. Lemoff, C. Jeffries, M. C. Connelly, and R.

M AN U

[38]

K. Guy, “Synthesis and Evaluation of Sulfonylnitrophenylthiazoles (SNPTs) as Thyroid Hormone Receptor–Coactivator Interaction Inhibitors,” J. Med. Chem., vol. 55, no. 5, pp. 2301– 2310, Feb. 2012.

J. Y. Hwang, D. Smithson, F. Zhu, G. Holbrook, M. C. Connelly, M. Kaiser, R. Brun,

TE D

[39]

and R. K. Guy, “Optimization of chloronitrobenzamides (CNBs) as therapeutic leads for human African trypanosomiasis (HAT).,” J. Med. Chem., vol. 56, no. 7, pp. 2850–2860, 2013. O. Rådmark, O. Werz, D. Steinhilber, and B. Samuelsson, “5-Lipoxygenase, a key

EP

[40]

enzyme for leukotriene biosynthesis in health and disease.,” Biochim. et Biophys. Acta, 2014. T. Kosaka, K. Ochiai, S. Ohba, T. Wakabayashi, and S. Murota, “A novel series of highly

AC C

[41]

potent 5-lipoxygenase inhibitors - 2-arylidienylbenzoxazolles,” Bioorganic and Medicinal Chemistry Letters, vol. 5. pp. 35–38, 1995. [42]

C. S. Demmer, J. C. Hansen, J. Kehler, and L. Bunch, “New Palladium-Catalyzed

Domino Reaction with Intramolecular Ring Closure of an N-(2-Chloro-3-heteroaryl)arylamide: First Synthesis of Oxazolo[4,5-b]pyrazines,” Adv. Synth. & Catal., vol. 356, no. 5, pp. 1047–

28

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

1055, Mar. 2014.

29

Benzoxazoles and oxazolopyridines in medicinal chemistry studies.

The benzoxazole heterocycle is often found in ligands targeting a plethora of receptors and enzymes. By analysis of published X-ray structures, this r...
2MB Sizes 3 Downloads 11 Views