U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Probe Reports from the NIH Molecular Libraries Program [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2010-.

Cover of Probe Reports from the NIH Molecular Libraries Program

Probe Reports from the NIH Molecular Libraries Program [Internet].

Show details

Identification of Modulators of the N370S Mutant Form of Glucocerebrosidase as a Potential Therapy for Gaucher Disease - Chemotype 1

, , , , , , and .

Author Information and Affiliations

Received: ; Last Update: May 5, 2011.

In this report, ML155 (CID 40225210) is described as a probe that is able to inhibit the hydrolytic activity of the N370S mutant form of glucocerebrosidase, as well as wild type glucocerebrosidase, in tissue homogenate assays. The probe does not inhibit purified glucocerebrosidase, but the cellular activity of the enzyme is known to be dependent on interactions with other factors, such as Saposin C. Importantly, the probe increased glucocerebrosidase translocation to the lysosome in Gaucher patient-derived fibroblasts homozygous for the N370S mutation, and can be used to study ER-lysosomal trafficking of clinically relevant GC mutants in vitro. This probe may be a useful lead for the clinical development of a chemical chaperone of glucocerebrosidase.

Assigned Assay Grant #: R03MH086442

Screening Center Name & PI: NIH Chemical Genomics Center, Dr. Christopher P. Austin

Chemistry Center Name & PI: NIH Chemical Genomics Center, Dr. Christopher P. Austin

Assay Submitter & Institution: National Human Genome Research Institute, Dr. Ellen Sidransky

PubChem Summary Bioassay Identifier (AID): 2593

Probe Structure & Characteristics

CID/ML#Target NameIC50/EC50 (nM) [SID, AID]Anti-target Name(s)IC50/EC50 (μM) [SID, AID]Fold SelectiveSecondary Assay(s) Name: IC50/EC50 (nM) [SID, AID]
40225210/ML155N370S GC*330 nM [85267237, 2590]Alpha-glucosidase>57μM [85267237, 2577]>100-foldChaperone activity in N370S GC fibroblasts: 500 nM [85267237, 2587]
40225210/ML155Alpha-galactosidase> 57μM [85267237, 2578]>100-fold
*

GC = glucocerebrosidase, also known as beta-glucosidase.

Recommendations for scientific use of the probe

The probe is able to inhibit the hydrolytic activity of the N370S mutant form of glucocerebrosidase, as well as wild type glucocerebrosidase, in tissue homogenate assays. The probe does not inhibit purified glucocerebrosidase, but the cellular activity of the enzyme is known to be dependent on interactions with other factors, such as Saposin C. Importantly, the probe increased glucocerebrosidase translocation to the lysosome in Gaucher patient-derived fibroblasts homozygous for the N370S mutation, and can be used to study ER-lysosomal trafficking of clinically relevant GC mutants in vitro. This probe may be a useful lead for the clinical development of a chemical chaperone of glucocerebrosidase.

1. Introduction

Gaucher disease is an autosomal recessive disorder resulting from mutations in the enzyme Glucocerebrosidase (EC 3.2.1.45; also known as acid beta-glucosidase) and affects 1 in 50,000 live births (1). The function of Glucocerebrosidase (GC) is to hydrolyze the beta glucosidic linkage of glucocerebrosidases, also called glucosylceramides (2). These glycosphingolipids are cell membrane components that maintain the stability of the lipid bilayer, function as cellular recognition elements and play an important role in cellular adherence (3). The deficiency of GC due to the genetic mutations results in the accumulation of glucosylceramides in lysosomes.

There are more than 200 recognized mutations of the GC gene (4). Although many GC mutants are still functional (5), conformational differences in mutant proteins reduce their recognition by transporters, resulting in ER protein accumulation and premature degradation in endosome. The inability of mutant proteins to reach their site of action causes lysosomal accumulation of glucosylceramides and lysosomal enlargement. Ultimately, this leads to liver and spleen enlargements, as well as the neurological symptoms in type 2 and type 3 Gaucher disease.

Currently, the only FDA approved treatment for Gaucher disease is enzyme replacement therapy using the human recombinant glucocerebrosidase, Cerezyme. Although this approach does address some aspects of the disease, the limited tissue distribution of the infused enzyme, for example poor CNS penetration, reduces its therapeutic benefits in type 2 and type 3 Gaucher patients (6). Small molecule chaperone therapy has been proposed as an alternative therapeutic strategy for Gaucher disease. The binding of small molecules to mutant protein may facilitate proper folding and translocation of the mutant protein to the lysosome (7–8). Several imino sugar inhibitors of glycosidases have been reported to have chaperone activity (9–20). Isofagomine is an imino sugar that can act as a chaperone for glucocerebrosidase, but was recently withdrawn from clinical trial testing.

Imino sugars inhibit glycosidases by mimicking the transition state of the glycosidic cleavage, and as such tend to be poorly selective (13). Therefore, it would be desirable to develop alternative, more selective series with chaperone activity. It is also important to remark that molecules with very potent inhibitors may have a lower probability of ultimately producing a therapeutic effect or an increase in GC activity in the lysosome. Though the GC protein translocation is increased, the tightly bound inhibitors may not be easily displaced by the natural substrate, and may even inhibit any residual activity of the enzyme (2), (8), (21). Previous chaperone molecules that have entered clinical trials are all potent GC inhibitors. Our goal is to identify a non-imino sugar series with modest inhibitory capacity and good chaperone activity.

GC activity is modulated in cells through the binding of an allosteric activator, Saposin C (27). In the GC enzyme assay with a purified enzyme, the addition of sodium taurocholate, a bile salt, is required to activate the enzyme (23). Assays by our group and others that screen for inhibitors of GC using purified enzyme have identified several interesting inhibitor chemotypes, but most have reduced or no activity when tested using tissue homogenate that contains both GC and Saposin C, as well as other components that may be required for GC function in cells. We speculate that this variation in activity is due to the GC conformational differences between the active conformation induced by detergent and the one induced by Saposin C and/or other factors in cells. Additionally, 70% of Gaucher patients carry the N370S mutation, and therefore we developed an assay utilizing the spleen (28) homogenate derived from a GC patient homozygous for the N370S mutation in the primary screen. This assay uses a fluorogenic substrate, 4-methylumbelliferone β-D-glucopyranoside, to monitor glucoerebrosidase specific activity, and the spleen tissue homogenate as the GC enzyme preparation. Upon hydrolysis, 4-methylumbelliferone (4-MU) is liberated, which produces a fluorescent emission at 440 nm when excited at 370 nm.

2. Materials and Methods

Unless otherwise stated, all reactions were carried out under an atmosphere of dry argon or nitrogen in dried glassware. Indicated reaction temperatures refer to those of the reaction bath, while room temperature (rt) is noted as 25oC. All solvents were of anhydrous quality purchased from Aldrich Chemical Co. and used as received. Commercially available starting materials and reagents were purchased from Aldrich, TCI and Acros and were used as received. Analytical thin layer chromatography (TLC) was performed with Sigma Aldrich TLC plates (5x 20cm, 60 Å, 250μm). Visualization was accomplished by irradiation under a 254nm UV lamp.

Chromatography on silica gel was performed using forced flow (liquid) of the indicated solvent system on Biotage KPSil pre-packed cartridges and using the Biotage SP-1 automated chromatography system. 1H NMR spectra were recorded on a Varian Inova 400 MHz spectrometer. Chemical shifts are reported in ppm with the solvent resonance as the internal standard (CDCl3 7.27 ppm, DMSO-d6 2.49 ppm, for 1H NMR). Data are reported as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, sep = septet, quin = quintet, br = broad, m = multiplet), coupling constants, and number of protons. Low resolution mass spectra (electrospray ionization) were acquired on an Agilent Technologies 6130 quadrupole spectrometer coupled to an Agilent Technologies 1200 series HPLC. The HPLC retention times were recorded through short standard gradient 4% to 100% acetonitrile (0.05% TFA) over 3 minutes (t1) or long standard gradient 4% to 100% acetonitrile (0.05% TFA) over 7 minutes (t2) using Luna C18 3 micron 3x 75mm column with a flow rate of 0.800 ml/min. High resolution mass spectral data was collected in-house using an Agilent 6210 time-of-flight mass spectrometer, also coupled to an Agilent Technologies 1200 series HPLC system.

2.1. Assays

PubChem AIDTypeTargetConc. RangeSamples TestedNotes
2101Primary qHTSN370S GC57.5μM–0.7nM326,770Tissue, blue
2590ConfirmatoryN370S GC54μM–0.01nM320Tissue, blue
2671ConfirmatoryN370S GC54μM–0.01nM142Tissue, blue
2613SecondaryN370S GC57.5μM–0.3nM83Tissue, red
2592SecondaryWildtype GC57.5μM–0.3nM21Tissue, blue
2588SecondaryWildtype GC50μM–0.1nM152Tissue, red
2595SecondaryWildtype GC77μM–0.3nM52Purified, blue
2597SecondaryN370S GC77μM–0.3nM52Purified, blue
2596SecondaryN370S GC230μM–0.1nM94Purified, natural substrate
2577Anti-targetAlpha- glucosidase57.5μM–0.3nM70Purified, blue
2578Anti-targetAlpha- galactosidase57.5μM–0.3nM70Purified, blue
2587TertiaryN370S GC100μM–10nM4Immunostaining of fibroblast lysosomes
2589TertiaryWildtype GC100μM–10nM4Immunostaining of fibroblast lysosomes
2593SummaryN370S GC

qHTS assay for activators and inhibitors of N370S Mutant Form of Glucocerebrosidase [AID:2101]

Assay details and protocol: This is a fluorogenic enzyme assay with 4-methylumbelliferyl-beta-D-glucopyranoside as the substrate and the glucocerebrosidase from spleen homogenate of a N370S Gaucher patient as the enzyme preparation. Upon the hydrolysis of this fluorogenic substrate, the resulting product, 4-methyllumbelliferone, can be excited at 365 nm and emits at 440 nm, which can be detected by a standard fluorescence plate reader. Data were normalized to the controls for basal activity (without enzyme) and 100% activity (with enzyme). The AC50 values were determined from concentration-response data modeled with the standard Hill equation.

The human spleen tissue was homogenized using a food blender at the maximal speed for 5 minutes, followed by 10 passes in a motor-driven 50 ml glass-Teflon homogenizer. The homogenate was centrifuged at 1000 ×g for 10 min. The supernatant was then filtered using a 40 um filter and aliquots of resultant spleen homogenate were frozen at −80°C until use. (Assay buffer: 50mM citric acid (titrated with potassium phosphate to pH 5.0), 100mM potassium chloride, 10mM sodium chloride, 1mM magnesium chloride, 0.01% Tween-20.)

1536-well assay protocol:

  1. Add 2μl/well of spleen homogenate (27μg final)
  2. Add 23nl compounds in DMSO solution. The final titration was 0.5nM to 58μM.
  3. Add 2μl of substrate (1mM final)
  4. Incubate at 37° C for 40 min.
  5. Add 2μl stop solution (1M NaOH and 1M Glycine mixture, pH 10)
  6. Detect the assay plate in a ViewLux plate reader (PerkinElmer) with Ex=365 nm and Em=440nm.

2.2. Probe Chemical Characterization

Structural verification information of probe SID: 40225210/ML155

1H NMR (400 MHz, DMSO-d6) δ ppm 3.28 (m, 4H), 3.84 (m, 4H), 7.16 (dd, 1H, J=1.6, 5.2 Hz), 7.42 (m, 1H), 7.729 (m, 6H), 7.80 (dd, 1H, J= 2.0, 8.8 Hz), 7.90 (m, 2H), 8.07 (d, 1H, J= 8.0 Hz), 8.20 (m, 2H), 8.49 (s, 1H).

LC/MS (Agilent system) Retention time t1 (short) = 3.22 min

Purity: UV220 > 99%, UV254 > 99%; MS m/z 487.1 (M+H);

Column: 3x 75mm Luna C18, 3 micron

Run time: 4.5 min (short)

Gradient: 4% to 100%

Mobile phase: Acetonitrile (0.025% TFA), water (0.05% TFA).

Flow rate: 0.8 to 1.0ml

Temperature: 50oC

UV wavelength: 220 nm, 254 nm

The corresponding CID is 40225210 and is commercially available for purchase from the following vendors:

Order number: ken-705035 and registry number: 1090708-11-4

Aurora Fine Chemicals LLC

7929 Silverton Ave.

Suite 609

San Diego, CA, 92126

USA

Phone: +1 858 549 4700

Fax: +1 858 549 4701

Email: moc.slacimehcenifarorua@arorua

Web: http://www.aurorafinechemicals.com

Order number: T6239298 and registry number: 1090708-11-4

Enamine

23 Alexandra Matrosova Street

Kiev, 01103

Ukraine

Phone: +380 44 537 32 18

Fax: +380 44 537 32 53

Email: ten.enimane@enimane

Web: http://www.enamine.net

Order number: T6239298 and registry number: 1090708-11-4

Ambinter

50, avenue de Versailles

Paris, F-75016

France

Phone: (33-1) 45 24 48 60

Fax: (33-1) 45 24 62 41

Email: moc.retnibma@tcatnoc

Web: http://www.ambinter.com

MLS IDSIDCIDNCGC IDMLType
MLS0034292328526723740225210NCGC00182292155Probe
MLS0026998138526740644246455NCGC00186073Analog
MLS002699814852672764787756NCGC00182327Analog
MLS0026998158526740325708371NCGC00186069Analog
MLS0026998168526740744246456NCGC00186444Analog
MLS0026998178526740844246457NCGC00186445Analog
MLS0026998189238662445136810NCGC00187483Analog
MLS0003939621042193152477651MLS000393962Analog
MLS000393919224108734035156MLS000393919Analog

2.3. Probe Preparation

With the confirmatory data in hand, we embarked on systematic SAR modifications based on commercially available quinazolines as shown in Table 1. Scheme 1 discloses the synthesis strategy for some of the modifications at the quinazoline core (26).

Table 1. SAR of commercial quinazolines.

Table 1

SAR of commercial quinazolines.

Scheme 1. General strategy for modification in the functional core.

Scheme 1

General strategy for modification in the functional core.

Commercially available Boc protected piperazine 14 was sulphonytaled, using the sulphonyl chloride 15 in the presence of a suitable base such as Diisopropylethylamine, followed by a quantitative deprotection of the Boc functional group to yield intermediate 16. The next step involves the selective chloro displacement at the core ring 17 to produce compound 18, followed by a Suzuki cross-coupling reaction with an aromatic or heteroaromatic boronic acid 19 to produce final compound 20. Application of this synthetic strategy allowed us the synthesis of analogs having quinazoline, pyrimidine and isoquinoline cores.

The synthesis of analogs with a purine core, Scheme 2, required the selective protection of the ring NH with a methyl pivalate functional group, followed by cross coupling and deprotection. Numerous attempts of carrying out Suzuki reactions with the free purine core or using a Boc protecting group failed to yield the intended coupling product.

Scheme 2. Synthesis of purine analogs.

Scheme 2

Synthesis of purine analogs.

For the SAR study of the sulphonamide portion of the molecule, we modified the synthetic procedure toward a more convenient methodology that allowed us to introduce variation at the last step of the synthesis. Scheme 3 shows how sulphonylation, carboxylic coupling and alkylation of the key intermediate 32 provided a variety of final compounds.

Scheme 3. Synthesis of analogs with modification of the aromatic sulphonamide.

Scheme 3

Synthesis of analogs with modification of the aromatic sulphonamide.

We also synthesized a number of analogs testing the flexibility of the diamine linker, as well as the activity of other amine-linked functionalities attached to the piperidine ring. Schemes 4 and 5 present example syntheses of some of these compounds, which were performed in a similar fashion to the previously described compound syntheses.

Scheme 4. Synthesis of analogs with a flexible linker.

Scheme 4

Synthesis of analogs with a flexible linker.

Scheme 5. Synthesis of additional analogues with a flexible linker.

Scheme 5

Synthesis of additional analogues with a flexible linker.

Regarding the synthesis of analogs with aromatic and heteroaromatic modifications at the 2 position, Scheme 6 shows the synthetic methodology involving the production of the organotin 63 as a convenient intermediate for last step diversification using Stille coupling. Thus, 2,4 quinazoline dione, 60, was converted in 2,4-dibromoquinazoline 61 with phosphorus oxybromide. Selective halogen displacement with substituted piperidine 21 and Lithium halogen exchange followed by reaction with tributyl tin chloride yielded compound 63. Cross coupling reaction between 63 and heteroaromatic halogens yielded final compounds 64.

Scheme 6. Synthesis of analogs with modifications at the 2 position.

Scheme 6

Synthesis of analogs with modifications at the 2 position.

Additional modifications at the same position 2 were obtained as show in Schemes 7 and 8. Thus, cross-coupling reaction with Zinc cyanide catalyzed by Palladium yielded intermediate 66. Hydrolysis and esterification of nitrile 66 yielded final compounds 67–69. Reduction of starting material 65 produced compound 70, and displacement of the chloro at position 2 yielded final compound 71.

Scheme 7. Additional modifications at the 2 position.

Scheme 7

Additional modifications at the 2 position.

Scheme 8. Synthesis of thiophene analogs with improved solubility.

Scheme 8

Synthesis of thiophene analogs with improved solubility.

3. Results

3.1. Summary of Screening Results

1300 1536-well plates were screened in qHTS format (most compounds were assayed in titration from 90nM to 57μM final concentration in the primary screen) with a Z’ of 0.80 +/− 0.16 across the entire set of plates, indicating robust performance of the assay. A total of 326,770 compounds were screened (22–23) (AID 2101).

83 of the more potent, high confidence inhibitors and activators were re-ordered for confirmation in N370S GC spleen homogenate using the resorufin-based ‘red’ fluorescent substrate (AID 2613). Several quinazoline analogs were confirmed to be active, including MLS000393962 (CID 2477651), which is compound 9 below. Based on similar activity in this secondary assay, we chose to develop the SAR of the series by purchasing available analogs from commercial vendors. Table 1 discloses the inhibitory activity of compounds from the primary screen and the initial SAR-by-catalog approach. The activity is reported as N370S spleen homogenate AC50, concentration necessary to inhibit 50 % of the 4-methylumbelliferone β-D-glucopyranoside hydrolysis.

3.2. Dose Response Curves for Probe

Figure 1. Inhibitory curves of compound 13 (CID 40225210), against spleen homogenate homozygous for N370S GC using 4-methylumbelliferyl-β-D-glucopyranoside (curve in black) and resorufin β-D-glucopyranoside as substrates (curve in blue), and wildtype spleen homogenate using 4-methylumbelliferyl-β-D-glucopyranoside (curve in cyan; AID 2592).

Figure 1Inhibitory curves of compound 13 (CID 40225210), against spleen homogenate homozygous for N370S GC using 4-methylumbelliferyl-β-D-glucopyranoside (curve in black) and resorufin β-D-glucopyranoside as substrates (curve in blue), and wildtype spleen homogenate using 4-methylumbelliferyl-β-D-glucopyranoside (curve in cyan; AID 2592)

3.3. Scaffold/Moiety Chemical Liabilities

The probe molecule does not contain any functional groups with potential chemical liabilities.

3.4. SAR Tables

Tables 38 show the capacity of compounds to inhibit the hydrolysis of 4-methylumbelliferone β-D-glucopyranoside. As a positive control, we measured the activity of isofagomine in the same assay (AC50 = 0.080μM).

Table 3. Analogs with sulfonamide aromatic modifications having a phenyl at the two position.

Table 3

Analogs with sulfonamide aromatic modifications having a phenyl at the two position.

Table 4. Analogs with sulfonamide aromatic modifications having a thiophene at the two position.

Table 4

Analogs with sulfonamide aromatic modifications having a thiophene at the two position.

Table 5. A continuation of Table 4 - analogs with sulfonamide aromatic modifications having a thiophene at the two position.

Table 5

A continuation of Table 4 - analogs with sulfonamide aromatic modifications having a thiophene at the two position.

Table 6. Analogs with the modifications at the linker.

Table 6

Analogs with the modifications at the linker.

Table 7. Analogs with modification at the 2 position of the quinazoline core.

Table 7

Analogs with modification at the 2 position of the quinazoline core.

Table 8. Analogs with modifications at the molecular core.

Table 8

Analogs with modifications at the molecular core.

3.5. Cellular Activity

Compound 13 was characterized in a direct assay of chaperone activity using patient fibroblasts; that is, we measured 13’ s ability to increase the translocation of GC to the lysosome (8), (16), (24), (25). Briefly, fibroblasts obtained from a Gaucher patient homozygous for the N370S mutation and from another donor with wildtype GC were incubated for five days with NCGC00182292 (CID 40225210), in a range of concentrations from 1nM to 50μM, followed by cell fixation and staining with a selective fluorescent GC-antibody (AID2587 and AID2589). Compounds able to increase the ER-Lysosomal traffic show increased fluorescent GC-antibody staining in lysosomes. DMSO and Isofagomine were use as negative and positive controls. Figure 2 shows the increment of signal in produced by 13, thereby confirming the chaperone activity of this compound.

Figure 2. Chaperone activity of compound 13, NCGC00182292 (CID 40225210), and others using wildtype and homozygous, mutant N370S GC fibroblasts.

Figure 2

Chaperone activity of compound 13, NCGC00182292 (CID 40225210), and others using wildtype and homozygous, mutant N370S GC fibroblasts. Two genotypes of fibroblasts, fibroblasts homozygous for wildtype GC (top) and fibroblasts homozygous for N370S GC (bottom) (more...)

3.6. Profiling Assays

Selectivity was measured against other two lysosomal hydrolases: alpha glucosidase (AID 2578) and alpha galactosidase (AID 2577). All tested compounds from this quinazoline series show selectivity for inhibiting glucocerebrosidase. None of them show inhibitory activity towards alpha glucosidase or alpha galactosidase at concentrations up to 57μM.

4. Discussion

The primary screen for this project assayed for GC enzyme in the context of spleen homogenate derived from a N370S Gaucher patient by monitoring the production of a fluorogenic substrate, 4-methylumbelliferone β-D-glucopyranoside. In addition, several other variations of GC enzyme assays were developed to help eliminate the false positive compounds in the primary screen, including a similar enzyme assay with a red-shifted fluorogenic substrate (resorufin-β-D-glucopyranoside) and an enzyme assay with a native substrate (glucosylceramide). Unfortunately, cleavage of glucosylceramide can only be done in the context of purified enzyme, as this enzyme assay uses a coupled enzyme reaction for detection in which the glucose oxidase/amplex red interacts with glucose, the product of glucosylceramide cleavage, to produce a fluorogenic signal. The strengths and weaknesses of these assays are described in detail below. In all the assays, activators and auto-fluorescent compounds may both give an apparent increase in signal and quenchers may give a decrease in signal. To eliminate nonspecific compounds, counterscreen enzyme assays with alpha-galactosidase and alpha-glucosidase were used. Activators and inhibitors were then confirmed using the primary screening assays as well as glucocerebrosidase assays using a red-shifted fluorogenic substrate, and product formation was also directly tracked using LC/MS. Once activity was confirmed, SAR studies were undertaken to characterize chemotypes in more detail.

Figure 3. Principles of enzyme reactions and product spectrums of two GC enzyme assays.

Figure 3Principles of enzyme reactions and product spectrums of two GC enzyme assays

(a) The “Red” GC enzyme assay. The pro-fluorescent substrate Res-β-glucopyranoside is hydrolyzed to form two products, glucose and resorufin, with an excitation perk of 573 nm and an emission perk of 590 nm. This assay is used for the primary screen. (b) The “Blue” GC enzyme assay. The pro-fluorescent substrate 4MU-β-Glc is hydrolyzed to form two products, glucose and 4MU, with an excitation perk of 365 nm and an emission peak of 440 nm.

An inhibitory chemical probe for this project is defined as a molecule having EC50 values in lower μM in the cell-based homogenate assay, and being selective against other glycosidic hydrolases (selective – EC50 values > 10 fold against α-galactosidase and α-glucosidase).

From the primary screen, we identified several series of modulators, including several imino sugar molecules previously reported to inhibit the enzyme such as isofagomine (Table 9).

Table 9. Compound representatives of initial hit series.

Table 9

Compound representatives of initial hit series. Isofagomine is an imino sugar previously reported to chaperone glucocerebrosidase. NCGC00092410 (CID 5067281) is a non-imino sugar previously reported to inhibit purified glucocerebrosidase (23).

4.1. Comparison to existing art and how the new probe is an improvement

The probe represents one of two new, non-imino sugar chemotypes that can act as a chaperone of glucocerebrosidase in vitro. There are not significant differences in activity between this probe and the other, ML156 (NCGC00159568, CID 9893924). Isofagomine (CID 447607) has been shown to chaperone glucocerebrosidase in vitro, but has been abandoned as a candidate for further clinical development (http://www.gaucher.org.uk/enews.php?id=303). NCGC00092410 (CID 5067281) is a non-imino sugar previously reported to inhibit purified glucocerebrosidase (Zheng et al, Proc. Natl. Acad. Sci. U.S.A. 2007, 104, 13192), but is much less active in tissue homogenate assays, likely indicating non-specific protein binding.

4.2. Mechanism of Action Studies

In enzyme kinetics assays, the probe, along with other glucocerebrosidase inhibitors such as isofagomine demonstrated mixed mode inhibition – neither competitive nor non-competitive enzyme kinetics. It is obvious, from x-ray crystal structure studies of isofagomine binding, that the compound binds in the active site. Thus, the current homogenate-based assays might be ill-suited for such kinetic characterization.

4.3. Planned Future Studies

No further characterization is planned at this time.

5. References

1.
Grabowski G. Phenotype, diagnosis, and treatment of Gaucher’s disease. The Lancet. 2008;372(9645):1263–1271. [PubMed: 19094956]
2.
Sawkar AR, D’Haeze W, Kelly JW. Therapeutic strategies to ameliorate lysosomal storage disorders – a focus on Gaucher disease. Cell. Mol. Life Sci. 2006;63:1179–1192. [PubMed: 16568247]
3.
Wedeking A, van Echten-Deckert G. Curr. Org. Chem. 2007;11:579.
4.
Hruska KS, LaMarca ME, Sidransky E. Futerman AH, Zimran A. Gaucher Disease. CRC Press; Boca Raton, FL: 2006. p. 13.
5.
Futerman AH, van Meer G. The Cell Biology of Lysosomal Storage Disorders. Nat. Rev. Mol. Cell. Biol. 2004;5:554–565. [PubMed: 15232573]
6.
Barton NW, Brady RO, Dambrosia JM, Dibisceglie AM, Doppelt SH, Hill SC, Mankin HJ, Murray GJ, Parker RI, Argoff CE, et al. Replacement therapy for inherited enzyme deficiency--macrophage-targeted glucocerebrosidase for Gaucher’s disease. N. Engl. J. Med. 1991;324:1464–1470. [PubMed: 2023606]
7.
Butters TD. Pharmacotherapeutic strategies using small molecules for the treatment of glycolipid lysosomal storage disorders. Expert Opin. Pharmacother. 2007;8:427–435. [PubMed: 17309337]
8.
Sawker AR, Cheng W-C, Beutler W, Wong C-H, Balch WE, Kelly JW. Chemical chaperones increase the cellular activity of N370S β-glucosidase: A therapeutic strategy for Gaucher disease. Proc. Natl. Acad. Sci. U.S.A. 2002;99:15428–15433. [PMC free article: PMC137733] [PubMed: 12434014]
9.
Sawker AR, Adamski-Werner SL, Cheng W-C, Wong C-H, Beutler E, Zimmer K-P, Kelly JW. Gaucher Disease-Associated Glucocerebrosidases Show Mutation-Dependent Chemical Chaperoning Profiles. Chem. Biol. 2005;12:1235–1244. [PubMed: 16298303]
10.
Yu Z, Sawker AR, Whalen LJ, Wong C-H, Kelly JWJ. Med. Chem. 2007;50:94. [PMC free article: PMC2543937] [PubMed: 17201413]
11.
Wennekes T, van der Berg RJBHN, Donker W, van der Marel GA, Strijland A, Aerts JMFG, Overkleeft HS. J. Org. Chem. 2007;72:1088. [PubMed: 17243712]
12.
Zhu X, Sheth KA, Li S, Chang H-H, Fan J-Q. Angew. Chem. Int. Ed. 2005;44:7450. [PubMed: 16231389]
13.
Butters TD, Dwek RA, Platt FM. Glycobiology. 2005;15:43R–52R. [PubMed: 15901676]
14.
Chang HH, Asano N, Ishii S, Ichikawa Y, Fan JQ. FEBS J. 2006;273:4082–4092. [PubMed: 16934036]
15.
Compain P, Martin OR, Boucheron C, Godin G, Yu L, Ikeda K, Asano N. Chem Bio Chem. 2006;7:1356–1359. [PubMed: 16871601]
16.
Sawkar AR, Schmitz M, Zimmer KP, Reczek D, Edmunds T, Balch WE, Kelly JW. ACS. Chem Biol. 2006;1:235–251. [PubMed: 17163678]
17.
Steet RA, Chung S, Wustman B, Powe A, Do H, Kornfeld SA. Proc Natl Acad Sci USA. 2006;103:13813–13818. [PMC free article: PMC1564243] [PubMed: 16945909]
18.
Yu L, Ikeda K, Kato A, Adachi I, Godin G, Compain P, Martin O, Asano N. Bioorg Med Chem. 2006;14:7736–7744. [PubMed: 16919960]
19.
Egido-Gabas M, Canals D, Casas J, Llebaria A, Delgado A. ChemMed-Chem. 2007 [PubMed: 17479993]
20.
Lei K, Ninomiya H, Suzuki M, Inoue T, Sawa M, Iida M, Ida H, Eto Y, Ogawa S, Ohno K, et al. Biochim Biophys Acta. 2007;1772:587–596. [PubMed: 17363227]
21.
Fan JQ. Trends Pharmacol Sci. 2003;24:355–360. [PubMed: 12871668]
22.
Inglese J, Auld DS, Jadhav A, Johnson RL, Simeonov A, Yasgar A, Zheng W, Austin CP. Proc Natl Acad Sci USA. 2006;103:11473–11478. [PMC free article: PMC1518803] [PubMed: 16864780]
23.
Zheng W, Padia J, Urban DJ, Jadhav A, Goker-Alpan O, Simeonov A, Goldin E, Auld D, LaMarca ME, Inglese J, Austin CP, Sidransky E. Three classes of glucocerebrosidase inhibitors identified by quantitative high-throughput screening are chaperone leads for Gaucher disease. Proc. Natl. Acad. Sci. U.S.A. 2007;104:13192. [PMC free article: PMC1936979] [PubMed: 17670938]
24.
Schmitz M, Alfalah M, Aerts JM, Naim HY, Zimmer KP. Int J Biochem Cell Biol. 2005;37:2310–2320. [PubMed: 15982918]
25.
Lieberman RL, Wustman BA, Huertas P, Powe AC Jr, Pine CW, Khanna R, Schlossmacher MG, Ringe D, Petsko GA. Nat Chem Biol. 2007;3:101–107. [PubMed: 17187079]
26.
Wéber C, Bielik A, Demeter A, Borza I, Szendrei GI, Keser GM, Greiner I. Solid-phase synthesis of 6-hydroxy-2,4-diaminoquinazolines. Tetrahedron. 2005;61:9375–9380.
27.
John M, Wendeler M, Heller M, Sandhoff K, Kessler H. Characterization of Human Saposins by NMR Spectroscopy. Biochemistry. 2006;45(16):5206–5216. [PubMed: 16618109]
28.
We evaluated several tissues finding that the spleen has one of the highest levels of GC activity.

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this page (584K)

Related information

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...