Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where B. Michael Silber is active.

Publication


Featured researches published by B. Michael Silber.


Journal of Medicinal Chemistry | 2011

2-Aminothiazoles as Therapeutic Leads for Prion Diseases

Alejandra Gallardo-Godoy; Joel R. Gever; Kimberly L. Fife; B. Michael Silber; Stanley B. Prusiner; Adam R. Renslo

2-Aminothiazoles are a new class of small molecules with antiprion activity in prion-infected neuroblastoma cell lines (J. Virol. 2010, 84, 3408). We report here structure-activity studies undertaken to improve the potency and physiochemical properties of 2-aminothiazoles, with a particular emphasis on achieving and sustaining high drug concentrations in the brain. The results of this effort include the generation of informative structure-activity relationships (SAR) and the identification of lead compounds that are orally absorbed and achieve high brain concentrations in animals. The new aminothiazole analogue (5-methylpyridin-2-yl)-[4-(3-phenylisoxazol-5-yl)-thiazol-2-yl]-amine (27), for example, exhibited an EC(50) of 0.94 μM in prion-infected neuroblastoma cells (ScN2a-cl3) and reached a concentration of ∼25 μM in the brains of mice following three days of oral administration in a rodent liquid diet. The studies described herein suggest 2-aminothiazoles as promising new leads in the search for effective therapeutics for prion diseases.


Journal of Pharmacology and Experimental Therapeutics | 2013

Biaryl Amides and Hydrazones as Therapeutics for Prion Disease in Transgenic Mice

Duo Lu; Kurt Giles; Zhe Li; Satish Rao; Elena Dolghih; Joel R. Gever; Michal Geva; Manuel Elepano; Abby Oehler; Clifford Bryant; Adam R. Renslo; Matthew P. Jacobson; Stephen J. DeArmond; B. Michael Silber; Stanley B. Prusiner

The only small-molecule compound demonstrated to substantially extend survival in prion-infected mice is a biaryl hydrazone termed “Compd B” (4-pyridinecarboxaldehyde,2-[4-(5-oxazolyl)phenyl]hydrazone). However, the hydrazone moiety of Compd B results in toxic metabolites, making it a poor candidate for further drug development. We developed a pharmacophore model based on diverse antiprion compounds identified by high-throughput screening; based on this model, we generated biaryl amide analogs of Compd B. Medicinal chemistry optimization led to multiple compounds with increased potency, increased brain concentrations, and greater metabolic stability, indicating that they could be promising candidates for antiprion therapy. Replacing the pyridyl ring of Compd B with a phenyl group containing an electron-donating substituent increased potency, while adding an aryl group to the oxazole moiety increased metabolic stability. To test the efficacy of Compd B, we applied bioluminescence imaging (BLI), which was previously shown to detect prion disease onset in live mice earlier than clinical signs. In our studies, Compd B showed good efficacy in two lines of transgenic mice infected with the mouse-adapted Rocky Mountain Laboratory (RML) strain of prions, but not in transgenic mice infected with human prions. The BLI system successfully predicted the efficacies in all cases long before extension in survival could be observed. Our studies suggest that this BLI system has good potential to be applied in future antiprion drug efficacy studies.


ChemMedChem | 2013

2-Aminothiazoles with Improved Pharmacotherapeutic Properties for Treatment of Prion Disease

Zhe Li; B. Michael Silber; Satish Rao; Joel R. Gever; Clifford Bryant; Alejandra Gallardo-Godoy; Elena Dolghih; Kartika Widjaja; Manuel Elepano; Matthew P. Jacobson; Stanley B. Prusiner; Adam R. Renslo

Recently, we described the aminothiazole lead (4‐biphenyl‐4‐ylthiazol‐2‐yl)‐(6‐methylpyridin‐2‐yl)‐amine (1), which exhibits many desirable properties, including excellent stability in liver microsomes, oral bioavailability of ∼40 %, and high exposure in the brains of mice. Despite its good pharmacokinetic properties, compound 1 exhibited only modest potency in mouse neuroblastoma cells overexpressing the disease‐causing prion protein PrPSc. Accordingly, we sought to identify analogues of 1 with improved antiprion potency in ScN2a‐cl3 cells while retaining similar or superior properties. Herein we report the discovery of improved lead compounds such as (6‐methylpyridin‐2‐yl)‐[4‐(4‐pyridin‐3‐yl‐phenyl)thiazol‐2‐yl]amine and cyclopropanecarboxylic acid (4‐biphenylthiazol‐2‐yl)amide, which exhibit brain exposure/EC50 ratios at least tenfold greater than that of compound 1.


European Journal of Medicinal Chemistry | 2011

Discovery of 6-substituted indole-3-glyoxylamides as lead antiprion agents with enhanced cell line activity, improved microsomal stability and low toxicity.

Mark J. Thompson; Jennifer C. Louth; Steven Ferrara; Matthew P. Jackson; Fiona J. Sorrell; Edward J. Cochrane; Joel R. Gever; B. Michael Silber; Henry Roehl; Beining Chen

A series of highly potent indole-3-glyoxylamide based antiprion agents was previously characterized, focusing on optimization of structure-activity relationship (SAR) at positions 1-3 of the indole system. New libraries interrogating the SAR at indole C-4 to C-7 now demonstrate that introducing electron-withdrawing substituents at C-6 may improve biological activity by up to an order of magnitude, and additionally confer higher metabolic stability. For the present screening libraries, both the degree of potency and trends in SAR were consistent across two cell line models of prion disease, and the large majority of compounds showed no evidence of toxic effects in zebrafish. The foregoing observations thus make the indole-3-glyoxylamides an attractive lead series for continuing development as potential therapeutic agents against prion disease.


Journal of Pharmacology and Experimental Therapeutics | 2015

Different 2-Aminothiazole Therapeutics Produce Distinct Patterns of Scrapie Prion Neuropathology in Mouse Brains

Kurt Giles; David B. Berry; Carlo Condello; Ronald C. Hawley; Alejandra Gallardo-Godoy; Clifford Bryant; Abby Oehler; Manuel Elepano; Sumita Bhardwaj; Smita Patel; B. Michael Silber; Shenheng Guan; Stephen J. DeArmond; Adam R. Renslo; Stanley B. Prusiner

Because no drug exists that halts or even slows any neurodegenerative disease, developing effective therapeutics for any prion disorder is urgent. We recently reported two compounds (IND24 and IND81) with the 2-aminothiazole (2-AMT) chemical scaffold that almost doubled the incubation times in scrapie prion-infected, wild-type (wt) FVB mice when given in a liquid diet. Remarkably, oral prophylactic treatment with IND24 beginning 14 days prior to intracerebral prion inoculation extended survival from ∼120 days to over 450 days. In addition to IND24, we evaluated the pharmacokinetics and efficacy of five additional 2-AMTs; one was not followed further because its brain penetration was poor. Of the remaining four new 2-AMTs, IND114338 doubled and IND125 tripled the incubation times of RML-inoculated wt and Tg4053 mice overexpressing wt mouse prion protein (PrP), respectively. Neuropathological examination of the brains from untreated controls showed a widespread deposition of self-propagating, β-sheet-rich “scrapie” isoform (PrPSc) prions accompanied by a profound astrocytic gliosis. In contrast, mice treated with 2-AMTs had lower levels of PrPSc and associated astrocytic gliosis, with each compound resulting in a distinct pattern of deposition. Notably, IND125 prevented both PrPSc accumulation and astrocytic gliosis in the cerebrum. Progressive central nervous system dysfunction in the IND125-treated mice was presumably due to the PrPSc that accumulated in their brainstems. Disappointingly, none of the four new 2-AMTs prolonged the lives of mice expressing a chimeric human/mouse PrP transgene inoculated with Creutzfeldt-Jakob disease prions.


Bioorganic & Medicinal Chemistry | 2014

Novel compounds lowering the cellular isoform of the human prion protein in cultured human cells

B. Michael Silber; Joel R. Gever; Satish Rao; Zhe Li; Adam R. Renslo; Kartika Widjaja; Casper Wong; Kurt Giles; Yevgeniy Freyman; Manuel Elepano; John J. Irwin; Matthew P. Jacobson; Stanley B. Prusiner

PURPOSE Previous studies showed that lowering PrP(C) concomitantly reduced PrP(Sc) in the brains of mice inoculated with prions. We aimed to develop assays that measure PrP(C) on the surface of human T98G glioblastoma and IMR32 neuroblastoma cells. Using these assays, we sought to identify chemical hits, confirmed hits, and scaffolds that potently lowered PrP(C) levels in human brains cells, without lethality, and that could achieve drug concentrations in the brain after oral or intraperitoneal dosing in mice. METHODS We utilized HTS ELISA assays to identify small molecules that lower PrP(C) levels by ≥30% on the cell surface of human glioblastoma (T98G) and neuroblastoma (IMR32) cells. RESULTS From 44,578 diverse chemical compounds tested, 138 hits were identified by single point confirmation (SPC) representing 7 chemical scaffolds in T98G cells, and 114 SPC hits representing 6 scaffolds found in IMR32 cells. When the confirmed SPC hits were combined with structurally related analogs, >300 compounds (representing 6 distinct chemical scaffolds) were tested for dose-response (EC₅₀) in both cell lines, only studies in T98G cells identified compounds that reduced PrP(C) without killing the cells. EC₅₀ values from 32 hits ranged from 65 nM to 4.1 μM. Twenty-eight were evaluated in vivo in pharmacokinetic studies after a single 10 mg/kg oral or intraperitoneal dose in mice. Our results showed brain concentrations as high as 16.2 μM, but only after intraperitoneal dosing. CONCLUSIONS Our studies identified leads for future studies to determine which compounds might lower PrP(C) levels in rodent brain, and provide the basis of a therapeutic for fatal disorders caused by PrP prions.


Bioorganic & Medicinal Chemistry | 2013

Antiprion compounds that reduce PrPSc levels in dividing and stationary-phase cells

B. Michael Silber; Joel R. Gever; Zhe Li; Alejandra Gallardo-Godoy; Adam R. Renslo; Kartika Widjaja; John J. Irwin; Satish Rao; Matthew P. Jacobson; Sina Ghaemmaghami; Stanley B. Prusiner

During prion diseases, a normally benign, host protein, denoted PrP(C), undergoes alternative folding into the aberrant isoform, PrP(Sc). We used ELISA to identify and confirm hits in order to develop leads that reduce PrP(Sc) in prion-infected dividing and stationary-phase mouse neuroblastoma (ScN2a-cl3) cells. We tested 52,830 diverse small molecules in dividing cells and 49,430 in stationary-phase cells. This led to 3100 HTS and 970 single point confirmed (SPC) hits in dividing cells, 331 HTS and 55 confirmed SPC hits in stationary-phase cells as well as 36 confirmed SPC hits active in both. Fourteen chemical leads were identified from confirmed SPC hits in dividing cells and three in stationary-phase cells. From more than 682 compounds tested in concentration-effect relationships in dividing cells to determine potency (EC50), 102 had EC50 values between 1 and 10 μM and 50 had EC50 values of <1 μM; none affected cell viability. We observed an excellent correlation between EC50 values determined by ELISA and Western immunoblotting for 28 representative compounds in dividing cells (R(2)=0.75; p <0.0001). Of the 55 confirmed SPC hits in stationary-phase cells, 23 were piperazine, indole, or urea leads. The EC50 values of one indole in stationary-phase and dividing ScN2a-cl3 cells were 7.5 and 1.6 μM, respectively. Unexpectedly, the number of hits in stationary-phase cells was ~10% of that in dividing cells. The explanation for this difference remains to be determined.


Annals of Internal Medicine | 2009

Toward a 21st-Century Health Care System: Recommendations for Health Care Reform

Kenneth J. Arrow; Alan J. Auerbach; John Bertko; Shannon Brownlee; Lawrence P. Casalino; Jim Cooper; Francis J. Crosson; Alain C. Enthoven; Elizabeth Falcone; Robert C. Feldman; Victor R. Fuchs; Alan M. Garber; Marthe R. Gold; Dana P. Goldman; Gillian K. Hadfield; Mark A. Hall; Ralph I. Horwitz; Michael Hooven; Peter D. Jacobson; Timothy Stoltzfus Jost; Lawrence Kotlikoff; Jonathan Levin; Sharon Levine; Richard M. Levy; Karen Linscott; Harold S. Luft; Robert Mashal; Daniel McFadden; David Mechanic; David O. Meltzer


Pharmaceutical Research | 2013

Pharmacokinetics and Metabolism of 2-Aminothiazoles with Antiprion Activity in Mice

B. Michael Silber; Satish Rao; Kimberly L. Fife; Alejandra Gallardo-Godoy; Adam R. Renslo; Deepak Dalvie; Kurt Giles; Yevgeniy Freyman; Manuel Elepano; Joel R. Gever; Zhe Li; Matthew P. Jacobson; Yong Huang; Leslie Z. Benet; Stanley B. Prusiner


ACS Medicinal Chemistry Letters | 2013

Towards optimization of arylamides as novel, potent, and brain-penetrant antiprion lead compounds

Zhe Li; Satish Rao; Joel R. Gever; Kartika Widjaja; Stanley B. Prusiner; B. Michael Silber

Collaboration


Dive into the B. Michael Silber's collaboration.

Top Co-Authors

Avatar

Joel R. Gever

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam R. Renslo

University of California

View shared research outputs
Top Co-Authors

Avatar

Zhe Li

University of California

View shared research outputs
Top Co-Authors

Avatar

Satish Rao

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kurt Giles

University of California

View shared research outputs
Top Co-Authors

Avatar

Manuel Elepano

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge