Network


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

Hotspot


Dive into the research topics where Brian Samas is active.

Publication


Featured researches published by Brian Samas.


International Journal of Pharmaceutics | 2003

An intravenous formulation decision tree for discovery compound formulation development.

Yung-Chi Lee; Philip Zocharski; Brian Samas

Discovery and pre-clinical animal efficacy assessment formulation development efforts are challenged by limited compound availability and stringent timelines. The implementation and use of a systematic discovery formulation scheme can facilitate this important process. We observed that nearly 85% of Pfizer, Ann Arbor discovery compounds (n>300) submitted for discovery and pre-clinical injectable formulation development in the year 2000 could be formulated by pH adjustment, cosolvent addition, or a combination of the two approaches. Based on the vehicle data generated by this laboratory, a discovery formulation decision tree, that utilizes the solubilization approaches described above, is proposed. The proposed decision tree can be adapted and modified by pharmaceutical scientists to conform to best practices put forth by their institutions for discovery animal studies requiring injectable dosage forms.


Journal of Medicinal Chemistry | 2011

Discovery of a Clinical Candidate from the Structurally Unique Dioxa-bicyclo[3.2.1]octane Class of Sodium-Dependent Glucose Cotransporter 2 Inhibitors

Vincent Mascitti; Tristan S. Maurer; Ralph P. Robinson; Jianwei Bian; Carine M. Boustany-Kari; Thomas A. Brandt; Benjamin Micah Collman; Amit S. Kalgutkar; Michelle K. Klenotic; Michael T. Leininger; André Lowe; Robert John Maguire; Victoria M. Masterson; Zhuang Miao; Emi Mukaiyama; Jigna D. Patel; John C. Pettersen; Cathy Préville; Brian Samas; Li She; Zhanna Sobol; Claire M. Steppan; Benjamin D. Stevens; Benjamin A. Thuma; Meera Tugnait; Dongxiang Zeng; Tong Zhu

Compound 4 (PF-04971729) belongs to a new class of potent and selective sodium-dependent glucose cotransporter 2 inhibitors incorporating a unique dioxa-bicyclo[3.2.1]octane (bridged ketal) ring system. In this paper we present the design, synthesis, preclinical evaluation, and human dose predictions related to 4. This compound demonstrated robust urinary glucose excretion in rats and an excellent preclinical safety profile. It is currently in phase 2 clinical trials and is being evaluated for the treatment of type 2 diabetes.


Organic Letters | 2009

A highly enantioselective allylic amination reaction using a commercially available chiral rhodium catalyst: resolution of racemic allylic carbonates.

Derek Vrieze; Garrett S. Hoge; Perrine Z. Hoerter; Jared T. Van Haitsma; Brian Samas

A novel method for the kinetic resolution of unsymmetrical acyclic allylic carbonates and the concurrent synthesis of enantioenriched secondary amines using a commercially available chiral catalyst is disclosed.


Bioorganic & Medicinal Chemistry Letters | 2013

The design and synthesis of a potent glucagon receptor antagonist with favorable physicochemical and pharmacokinetic properties as a candidate for the treatment of type 2 diabetes mellitus.

Angel Guzman-Perez; Jeffrey A. Pfefferkorn; Esther Cheng Yin Lee; Benjamin D. Stevens; Gary E. Aspnes; Jianwei Bian; Mary Theresa Didiuk; Kevin J. Filipski; Dianna E. Moore; Christian Perreault; Matthew F. Sammons; Meihua Tu; Janice A. Brown; Karen Atkinson; John Litchfield; Beijing Tan; Brian Samas; William J. Zavadoski; Christopher T. Salatto; Judith L. Treadway

A novel and potent small molecule glucagon receptor antagonist for the treatment of diabetes mellitus is reported. This candidate, (S)-3-[4-(1-{3,5-dimethyl-4-[4-(trifluoromethyl)-1H-pyrazol-1-yl]phenoxy}butyl)benzamido]propanoic acid, has lower molecular weight and lipophilicity than historical glucagon receptor antagonists, resulting in excellent selectivity in broad-panel screening, lower cytotoxicity, and excellent overall in vivo safety in early pre-clinical testing. Additionally, it displays low in vivo clearance and excellent oral bioavailability in both rats and dogs. In a rat glucagon challenge model, it was shown to reduce the glucagon-elicited glucose excursion in a dose-dependent manner and at a concentration consistent with its rat in vitro potency. Its properties make it an excellent candidate for further investigation.


Journal of Medicinal Chemistry | 2016

Identification of a Chemical Probe for Family VIII Bromodomains through Optimization of a Fragment Hit

Brian S. Gerstenberger; John David Trzupek; Cynthia Tallant; Oleg Fedorov; Panagis Filippakopoulos; Paul E. Brennan; Vita Fedele; Sarah Martin; Sarah Picaud; Catherine Rogers; Mihir D. Parikh; Alexandria P. Taylor; Brian Samas; Alison O’Mahony; Ellen Berg; Gabriel Pallares; Adam Torrey; Daniel Kelly Treiber; Ivan Samardjiev; Brian T. Nasipak; Teresita Padilla-Benavides; Qiong Wu; Anthony N. Imbalzano; Jeffrey A. Nickerson; Mark Edward Bunnage; Susanne Müller; Stefan Knapp; Dafydd R. Owen

The acetyl post-translational modification of chromatin at selected histone lysine residues is interpreted by an acetyl-lysine specific interaction with bromodomain reader modules. Here we report the discovery of the potent, acetyl-lysine-competitive, and cell active inhibitor PFI-3 that binds to certain family VIII bromodomains while displaying significant, broader bromodomain family selectivity. The high specificity of PFI-3 for family VIII was achieved through a novel bromodomain binding mode of a phenolic headgroup that led to the unusual displacement of water molecules that are generally retained by most other bromodomain inhibitors reported to date. The medicinal chemistry program that led to PFI-3 from an initial fragment screening hit is described in detail, and additional analogues with differing family VIII bromodomain selectivity profiles are also reported. We also describe the full pharmacological characterization of PFI-3 as a chemical probe, along with phenotypic data on adipocyte and myoblast cell differentiation assays.


ChemMedChem | 2012

Discovery and characterization of atropisomer PH-797804, a p38 MAP kinase inhibitor, as a clinical drug candidate.

Li Xing; Balekudru Devadas; Rajesh V. Devraj; Shaun R. Selness; Huey Shieh; John K. Walker; Michael Mao; Dean Messing; Brian Samas; Jerry Z. Yang; Gary D. Anderson; Elizabeth G. Webb; Joseph B. Monahan

PH‐797804 ((aS)‐3‐{3‐bromo‐4‐[(2,4‐difluorobenzyl)oxy]‐6‐methyl‐2‐oxopyridin‐1(2H)‐yl}‐N,4‐dimethylbenzamde) is a diarylpyridinone inhibitor of p38 mitogen‐activated protein (MAP) kinase derived from a racemic mixture as the more potent atropisomer (aS), first proposed by molecular modeling and subsequently confirmed by experiments. Due to steric constraints imposed by the pyridinone carbonyl group and the 6‐ and 6′‐methyl substituents of PH‐797804, rotation around the connecting bond of the pyridinone and the N‐phenyl ring is restricted. Density functional theory predicts a remarkably high rotational energy barrier of >30 kcal mol−1, corresponding to a half‐life of more than one hundred years at room temperature. This gives rise to discrete conformational spaces for the N‐phenylpyridinone group, and as a result, two atropic isomers that do not interconvert under ambient conditions. Molecular modeling studies predict that the two isomers should differ in their binding affinity for p38α kinase; whereas the atropic S (aS) isomer binds favorably, the opposite aR isomer incurs significant steric interference with p38α kinase. The two isomers were subsequently identified and separated by chiral chromatography. IC50 values from p38α kinase assays confirm that one atropisomer is >100‐fold more potent than the other. It was ultimately confirmed by small‐molecule X‐ray diffraction that the more potent atropisomer, PH‐797804, is the aS isomer of the racemic pair. Extensive pharmacological characterization supports that PH‐797804 carries most activity both in vitro and in vivo, and it has a stability profile compatible with oral formulation and delivery options.


Journal of Physical Chemistry A | 2011

Long, Directional Interactions in Cofacial Silicon Phthalocyanine Oligomers

Yang Yang; Brian Samas; Vance O. Kennedy; Dainius Macikenas; Brian L. Chaloux; Jacob A. Miller; Richard L. Speer; John D. Protasiewicz; A. Alan Pinkerton; Malcolm E. Kenney

Single crystal structures have been determined for the three cofacial, oxygen-bridged, silicon phthalocyanine oligomers, [((CH(3))(3)SiO)(2)(CH(3))SiO](SiPcO)(2-4)[Si(CH(3))(OSi(CH(3))(3))(2)], and for the corresponding monomer. The data for the oligomers give structural parameters for a matching set of three cofacial, oxygen-bridged silicon phthalocyanine oligomers for the first time. The staggering angles between the six adjacent cofacial ring pairs in the three oligomers are not in a random distribution nor in a cluster at the intuitively expected angle of 45° but rather are in two clusters, one at an angle of 15° and the other at an angle of 41°. These two clusters lead to the conclusion that long, directional interactions (LDI) exist between the adjacent ring pairs. An understanding of these interactions is provided by atoms-in-molecules (AIM) and reduced-density-gradient (RDG) studies. A survey of the staggering angles in other single-atom-bridged, cofacial phthalocyanine oligomers provides further evidence for the existence of LDI between cofacial phthalocyanine ring pairs in single-atom-bridged phthalocyanine oligomers.


Journal of Medicinal Chemistry | 2008

Rational design and synthesis of 4-((1R,2R)-2-hydroxycyclohexyl)-2(trifluoromethyl)benzonitrile (PF-998425), a novel, nonsteroidal androgen receptor antagonist devoid of phototoxicity for dermatological indications.

Jie Jack Li; Donna M. Iula; Maria N. Nguyen; Lain-Yen Hu; Danielle Dettling; Theodore R. Johnson; Daniel Y. Du; Veerabahu Shanmugasundaram; Jennifer A. Van Camp; Zhi Wang; William Glen Harter; Wen-Song Yue; Mark L. Boys; Kimberly Wade; Elena M. Drummond; Brian Samas; Bruce Allen Lefker; Garrett S. Hoge; Mark J. Lovdahl; Jeffrey Asbill; Matthew Carroll; Mary Ann Meade; Susan Ciotti; Theresa Krieger-Burke

4-((1 R,2 R)-2-Hydroxycyclohexyl)-2(trifluoromethyl)benzonitrile [PF-0998425, (-)- 6a] is a novel, nonsteroidal androgen receptor antagonist for sebum control and treatment of androgenetic alopecia. It is potent, selective, and active in vivo. The compound is rapidly metabolized systemically, thereby reducing the risk of unwanted systemic side effects due to its primary pharmacology. (-)- 6a was tested negative in the 3T3 NRU assay, validating our rationale that reduction of conjugation might reduce potential phototoxicity.


Journal of Medicinal Chemistry | 2017

Discovery of Clinical Candidate 1-{[(2S,3S,4S)-3-Ethyl-4-fluoro-5-oxopyrrolidin-2-yl]methoxy}-7-methoxyisoquinoline-6-carboxamide (PF-06650833), a Potent, Selective Inhibitor of Interleukin-1 Receptor Associated Kinase 4 (IRAK4), by Fragment-Based Drug Design

Katherine L. Lee; Catherine M. Ambler; David R. Anderson; Brian P. Boscoe; Andrea G Bree; Joanne Brodfuehrer; Jeanne S. Chang; Chulho Choi; Seung Won Chung; Kevin J. Curran; Jacqueline E. Day; Christoph Martin Dehnhardt; Ken Dower; Susan E. Drozda; Richard K. Frisbie; Lori Krim Gavrin; Joel Adam Goldberg; Seungil Han; Martin Hegen; David Hepworth; Heidi R. Hope; Satwik Kamtekar; Iain Kilty; Arthur Lee; Lih-Ling Lin; Frank Lovering; Michael Dennis Lowe; John Paul Mathias; Heidi M Morgan; Elizabeth Murphy

Through fragment-based drug design focused on engaging the active site of IRAK4 and leveraging three-dimensional topology in a ligand-efficient manner, a micromolar hit identified from a screen of a Pfizer fragment library was optimized to afford IRAK4 inhibitors with nanomolar potency in cellular assays. The medicinal chemistry effort featured the judicious placement of lipophilicity, informed by co-crystal structures with IRAK4 and optimization of ADME properties to deliver clinical candidate PF-06650833 (compound 40). This compound displays a 5-unit increase in lipophilic efficiency from the fragment hit, excellent kinase selectivity, and pharmacokinetic properties suitable for oral administration.


MedChemComm | 2013

On the importance of synthetic organic chemistry in drug discovery: reflections on the discovery of antidiabetic agent ertugliflozin

Vincent Mascitti; Benjamin A. Thuma; Aaron Smith; Ralph P. Robinson; Thomas A. Brandt; Amit S. Kalgutkar; Tristan S. Maurer; Brian Samas; Raman Sharma

The discovery of antidiabetic agent ertugliflozin is described. The compound belongs to a new class of SGLT2 inhibitors bearing a dioxa-bicyclo[3.2.1]octane motif. This article describes the critical role that organic synthesis played in both influencing our medicinal chemistry strategy and speeding up the progression of our program.

Collaboration


Dive into the Brian Samas's collaboration.

Researchain Logo
Decentralizing Knowledge