Network


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

Hotspot


Dive into the research topics where Brad D. Maxwell is active.

Publication


Featured researches published by Brad D. Maxwell.


Drug Metabolism and Disposition | 2010

In Vitro Assessment of Metabolic Drug-Drug Interaction Potential of Apixaban through Cytochrome P450 Phenotyping, Inhibition, and Induction Studies

Lifei Wang; Donglu Zhang; Nirmala Raghavan; Ming Yao; Li Ma; Charles A Frost; Brad D. Maxwell; Shiang-Yuan Chen; Kan He; Theunis C. Goosen; W. Griffith Humphreys; Scott J. Grossman

Apixaban is an oral, direct, and highly selective factor Xa inhibitor in late-stage clinical development for the prevention and treatment of thromboembolic diseases. The metabolic drug-drug interaction potential of apixaban was evaluated in vitro. The compound did not show cytochrome P450 inhibition (IC50 values >20 μM) in incubations of human liver microsomes with the probe substrates of CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6, or 3A4/5. Apixaban did not show any effect at concentrations up to 20 μM on enzyme activities or mRNA levels of selected P450 enzymes (CYP1A2, 2B6, and 3A4/5) that are sensitive to induction in incubations with primary human hepatocytes. Apixaban showed a slow metabolic turnover in incubations of human liver microsomes with formation of O-demethylation (M2) and hydroxylation products (M4 and M7) as prominent in vitro metabolites. Experiments with human cDNA-expressed P450 enzymes and P450 chemical inhibitors and correlation with P450 activities in individual human liver microsomes demonstrated that the oxidative metabolism of apixaban for formation of all metabolites was predominantly catalyzed by CYP3A4/5 with a minor contribution of CYP1A2 and CYP2J2 for formation of M2. The contribution of CYP2C8, 2C9, and 2C19 to metabolism of apixaban was less significant. In addition, a human absorption, distribution, metabolism, and excretion study showed that more than half of the dose was excreted as unchanged parent (fm CYP <0.5), thus significantly reducing the overall metabolic drug-drug interaction potential of apixaban. Together with a low clinical efficacious concentration and multiple clearance pathways, these results demonstrate that the metabolic drug-drug interaction potential between apixaban and coadministered drugs is low.


Science | 2016

A general alkyl-alkyl cross-coupling enabled by redox-active esters and alkylzinc reagents

Tian Qin; Josep Cornella; Chao Li; Lara R. Malins; Jacob T. Edwards; Shuhei Kawamura; Brad D. Maxwell; Martin D. Eastgate; Phil S. Baran

Carbon links without helpful neighbors Its an irony of modern organic chemistry that the simplest-looking carbon-carbon bonds are often the hardest to make. Most reactions owe their efficiency to neighboring double bonds or oxygen and nitrogen atoms that linger in the products. Qin et al. now present a broadly applicable protocol for making C-C bonds in the absence of such surrounding help. The nickel-catalyzed process couples a zinc-activated carbon center to an ester thats poised to lose CO2. The ready availability of numerous carboxylic acids (which are easily converted to esters) contributes to the reactions versatility. Science, this issue p. 801 A versatile nickel-catalyzed reaction forms carbon–carbon bonds, with no need for adjacent functionality in the product. Alkyl carboxylic acids are ubiquitous in all facets of chemical science, from natural products to polymers, and represent an ideal starting material with which to forge new connections. This study demonstrates how the same activating principles used for decades to make simple C–N (amide) bonds from carboxylic acids with loss of water can be used to make C–C bonds through coupling with dialkylzinc reagents and loss of carbon dioxide. This disconnection strategy benefits from the use of a simple, inexpensive nickel catalyst and exhibits a remarkably broad scope across a range of substrates (>70 examples).


Drug Metabolism and Disposition | 2009

Comparative Metabolism of 14C-Labeled Apixaban in Mice, Rats, Rabbits, Dogs, and Humans

Donglu Zhang; Kan He; Nirmala Raghavan; Lifei Wang; James Mitroka; Brad D. Maxwell; Robert M. Knabb; Charles A Frost; Alan Schuster; Feng Hao; Zheming Gu; W. Griffith Humphreys; Scott J. Grossman

The metabolism and disposition of [14C]apixaban, a potent, reversible, and direct inhibitor of coagulation factor Xa, were investigated in mice, rats, rabbits, dogs, and humans after a single oral administration and in incubations with hepatocytes. In plasma, the parent compound was the major circulating component in mice, rats, dogs, and humans. O-Demethyl apixaban sulfate (M1) represented approximately 25% of the parent area under the time curve in human plasma. This sulfate metabolite was present, but in lower amounts relative to the parent, in plasma from mice, rats, and dogs. Rabbits showed a plasma metabolite profile distinct from that of other species with apixaban as a minor component and M2 (O-demethyl apixaban) and M14 (O-demethyl apixaban glucuronide) as prominent components. The fecal route was a major elimination pathway, accounting for >54% of the dose in animals and >46% in humans. The urinary route accounted for <15% of the dose in animals and 25 to 28% in humans. Apixaban was the major component in feces of every species and in urine of all species except rabbit. M1 and M2 were common prominent metabolites in urine and feces of all species as well as in bile of rats and humans. In vivo metabolite profiles showed quantitative differences between species and from in vitro metabolite profiles, but all human metabolites were found in animal species. After intravenous administration of [14C]apixaban to bile duct-cannulated rats, the significant portion (approximately 22%) of the dose was recovered as parent drug in the feces, suggesting direct excretion of the drug from gastrointestinal tracts of rats. Overall, apixaban was effectively eliminated via multiple elimination pathways in animals and humans, including oxidative metabolism, and direct renal and intestinal excretion.


Journal of the American Chemical Society | 2015

Hydromethylation of Unactivated Olefins

Hai T. Dao; Chao Li; Quentin Michaudel; Brad D. Maxwell; Phil S. Baran

A solution to the classic unsolved problem of olefin hydromethylation is presented. This highly chemoselective method can tolerate labile and reactive chemical functionalities and uses a simple set of reagents. An array of olefins, including mono-, di-, and trisubstituted olefins, are all smoothly hydromethylated. This mild protocol can be used to simplify the synthesis of a specific target or to directly “edit” complex natural products and other advanced materials. The method is also amenable to the simple installation of radioactive and stable labeled methyl groups.


Drug Metabolism and Disposition | 2011

Tissue Distribution and Elimination of [14C]Apixaban in Rats

Lifei Wang; Kan He; Brad D. Maxwell; Scott J. Grossman; Larry M. Tremaine; W. Griffith Humphreys; Donglu Zhang

Apixaban, a potent and highly selective factor Xa inhibitor, is currently under development for treatment of arterial and venous thrombotic diseases. The distribution, metabolism, and elimination of [14C]apixaban were investigated in male, female, pregnant, and lactating rats after single oral doses. Tissue distribution of radioactivity in rats was measured using quantitative whole-body autoradiography. After a single oral administration, radioactivity distributed quickly in rats with Cmax at 1 h for most tissues. The elimination t1/2 of radioactivity in blood was 1.7 to 4.2 h. The blood area under the plasma concentration-time curve of radioactivity was similar between male and female rats and was slightly higher in pregnant rats and lower in lactating rats. The radioactivity concentration in tissues involved in elimination was greater than that in blood with the highest concentration in the gastrointestinal tract, liver, and urinary bladder/contents and lowest level in brains. In pregnant rats, the whole-body autoradiogram showed that low levels of radioactivity were present in fetal blood, liver, and kidney and were much lower than the radioactivity in the respective maternal organs. The fecal route was the major pathway (74% of dose), and the urinary route was the minor pathway (14%) for apixaban elimination. After single oral doses of [14C]apixaban to lactating rats, apixaban exhibited extensive lacteal excretion with apixaban as the major component. In summary, tissue distribution of apixaban in rats was extensive but with limited transfer to fetal and brain tissues and extensive secretion into rat milk with the parent drug as the major component. Milk excretion could account for 10% of apixaban dose, which was comparable to urinary elimination in rats. Tissue distribution and drug excretion of apixaban are consistent with those for a moderately permeable drug that is a substrate for P-glycoprotein and breast cancer resistance protein efflux transporters.


Journal of Medicinal Chemistry | 2014

Identification of a nonbasic melanin hormone receptor 1 antagonist as an antiobesity clinical candidate.

William N. Washburn; Mark Manfredi; Pratik Devasthale; Guohua Zhao; Saleem Ahmad; Andres Hernandez; Jeffrey A. Robl; Wei Wang; James Mignone; Zhenghua Wang; Khehyong Ngu; Mary Ann Pelleymounter; Daniel Longhi; Rulin Zhao; Bei Wang; Ning Huang; Neil Flynn; Anthony V. Azzara; Joel C. Barrish; Kenneth Rohrbach; James Devenny; Michael J. Thomas; Susan Glick; Helen E. Godonis; Susan J. Harvey; Mary Jane Cullen; Hongwei Zhang; Christian Caporuscio; Paul Stetsko; Mary F. Grubb

Identification of MCHR1 antagonists with a preclinical safety profile to support clinical evaluation as antiobesity agents has been a challenge. Our finding that a basic moiety is not required for MCHR1 antagonists to achieve high affinity allowed us to explore structures less prone to off-target activities such as hERG inhibition. We report the SAR evolution of hydroxylated thienopyrimidinone ethers culminating in the identification of 27 (BMS-819881), which entered obesity clinical trials as the phosphate ester prodrug 35 (BMS-830216).


Journal of Medicinal Chemistry | 2017

Discovery of a Parenteral Small Molecule Coagulation Factor XIa Inhibitor Clinical Candidate (BMS-962212)

Donald J. P. Pinto; Michael J. Orwat; Leon M Smith; Mimi L. Quan; Patrick Y. S. Lam; Karen A. Rossi; Atsu Apedo; Jeffrery M. Bozarth; Yiming Wu; Joanna J. Zheng; Baomin Xin; Nathalie Toussaint; Paul Stetsko; Olafur S. Gudmundsson; Brad D. Maxwell; Earl J. Crain; Pancras C. Wong; Zhen Lou; Timothy W. Harper; Silvi A. Chacko; Joseph E. Myers; Steven Sheriff; Huiping Zhang; Xiaoping Hou; Arvind Mathur; Dietmar A. Seiffert; Ruth R. Wexler; Joseph M. Luettgen; William R. Ewing

Factor XIa (FXIa) is a blood coagulation enzyme that is involved in the amplification of thrombin generation. Mounting evidence suggests that direct inhibition of FXIa can block pathologic thrombus formation while preserving normal hemostasis. Preclinical studies using a variety of approaches to reduce FXIa activity, including direct inhibitors of FXIa, have demonstrated good antithrombotic efficacy without increasing bleeding. On the basis of this potential, we targeted our efforts at identifying potent inhibitors of FXIa with a focus on discovering an acute antithrombotic agent for use in a hospital setting. Herein we describe the discovery of a potent FXIa clinical candidate, 55 (FXIa Ki = 0.7 nM), with excellent preclinical efficacy in thrombosis models and aqueous solubility suitable for intravenous administration. BMS-962212 is a reversible, direct, and highly selective small molecule inhibitor of FXIa.


Journal of Organic Chemistry | 2015

Synthesis of Biologically Active Piperidine Metabolites of Clopidogrel: Determination of Structure and Analyte Development

Scott A. Shaw; Balu Balasubramanian; Samuel J. Bonacorsi; Janet Caceres Cortes; Kevin Cao; Bang-Chi Chen; Jun Dai; Carl P. Decicco; Animesh Goswami; Zhiwei Guo; Ronald L. Hanson; W. Griffith Humphreys; Patrick Y. S. Lam; Wenying Li; Arvind Mathur; Brad D. Maxwell; Quentin Michaudel; Li Peng; Andrew T. Pudzianowski; Feng Qiu; Shun Su; Dawn Sun; Adrienne A. Tymiak; Benjamin P. Vokits; Bei Wang; Ruth R. Wexler; Dauh-Rurng Wu; Yingru Zhang; Rulin Zhao; Phil S. Baran

Clopidogrel is a prodrug anticoagulant with active metabolites that irreversibly inhibit the platelet surface GPCR P2Y12 and thus inhibit platelet activation. However, gaining an understanding of patient response has been limited due to imprecise understanding of metabolite activity and stereochemistry, and a lack of acceptable analytes for quantifying in vivo metabolite formation. Methods for the production of all bioactive metabolites of clopidogrel, their stereochemical assignment, and the development of stable analytes via three conceptually orthogonal routes are disclosed.


Journal of Medicinal Chemistry | 2017

Discovery of Pyrrolidine-Containing GPR40 Agonists: Stereochemistry Effects a Change in Binding Mode

Elizabeth A. Jurica; Ximao Wu; Kristin N. Williams; Andres S. Hernandez; David S. Nirschl; Richard Rampulla; Arvind Mathur; Min Zhou; Gary Cao; Chunshan Xie; Biji Jacob; Hong Cai; Tao Wang; Brian J. Murphy; Heng Liu; Carrie Xu; Lori Kunselman; Michael B. Hicks; Qin Sun; Dora M. Schnur; Doree Sitkoff; Elizabeth A. Dierks; Atsu Apedo; Douglas B. Moore; Kimberly A. Foster; Mary Ellen Cvijic; Reshma Panemangalore; Neil Flynn; Brad D. Maxwell; Yang Hong

A novel series of pyrrolidine-containing GPR40 agonists is described as a potential treatment for type 2 diabetes. The initial pyrrolidine hit was modified by moving the position of the carboxylic acid, a key pharmacophore for GPR40. Addition of a 4-cis-CF3 to the pyrrolidine improves the human GPR40 binding Ki and agonist efficacy. After further optimization, the discovery of a minor enantiomeric impurity with agonist activity led to the finding that enantiomers (R,R)-68 and (S,S)-68 have differential effects on the radioligand used for the binding assay, with (R,R)-68 potentiating the radioligand and (S,S)-68 displacing the radioligand. Compound (R,R)-68 activates both Gq-coupled intracellular Ca2+ flux and Gs-coupled cAMP accumulation. This signaling bias results in a dual mechanism of action for compound (R,R)-68, demonstrating glucose-dependent insulin and GLP-1 secretion in vitro. In vivo, compound (R,R)-68 significantly lowers plasma glucose levels in mice during an oral glucose challenge, encouraging further development of the series.


Journal of Labelled Compounds and Radiopharmaceuticals | 2013

The synthesis of a carbon-14 labeled pegylated Adnectin™ for placental transfer studies in guinea pigs†

Brad D. Maxwell; Kai Cao; Samuel J. Bonacorsi; Lakshmi Sivaraman; Bonnie Wang; MaryEllen McNerney; Lifei Wang; Lisa J. Christopher; Nga Kit Eliza Fung; Jian Wang; Mark E. Arnold

Adnectins™ are novel fibronectin-based proteins containing domains engineered to bind to targets of therapeutic interest. The molecular weights of adnectins are less than conventional monoclonal antibodies but larger than traditional small molecules. Until now, there has been no information on the placental transfer of adnectins. To assess placental permeability to adnectins in pregnant guinea pigs, a radiolabeled adnectin, ATI-1072, bound to polyethylene glycol through a [(14) C]Maleimide linker, was synthesized from [1,4-(14) C]Maleic acid. This publication describes the synthesis and analysis of PEG-[(14) C]Maleimide-adnectin ([(14) C]ATI-1072).

Collaboration


Dive into the Brad D. Maxwell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kai Cao

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge