Network


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

Hotspot


Dive into the research topics where Angel Guzman-Perez is active.

Publication


Featured researches published by Angel Guzman-Perez.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Proline-rich tyrosine kinase 2 regulates osteoprogenitor cells and bone formation, and offers an anabolic treatment approach for osteoporosis

Leonard Buckbinder; D.T. Crawford; Hong Qi; Hua Zhu Ke; Lisa M. Olson; Kelly R. Long; Peter C. Bonnette; Amy Baumann; John E. Hambor; William A. Grasser; Lydia Codetta Pan; Thomas A. Owen; Michael Joseph Luzzio; Catherine A. Hulford; David Gebhard; Vishwas M. Paralkar; H.A. Simmons; John Charles Kath; W. Gregory Roberts; Steven L. Smock; Angel Guzman-Perez; Thomas A. Brown; Mei Li

Bone is accrued and maintained primarily through the coupled actions of bone-forming osteoblasts and bone-resorbing osteoclasts. Cumulative in vitro studies indicated that proline-rich tyrosine kinase 2 (PYK2) is a positive mediator of osteoclast function and activity. However, our investigation of PYK2−/− mice did not reveal evidence supporting an essential function for PYK2 in osteoclasts either in vivo or in culture. We find that PYK2−/− mice have high bone mass resulting from an unexpected increase in bone formation. Consistent with the in vivo findings, mouse bone marrow cultures show that PYK2 deficiency enhances differentiation and activity of osteoprogenitor cells, as does expressing a PYK2-specific short hairpin RNA or dominantly interfering proteins in human mesenchymal stem cells. Furthermore, the daily administration of a small-molecule PYK2 inhibitor increases bone formation and protects against bone loss in ovariectomized rats, an established preclinical model of postmenopausal osteoporosis. In summary, we find that PYK2 regulates the differentiation of early osteoprogenitor cells across species and that inhibitors of the PYK2 have potential as a bone anabolic approach for the treatment of osteoporosis.


Journal of Medicinal Chemistry | 2012

Discovery of (S)-6-(3-Cyclopentyl-2-(4-(trifluoromethyl)-1H-imidazol-1-yl)propanamido)nicotinic Acid as a Hepatoselective Glucokinase Activator Clinical Candidate for Treating Type 2 Diabetes Mellitus

Jeffrey A. Pfefferkorn; Angel Guzman-Perez; John Litchfield; Robert J. Aiello; Judith L. Treadway; John C. Pettersen; Martha L. Minich; Kevin J. Filipski; Christopher S. Jones; Meihua Tu; Gary E. Aspnes; Hud Risley; Jianwei Bian; Benjamin D. Stevens; Patricia Bourassa; Theresa D’Aquila; Levenia Baker; Nicole Barucci; Alan Robertson; Francis Bourbonais; David R. Derksen; Margit MacDougall; Over Cabrera; Jing Chen; Amanda Lee Lapworth; James A. Landro; William J. Zavadoski; Karen Atkinson; Nahor Haddish-Berhane; Beijing Tan

Glucokinase is a key regulator of glucose homeostasis, and small molecule allosteric activators of this enzyme represent a promising opportunity for the treatment of type 2 diabetes. Systemically acting glucokinase activators (liver and pancreas) have been reported to be efficacious but in many cases present hypoglycaemia risk due to activation of the enzyme at low glucose levels in the pancreas, leading to inappropriately excessive insulin secretion. It was therefore postulated that a liver selective activator may offer effective glycemic control with reduced hypoglycemia risk. Herein, we report structure-activity studies on a carboxylic acid containing series of glucokinase activators with preferential activity in hepatocytes versus pancreatic β-cells. These activators were designed to have low passive permeability thereby minimizing distribution into extrahepatic tissues; concurrently, they were also optimized as substrates for active liver uptake via members of the organic anion transporting polypeptide (OATP) family. These studies lead to the identification of 19 as a potent glucokinase activator with a greater than 50-fold liver-to-pancreas ratio of tissue distribution in rodent and non-rodent species. In preclinical diabetic animals, 19 was found to robustly lower fasting and postprandial glucose with no hypoglycemia, leading to its selection as a clinical development candidate for treating type 2 diabetes.


Bioorganic & Medicinal Chemistry Letters | 2001

Discovery of zoniporide: A potent and selective sodium–hydrogen exchanger type 1 (NHE-1) inhibitor with high aqueous solubility

Angel Guzman-Perez; Ronald Thure Wester; Mary C. Allen; Janice A. Brown; Allan R. Buchholz; Ewell R. Cook; Wesley W. Day; Ernest Seiichi Hamanaka; Scott P. Kennedy; Delvin R. Knight; Paul J. Kowalczyk; Ravi B. Marala; Christian J. Mularski; William Albert Novomisle; Roger Benjamin Ruggeri; W. Ross Tracey; Roger J. Hill

Zoniporide (CP-597,396) is a potent and selective inhibitor of NHE-1, which exhibits high aqueous solubility and acceptable pharmacokinetics for intravenous administration. The discovery, synthesis, activities, and rat and dog pharmacokinetics of this compound are presented. The potency and selectivity of zoniporide may be due to the conformation that the molecule adopts due to the presence of a cyclopropyl and a 5-quinolinyl substituent on the central pyrazole ring of the molecule.


ACS Medicinal Chemistry Letters | 2011

Discovery of PF-04620110, a Potent, Selective, and Orally Bioavailable Inhibitor of DGAT-1.

Robert L. Dow; Jian-Cheng Li; Michael P. Pence; E. Michael Gibbs; Jennifer L. LaPerle; John Litchfield; David W. Piotrowski; Michael John Munchhof; Tara B. Manion; William J. Zavadoski; Gregory S. Walker; R. Kirk McPherson; Susan Tapley; Eliot Sugarman; Angel Guzman-Perez; Paul DaSilva-Jardine

Acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1) catalyzes the final committed step in the biosynthesis of triglycerides. DGAT-1 knockout mice have been shown to be resistant to diet-induced obesity and have increased insulin sensitivity. Thus, inhibition of DGAT-1 may represent an attractive target for the treatment of obesity or type II diabetes. Herein, we report the discovery and characterization of a potent and selective DGAT-1 inhibitor PF-04620110 (3). Compound 3 inhibits DGAT-1 with an IC50 of 19 nM and shows high selectivity versus a broad panel of off-target pharmacologic end points. In vivo DGAT-1 inhibition has been demonstrated through reduction of plasma triglyceride levels in rodents at doses of ≥0.1 mg/kg following a lipid challenge. On the basis of this pharmacologic and pharmacokinetic profile, compound 3 has been advanced to human clinical studies.


Bioorganic & Medicinal Chemistry Letters | 2008

Trifluoromethylpyrimidine-based inhibitors of proline-rich tyrosine kinase 2 (PYK2): structure-activity relationships and strategies for the elimination of reactive metabolite formation.

Daniel P. Walker; F. Christopher Bi; Amit S. Kalgutkar; Jonathan N. Bauman; Sabrina X. Zhao; John R. Soglia; Gary E. Aspnes; Daniel W. Kung; Jacquelyn Klug-McLeod; Michael P. Zawistoski; Molly A. McGlynn; Robert M. Oliver; Matthew Francis Dunn; Jian-Cheng Li; Daniel T. Richter; Beth Cooper; John Charles Kath; Catherine A. Hulford; Christopher Autry; Michael Joseph Luzzio; Ethan Ung; W. Gregory Roberts; Peter C. Bonnette; Leonard Buckbinder; Anil Mistry; Matthew C. Griffor; Seungil Han; Angel Guzman-Perez

The synthesis and SAR for a series of diaminopyrimidines as PYK2 inhibitors are described. Using a combination of library and traditional medicinal chemistry techniques, a FAK-selective chemical series was transformed into compounds possessing good PYK2 potency and 10- to 20-fold selectivity against FAK. Subsequent studies found that the majority of the compounds were positive in a reactive metabolite assay, an indicator for potential toxicological liabilities. Based on the proposed mechanism for bioactivation, as well as a combination of structure-based drug design and traditional medicinal chemistry techniques, a follow-up series of PYK2 inhibitors was identified that maintained PYK2 potency, FAK selectivity and HLM stability, yet were negative in the RM assay.


MedChemComm | 2011

Designing glucokinase activators with reduced hypoglycemia risk: discovery of N,N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as a clinical candidate for the treatment of type 2 diabetes mellitus

Jeffrey A. Pfefferkorn; Angel Guzman-Perez; Peter J. Oates; John Litchfield; Gary E. Aspnes; Arindrajit Basak; John William Benbow; Martin A. Berliner; Jianwei Bian; Chulho Choi; Kevin Daniel Freeman-Cook; Jeffrey W. Corbett; Mary Theresa Didiuk; Joshua R. Dunetz; Kevin J. Filipski; William M. Hungerford; Christopher S. Jones; Kapil Karki; Anthony Lai Ling; Jian-Cheng Li; Leena Patel; Christian Perreault; Hud Risley; James Saenz; Wei Song; Meihua Tu; Robert J. Aiello; Karen Atkinson; Nicole Barucci; David A. Beebe

Glucokinase is a key regulator of glucose homeostasis and small molecule activators of this enzyme represent a promising opportunity for the treatment of Type 2 diabetes. Several glucokinase activators have advanced to clinical studies and demonstrated promising efficacy; however, many of these early candidates also revealed hypoglycemia as a key risk. In an effort to mitigate this hypoglycemia risk while maintaining the promising efficacy of this mechanism, we have investigated a series of substituted 2-methylbenzofurans as “partial activators” of the glucokinase enzyme leading to the identification of N,N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as an early development candidate.


Bioorganic & Medicinal Chemistry Letters | 2009

Sulfoximine-substituted trifluoromethylpyrimidine analogs as inhibitors of proline-rich tyrosine kinase 2 (PYK2) show reduced hERG activity

Daniel P. Walker; Michael P. Zawistoski; Molly A. McGlynn; Jian-Cheng Li; Daniel W. Kung; Peter C. Bonnette; Amy Baumann; Leonard Buckbinder; Janet A. Houser; Jason Boer; Anil Mistry; Seungil Han; Li Xing; Angel Guzman-Perez

The synthesis, in vitro properties, and in vivo pharmacokinetics for a series of sulfoximine-substituted trifluoromethylpyrimidines as inhibitors of proline-rich tyrosine kinase, a target for the possible treatment of osteoporosis, are described. These compounds were prepared as surrogates of the corresponding sulfone compound 1. Sulfone 1 was an attractive PYK2 lead compound; however, subsequent studies determined this compound possessed high dofetilide binding, which is an early indicator of cardiovascular safety. Surprisingly, the corresponding sulfoximine analogs displayed significantly lower dofetilide binding, which, for N-methylsulfoximine (S)-14a, translated to lower activity in a patch clamp hERG K(+) ion channel screen. In addition, compound (S)-14a shows good oral exposure in a rat pharmacokinetic model.


Organic Letters | 2010

Functionalization of aromatic amino acids via direct C-H activation: generation of versatile building blocks for accessing novel peptide space.

Falco-Magnus Meyer; Spiros Liras; Angel Guzman-Perez; Christian Perreault; Jianwei Bian; Keith James

Functionalized alpha-amino acid building blocks have been prepared in good yield with high regiocontrol and preservation of stereochemistry via iridium-catalyzed borylation of suitably protected aromatic alpha-amino acid derivatives. The utility of these systems in peptide couplings and Suzuki reactions has been demonstrated.


Journal of Medicinal Chemistry | 2013

Discovery of Novel, Induced-Pocket Binding Oxazolidinones as Potent, Selective, and Orally Bioavailable Tankyrase Inhibitors

Howard Bregman; Nagasree Chakka; Angel Guzman-Perez; Hakan Gunaydin; Yan Gu; Xin Huang; Virginia Berry; Jingzhou Liu; Yohannes Teffera; Liyue Huang; Bryan Egge; Erin L. Mullady; Steve Schneider; Paul S. Andrews; Ankita Mishra; John Newcomb; Randy Serafino; Craig A. Strathdee; Susan M. Turci; Cindy Wilson; Erin F. DiMauro

Tankyrase (TNKS) is a poly-ADP-ribosylating protein (PARP) whose activity suppresses cellular axin protein levels and elevates β-catenin concentrations, resulting in increased oncogene expression. The inhibition of tankyrase (TNKS1 and 2) may reduce the levels of β-catenin-mediated transcription and inhibit tumorigenesis. Compound 1 is a previously described moderately potent tankyrase inhibitor that suffers from poor pharmacokinetic properties. Herein, we describe the utilization of structure-based design and molecular modeling toward novel, potent, and selective tankyrase inhibitors with improved pharmacokinetic properties (39, 40).


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.

Collaboration


Dive into the Angel Guzman-Perez's collaboration.

Researchain Logo
Decentralizing Knowledge