Network


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

Hotspot


Dive into the research topics where W. Gregory Roberts is active.

Publication


Featured researches published by W. Gregory Roberts.


Journal of Biological Chemistry | 2007

Cellular Characterization of a Novel Focal Adhesion Kinase Inhibitor

Jill K. Slack-Davis; Karen H. Martin; Robert W. Tilghman; Marcin P. Iwanicki; Ethan Ung; Christopher Autry; Michael Joseph Luzzio; Beth Cooper; John Charles Kath; W. Gregory Roberts; J. Thomas Parsons

Focal adhesion kinase (FAK) is a member of a family of non-receptor protein-tyrosine kinases that regulates integrin and growth factor signaling pathways involved in cell migration, proliferation, and survival. FAK expression is increased in many cancers, including breast and prostate cancer. Here we describe perturbation of adhesion-mediated signaling with a FAK inhibitor, PF-573,228. In vitro, this compound inhibited purified recombinant catalytic fragment of FAK with an IC50 of 4 nm. In cultured cells, PF-573,228 inhibited FAK phosphorylation on Tyr397 with an IC50 of 30–100 nm. Treatment of cells with concentrations of PF-573,228 that significantly decreased FAK Tyr397 phosphorylation failed to inhibit cell growth or induce apoptosis. In contrast, treatment with PF-573,228 inhibited both chemotactic and haptotactic migration concomitant with the inhibition of focal adhesion turnover. These studies show that PF-573,228 serves as a useful tool to dissect the functions of FAK in integrin-dependent signaling pathways in normal and cancer cells and forms the basis for the generation of compounds amenable for preclinical and patient trials.


Journal of Clinical Oncology | 2012

Safety, Pharmacokinetic, and Pharmacodynamic Phase I Dose-Escalation Trial of PF-00562271, an Inhibitor of Focal Adhesion Kinase, in Advanced Solid Tumors

Jeffrey R. Infante; D. Ross Camidge; Linda Mileshkin; Eric X. Chen; Rodney J. Hicks; Danny Rischin; Howard Fingert; Kristen J. Pierce; Huiping Xu; W. Gregory Roberts; S. Martin Shreeve; Howard A. Burris; Lillian L. Siu

PURPOSE PF-00562271 is a novel inhibitor of focal adhesion kinase (FAK). The objectives of this study were to identify the recommended phase II dose (RP2D) and assess safety and tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and antitumor activity of PF-00562271. PATIENTS AND METHODS Part 1 was a dose escalation without and with food. Part 2 enrolled specific tumor types in an expansion at the RP2D and also assessed the effect of PF-00562271 on single-dose midazolam PK in a subgroup of patients. RESULTS Ninety-nine patients (median age, 60 years; 98% with Eastern Cooperative Oncology Group performance status of 0 or 1) were treated in 12 fasting and three fed cohorts. The 125-mg twice-per-day fed dose was deemed the maximum-tolerated dose (MTD) and RP2D. Grade 3 dose-limiting toxicities included headache, nausea/vomiting, dehydration, and edema. Nausea was the most frequently observed toxicity (60% of patients, all grades 1 or 2 at RP2D). PF-00562271 exposure increased with increasing dose; serum concentration-time profiles showed characteristic nonlinear disposition. Steady-state exposures were reached within 1 week. On coadministration, geometric mean values of midazolam maximal observed serum concentration and area under the serum concentration-time curve increased by 60% and more than two-fold, respectively. Of 14 patients evaluable by [(18)F]fluorodeoxyglucose positron emission tomography in the expansion cohorts, seven metabolic responses were observed. With conventional imaging, 31 patients had stable disease at first restaging scans, and 15 of these remained stable for six or more cycles. CONCLUSION The MTD and RP2D of PF-00562271 is 125 mg twice per day with food. PF-00562271 displayed time- and dose-dependent nonlinear PK and is likely a potent CYP 3A inhibitor. This first-in-class study supports further investigation of FAK as a promising therapeutic target.


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.


Clinical Cancer Research | 2008

Focal Adhesion Kinase: Targeting Adhesion Signaling Pathways for Therapeutic Intervention

J. Thomas Parsons; Jill K. Slack-Davis; Robert W. Tilghman; W. Gregory Roberts

The tumor microenvironment plays a central role in cancer progression and metastasis. Within this environment, cancer cells respond to a host of signals including growth factors and chemotactic factors, as well as signals from adjacent cells, cells in the surrounding stroma, and signals from the extracellular matrix. Targeting the pathways that mediate many of these signals has been a major goal in the effort to develop therapeutics.


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.


Clinical Genitourinary Cancer | 2015

Long-Term Safety With Axitinib in Previously Treated Patients With Metastatic Renal Cell Carcinoma

Brian I. Rini; Bernard Escudier; Subramanian Hariharan; W. Gregory Roberts; Jamal Tarazi; Brad Rosbrook; Zena Askerova; Liza DeAnnuntis; Robert J. Motzer

BACKGROUND Axitinib is an approved treatment for advanced renal cell carcinoma (RCC) after failure of 1 systemic therapy. PATIENTS AND METHODS Long-term safety with single-agent axitinib was analyzed using pooled data from clinical trials in 672 previously treated patients with metastatic RCC (mRCC) and in 1304 patients with different advanced solid tumors. In all studies, except the phase I first-in-human, dose-finding study, the starting dose of oral axitinib was 5 mg twice daily continuously. Common long-term treatment-emergent adverse events (AEs) were identified in patients who received axitinib for ≥ 2 years, then evaluated in all patients, and assessed using interval, cumulative, and latency analyses. RESULTS In all, 108 (16%) previously treated patients with mRCC received axitinib for ≥ 2 years. In interval analysis, most AEs occurred during the first 6 months of treatment, with rates stable or decreased over time; rates increased for proteinuria, peripheral edema, and increased blood creatinine. Common Grade ≥ 3 AE rates declined or plateaued over time, except for increased amylase and myocardial infarction. Results were similar in cumulative analysis in this population, and in interval and cumulative analyses in all patients with mRCC and those with advanced solid tumors. CONCLUSION Declining or stable rates of most AEs support an acceptable long-term safety profile for axitinib in patients with mRCC. However, increases in the rates of some AEs warrant monitoring. This analysis is limited in that it was retrospective and included a relatively small patient population.


Cancer Research | 1997

Neovasculature Induced by Vascular Endothelial Growth Factor Is Fenestrated

W. Gregory Roberts; George E. Palade


Cancer Research | 1999

Tumor-derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function.

Jeremy Grunstein; W. Gregory Roberts; Odile Mathieu-Costello; Douglas Hanahan; Randall S. Johnson


Cancer Research | 2003

Pharmacological Characterization of CP-547,632, a Novel Vascular Endothelial Growth Factor Receptor-2 Tyrosine Kinase Inhibitor for Cancer Therapy

Jean Beebe; Jitesh P. Jani; Elisabeth Knauth; Peter Goodwin; Carla Higdon; Ann Marie Rossi; Erling O. Emerson; Martin Finkelstein; Eugenia Floyd; Shawn Harriman; Jim Atherton; Steve Hillerman; Cathy Soderstrom; Kou Kou; Tom Gant; Mark C. Noe; Barb Foster; Farzan Rastinejad; Matthew A. Marx; Tracey Lee Schaeffer; Pamela Whalen; W. Gregory Roberts


Cancer Research | 2005

Antiangiogenic and Antitumor Activity of a Selective PDGFR Tyrosine Kinase Inhibitor, CP-673,451

W. Gregory Roberts; Pamela Whalen; Erik Soderstrom; Garrett Moraski; Joseph P. Lyssikatos; Huifen-F. Wang; Beth Cooper; Deborah A. Baker; Douglas Savage; Deepak Dalvie; James A. Atherton; Sherry Ralston; Ruby Anthea Szewc; John Charles Kath; Jing Lin; Cathy Soderstrom; George T. Tkalcevic; Bruce D. Cohen; Vince Pollack; Wayne E. Barth; Will Hungerford; Ethan Ung

Collaboration


Dive into the W. Gregory Roberts's collaboration.

Researchain Logo
Decentralizing Knowledge