Network


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

Hotspot


Dive into the research topics where Patrick W. Vincent is active.

Publication


Featured researches published by Patrick W. Vincent.


Cancer Research | 2007

PF00299804, an Irreversible Pan-ERBB Inhibitor, Is Effective in Lung Cancer Models with EGFR and ERBB2 Mutations that Are Resistant to Gefitinib

Jeffrey A. Engelman; Kreshnik Zejnullahu; Christopher Michael Gale; Eugene Lifshits; Andrea J. Gonzales; Takeshi Shimamura; Feng Zhao; Patrick W. Vincent; George N. Naumov; James E. Bradner; Irene W. Althaus; Leena Gandhi; Geoffrey I. Shapiro; James M. Nelson; John V. Heymach; Matthew Meyerson; Kwok-Kin Wong; Pasi A. Jänne

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors gefitinib and erlotinib are effective treatments for a subset of non-small cell lung cancers. In particular, cancers with specific EGFR-activating mutations seem to be the most sensitive to these agents. However, despite their initial response, such cancers almost invariably develop resistance. In 50% of such cancers, a secondary EGFR mutation, T790M, has been identified that renders gefitinib and erlotinib ineffective inhibitors of EGFR kinase activity. Thus, there is a clinical need to develop novel EGFR inhibitors that can effectively inactivate T790M-containing EGFR proteins. In this study, we evaluate the effectiveness of a novel compound, PF00299804, an irreversible pan-ERBB inhibitor. The results from these studies show that PF00299804 is a potent inhibitor of EGFR-activating mutations as well as the EGFR T790M resistance mutation both in vitro and in vivo. Additionally, PF00299804 is a highly effective inhibitor of both the wild-type ERBB2 and the gefitinib-resistant oncogenic ERBB2 mutation identified in lung cancers. These preclinical evaluations support further clinical development of PF00299804 for cancers with mutations and/or amplifications of ERBB family members.


Molecular Cancer Therapeutics | 2008

Antitumor activity and pharmacokinetic properties of PF-00299804, a second-generation irreversible pan-erbB receptor tyrosine kinase inhibitor

Andrea J. Gonzales; Kenneth E. Hook; Irene W. Althaus; Paul A. Ellis; Erin Trachet; Amy Delaney; Patricia J. Harvey; Teresa Ellis; Danielle M. Amato; James M. Nelson; David W. Fry; Tong Zhu; Cho-Ming Loi; Stephen A. Fakhoury; Kevin Matthew Schlosser; R. Thomas Winters; Jessica Elizabeth Reed; Alex J. Bridges; Daniel J. Lettiere; Deborah A. Baker; Jianxin Yang; Helen T. Lee; Haile Tecle; Patrick W. Vincent

Signaling through the erbB receptor family of tyrosine kinases contributes to the proliferation, differentiation, migration, and survival of a variety of cell types. Abnormalities in members of this receptor family have been shown to play a role in oncogenesis, thus making them attractive targets for anticancer treatments. PF-00299804 is a second-generation irreversible pan-erbB receptor tyrosine kinase inhibitor currently in phase I clinical trials. PF-00299804 is believed to irreversibly inhibit erbB tyrosine kinase activity through binding at the ATP site and covalent modification of nucleophilic cysteine residues in the catalytic domains of erbB family members. Oral administration of PF-00299804 causes significant antitumor activity, including marked tumor regressions in a variety of human tumor xenograft models that express and/or overexpress erbB family members or contain the double mutation (L858R/T790M) in erbB1 (EGFR) associated with resistance to gefitinib and erlotinib. Furthermore, PF-00299804 shows exceptional distribution to human tumor xenografts and excellent pharmacokinetic properties across species. [Mol Cancer Ther 2008;7(7):1880–9]


The Journal of Nuclear Medicine | 2007

Preclinical Efficacy of the c-Met Inhibitor CE-355621 in a U87 MG Mouse Xenograft Model Evaluated by 18F-FDG Small-Animal PET

Jeffrey R. Tseng; Keon Wook Kang; Mangal Dandekar; Shahriar S. Yaghoubi; Joseph H. Lee; James G. Christensen; Stephen Muir; Patrick W. Vincent; Neil R. Michaud; Sanjiv S. Gambhir

The purpose of this study was to evaluate the efficacy of CE-355621, a novel antibody against c-Met, in a subcutaneous U87 MG xenograft mouse model using 18F-FDG small-animal PET. Methods: CE-355621 or control vehicle was administered intraperitoneally into nude mice (drug-treated group, n = 12; control group, n = 14) with U87 MG subcutaneous tumor xenografts. Drug efficacy was evaluated over 2 wk using 18F-FDG small-animal PET and compared with tumor volume growth curves. Results: The maximum %ID/g (percentage injected dose per gram of tissue) of 18F-FDG accumulation in mice treated with CE-355621 remained essentially unchanged over 2 wk, whereas the %ID/g of the control tumors increased 66% compared with the baseline. Significant inhibition of 18F-FDG accumulation was seen 3 d after drug treatment, which was earlier than the inhibition of tumor volume growth seen at 7 d after drug treatment. Conclusion: CE-355621 is an efficacious novel antineoplastic chemotherapeutic agent that inhibits 18F-FDG accumulation earlier than tumor volume changes in a mouse xenograft model. These results support the use of 18F-FDG PET to assess early tumor response for CE-355621.


Bioorganic & Medicinal Chemistry Letters | 2008

Synthesis and structure based optimization of novel Akt inhibitors

Blaise Lippa; Gonghua Pan; Matthew Corbett; Chao Li; Goss Stryker Kauffman; Jayvardhan Pandit; Shaughnessy Robinson; Liuqing Wei; Ekaterina Kozina; Eric S. Marr; Gary Borzillo; Elisabeth Knauth; Elsa G. Barbacci-Tobin; Patrick W. Vincent; Merin Troutman; Deborah A. Baker; Francis Rajamohan; Shefali Kakar; Tracey Clark; Joel Morris

Based on a high throughput screening hit, pyrrolopyrimidine inhibitors of the Akt kinase are explored. X-ray co-crystal structures of two lead series results in the understanding of key binding interactions, the design of new lead series, and enhanced potency. The syntheses of these series and their biological activities are described. Spiroindoline 13j is found to have an Akt1 kinase IC(50) of 2.4+/-0.6 nM, Akt cell potency of 50+/-19 nM, and provides 68% inhibition of tumor growth in a mouse xenograft model (50 mg/kg, qd, po).


Molecular Cancer Therapeutics | 2005

Plasma vascular endothelial growth factor and interleukin-8 as biomarkers of antitumor efficacy of a prototypical erbB family tyrosine kinase inhibitor

James G. Christensen; Patrick W. Vincent; Wayne D. Klohs; David W. Fry; Wilbur R. Leopold; William L. Elliott

CI-1033 (N-[4-[N-(3-chloro-4-fluorophenyl)amino-7-[3-(4-morpholynyl)propoxy]quinazolin-6-yl]acrylamide, PD 0183805-mesylate salt) was identified as a potent, selective inhibitor of erbB family tyrosine kinases, which are overexpressed in a number of solid tumors and have been shown to be involved in tumor progression. Because objective response of clinical patients to erbB-targeted therapies like CI-1033 has been observed only in a subset of cancer patients that exhibit the intended molecular targets, much emphasis has been placed on the identification of biomarkers of antitumor efficacy. Vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) were considered as potential biomarkers for CI-1033 due to ease of detection in patient plasma and showed roles in angiogenesis and cancer progression and positive regulation by the erbB receptor family. In the present studies, mice bearing established xenografts (A431 epidermoid carcinoma, H125 non–small cell lung carcinoma, SF767 glioblastoma, and MDA-MB-468 mammary carcinoma) were treated with efficacious and subefficacious doses of CI-1033, and plasma levels and xenograft gene expression of VEGF and IL-8 were evaluated. Oral administration of CI-1033 to tumor-bearing mice at efficacious doses resulted in markedly decreased levels of VEGF and/or IL-8 plasma levels and tumor mRNA levels relative to vehicle-treated control mice in xenograft models that exhibited evaluable levels of these markers. In contrast, subefficacious doses of CI-1033 did not significantly affect VEGF or IL-8 levels in any of the xenograft models. These studies indicate that plasma VEGF and IL-8 may have use as biomarkers of antitumor efficacy for epidermal growth factor receptor/erbB–targeted therapies such as CI-1033 and suggest that further clinical study of these markers in cancer patients are warranted.


mAbs | 2012

Biochemical and pharmacological characterization of human c-Met neutralizing monoclonal antibody CE-355621.

Neil R. Michaud; Jitesh P. Jani; Stephen M. Hillerman; Konstantinos Tsaparikos; Elsa G. Barbacci-Tobin; Elisabeth Knauth; Henry Putz; Mary Campbell; George A. Karam; Boris A. Chrunyk; David F. Gebhard; Larry L. Green; Jinghai J. Xu; Margaret C. Dunn; Tim M. Coskran; Jean-Martin Lapointe; Bruce D. Cohen; Kevin Coleman; Vahe Bedian; Patrick W. Vincent; Shama Kajiji; Stefan J. Steyn; Gary Borzillo; Gerrit Los

The c-Met proto-oncogene is a multifunctional receptor tyrosine kinase that is stimulated by its ligand, hepatocyte growth factor (HGF), to induce cell growth, motility and morphogenesis. Dysregulation of c-Met function, through mutational activation or overexpression, has been observed in many types of cancer and is thought to contribute to tumor growth and metastasis by affecting mitogenesis, invasion, and angiogenesis. We identified human monoclonal antibodies that bind to the extracellular domain of c-Met and inhibit tumor growth by interfering with ligand-dependent c-Met activation. We identified antibodies representing four independent epitope classes that inhibited both ligand binding and ligand-dependent activation of c-Met in A549 cells. In cells, the antibodies antagonized c-Met function by blocking receptor activation and by subsequently inducing downregulation of the receptor, translating to phenotypic effects in soft agar growth and tubular morphogenesis assays. Further characterization of the antibodies in vivo revealed significant inhibition of c-Met activity (≥ 80% lasting for 72–96 h) in excised tumors corresponded to tumor growth inhibition in multiple xenograft tumor models. Several of the antibodies identified inhibited the growth of tumors engineered to overexpress human HGF and human c-Met (S114 NIH 3T3) when grown subcutaneously in athymic mice. Furthermore, lead candidate antibody CE-355621 inhibited the growth of U87MG human glioblastoma and GTL-16 gastric xenografts by up to 98%. The findings support published pre-clinical and clinical data indicating that targeting c-Met with human monoclonal antibodies is a promising therapeutic approach for the treatment of cancer.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery and synthesis of novel 4-aminopyrrolopyrimidine Tie-2 kinase inhibitors for the treatment of solid tumors.

Jean Beebe; Martin A. Berliner; Vincent Bernardo; Merin Boehm; Gary Borzillo; Tracey Clark; Bruce D. Cohen; Richard D. Connell; Heather N. Frost; Deborah Gordon; William M. Hungerford; Shefali Kakar; Aaron Kanter; Nandell F. Keene; Elizabeth Knauth; Susan Deborah Lagreca; Yong Lu; Louis Martinez-Alsina; Matthew A. Marx; Joel Morris; Nandini Chaturbhai Patel; Doug Savage; Cathy Soderstrom; Carl Thompson; George T. Tkalcevic; Norma Jacqueline Tom; Felix Vajdos; James J. Valentine; Patrick W. Vincent; Matthew D. Wessel

The synthesis and biological evaluation of novel Tie-2 kinase inhibitors are presented. Based on the pyrrolopyrimidine chemotype, several new series are described, including the benzimidazole series by linking a benzimidazole to the C5-position of the 4-amino-pyrrolopyrimidine core and the ketophenyl series synthesized by incorporating a ketophenyl group to the C5-position. Medicinal chemistry efforts led to potent Tie-2 inhibitors. Compound 15, a ketophenyl pyrrolopyrimidine urea analog with improved physicochemical properties, demonstrated favorable in vitro attributes as well as dose responsive and robust oral tumor growth inhibition in animal models.


Journal of Medicinal Chemistry | 1998

2-Substituted aminopyrido[2,3-d]pyrimidin-7(8H)-ones. Structure-activity relationships against selected tyrosine kinases and in vitro and in vivo anticancer activity

Sylvester Klutchko; James Marino Hamby; Diane H. Boschelli; Zhipei Wu; Alan J. Kraker; Aneesa M. Amar; Brian G. Hartl; Cynthia Shen; Wayne D. Klohs; Randall W. Steinkampf; Denise L. Driscoll; James M. Nelson; William L. Elliott; Billy J. Roberts; Chad L. Stoner; Patrick W. Vincent; Donald J. Dykes; Robert L. Panek; Gina H. Lu; Terry C. Major; Tawny K. Dahring; Hussein Hallak; Laura A. Bradford; H. D. Hollis Showalter; Annette Marian Doherty


Journal of Medicinal Chemistry | 1998

Tyrosine Kinase Inhibitors. 14. Structure−Activity Relationships for Methyl- amino-Substituted Derivatives of 4-[(3-Bromophenyl)amino]-6-(methylamino)- pyrido[3,4-d]pyrimidine (PD 158780), a Potent and Specific Inhibitor of the Tyrosine Kinase Activity of Receptors for the EGF Family of Growth Factors

Gordon W. Rewcastle; D. K. Murray; William L. Elliott; David W. Fry; Curtis T. Howard; James M. Nelson; Billy J. Roberts; Patrick W. Vincent; H. D. Hollis Showalter; R. T. Winters; William A. Denny


Cancer Chemotherapy and Pharmacology | 2006

Sorafenib is efficacious and tolerated in combination with cytotoxic or cytostatic agents in preclinical models of human non-small cell lung carcinoma

Christopher Carter; Charles Chen; Cheryl Brink; Patrick W. Vincent; Yulia Y. Maxuitenko; Karen S. Gilbert; William R. Waud; Xiaomei Zhang

Collaboration


Dive into the Patrick W. Vincent's collaboration.

Researchain Logo
Decentralizing Knowledge