Network


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

Hotspot


Dive into the research topics where Kenneth E. Hook is active.

Publication


Featured researches published by Kenneth E. Hook.


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]


Investigational New Drugs | 1995

Inhibition of the epidermal growth factor receptor tyrosine kinase by PD153035 in human A431 tumors in athymic nude mice

Mark W. Kunkel; Kenneth E. Hook; Curtis T. Howard; Sally Przybranowski; Billy J. Roberts; William L. Elliott; Wilbur R. Leopold

SummaryPD153035 is a potent (Ki=6 pm) and specific inhibitor of the epidermal growth factor (EOF) receptor tyrosine kinase that suppresses tyrosine phosphorylation of the EGF receptor in A431 cells at nanomolar concentrations in cell culture. We have examined the pharmacokinetics of this compound and its ability to rapidly suppress phosphorylation of the EGF receptor in A431 human epidermoid tumors grown as xenografts in immunodeficient nude mice. Following a single i.p. dose of 80 mg/kg, the drug levels in the plasma and tumor rose to 50 and 22 μM within 15 minutes. While the plasma levels of PD153035 fell below 1 μM by 3 hours, in the tumors it remained at micromolar concentrations for at least 12 hours. The tyrosine phosphorylation of the EGF receptor was rapidly suppressed by 80–90% in the tumors. However receptor phosphorylation returned to control levels after 3 hours despite the continued presence of the drug at concentrations which, based on previousin vitro results, were predicted to maintain inhibition. EGF-stimulated tyrosine kinase activity in tumor extracts was decreased and recovered in parallel with the effects of PD153035 on receptor phosphorylation though the activity had reached only about half of the control activity after three hours. These results demonstrate the potential for using small molecule inhibitors to inhibit the EGF receptor tyrosine kinasein vivo, though a fair evaluation of their potential anti-cancer activity will have to wait for solutions to problems with sustained delivery which may allow us to maintain suppression of EGF receptor phosphorylation.


Molecular Cancer Therapeutics | 2012

An integrated genomic approach to identify predictive biomarkers of response to the aurora kinase inhibitor PF-03814735.

Kenneth E. Hook; Scott J. Garza; Maruja E. Lira; Keith Ching; Nathan V. Lee; Joan Cao; Jing Yuan; Jingjing Ye; Mark Ozeck; Stephanie Shi; Xianxian Zheng; Paul A. Rejto; Julie L.C. Kan; James G. Christensen; Adam Pavlicek

PF-03814735 is a novel, reversible inhibitor of Aurora kinases A and B that finished a phase I clinical trial for the treatment of advanced solid tumors. To find predictive biomarkers of drug sensitivity, we screened a diverse panel of 87 cancer cell lines for growth inhibition upon PF-03814735 treatment. Small cell lung cancer (SCLC) and, to a lesser extent, colon cancer lines were very sensitive to PF-03814735. The status of the Myc gene family and retinoblastoma pathway members significantly correlated with the efficacy of PF-03814735. Whereas RB1 inactivation, intact CDKN2A/p16, and normal CCND1/Cyclin D1 status are hallmarks of SCLC, activation or amplification of any of the three Myc genes (MYC, MYCL1, and MYCN) clearly differentiated cell line sensitivity within the SCLC panel. By contrast, we found that expression of Aurora A and B were weak predictors of response. We observed a decrease in histone H3 phosphorylation and polyploidization of sensitive lines, consistent with the phenotype of Aurora B inhibition. In vivo experiments with two SCLC xenograft models confirmed the sensitivity of Myc gene-driven models to PF-03814735 and a possible schedule dependence of MYC/c-Myc–driven tumors. Altogether our results suggest that SCLC and other malignancies driven by the Myc family genes may be suitable indications for treatment by Aurora B kinase inhibitors. Mol Cancer Ther; 11(3); 710–9. ©2012 AACR.


Molecular Cancer Therapeutics | 2010

PF-03814735, an Orally Bioavailable Small Molecule Aurora Kinase Inhibitor for Cancer Therapy

Jitesh P. Jani; Vincent Bernardo; Samit Kumar Bhattacharya; David Briere; Bruce D. Cohen; Kevin Coleman; James G. Christensen; Erling O. Emerson; Amy B. Jakowski; Kenneth E. Hook; Gerrit Los; James D. Moyer; Ingrid Pruimboom-Brees; Leslie R. Pustilnik; Ann Marie Rossi; Stefan J. Steyn; Chunyan Su; Konstantinos Tsaparikos; Donn G. Wishka; Kwansik Yoon; John Jakubczak

The Aurora family of highly related serine/threonine kinases plays a key role in the regulation of mitosis. Aurora1 and Aurora2 play important but distinct roles in the G2 and M phases of the cell cycle and are essential for proper chromosome segregation and cell division. Overexpression and amplification of Aurora2 have been reported in different tumor types, including breast, colon, pancreatic, ovarian, and gastric cancer. PF-03814735 is a novel, potent, orally bioavailable, reversible inhibitor of both Aurora1 and Aurora2 kinases that is currently in phase I clinical trials for the treatment of advanced solid tumors. In intact cells, the inhibitory activity of PF-03814735 on the Aurora1 and Aurora2 kinases reduces levels of phospho-Aurora1, phosphohistone H3, and phospho-Aurora2. PF-03814735 produces a block in cytokinesis, resulting in inhibition of cell proliferation and the formation of polyploid multinucleated cells. Although PF-03814735 produces significant inhibition of several other protein kinases, the predominant biochemical effects in cellular assays are consistent with inhibition of Aurora kinases. Once-daily oral administration of PF-03814735 to mice bearing human xenograft tumors produces a reduction in phosphohistone H3 in tumors at doses that are tolerable and that result in significant inhibition of tumor growth. The combination of PF-03814735 and docetaxel in xenograft mouse tumor models shows additive tumor growth inhibition. These results support the clinical evaluation of PF-03814735 in cancer patients. Mol Cancer Ther; 9(4); 883–94. ©2010 AACR.


Advances in Enzyme Regulation | 1985

The biochemical pharmacology of CI-920, a structurally novel antibiotic with antileukemic activity

Robert C. Jackson; David W. Fry; Theodore J. Boritzki; Billy J. Roberts; Kenneth E. Hook; Wilbur R. Leopold

CI-920 is a structurally novel, phosphate-containing polyene lactone antitumor agent isolated from a previously undescribed subspecies of Streptomyces pulveraceus cultured from a Brazilian soil sample. CI-920 was active against murine leukemia P388, and highly active and curative against L1210 leukemia in vivo. CI-920 was less active or inactive against the murine solid tumors tested. Daily administration for five to nine days was more effective against L1210 leukemia than a single dose or doses every four days. Given three times daily for five days, CI-920 was more toxic and less active. CI-920 had similar activity intravenously and intraperitoneally. Oral administration was inactive and nontoxic. Subcutaneous treatment was less effective and more toxic. Structure-activity relationship studies showed that the phosphate group was essential for antitumor activity in vivo and in vitro. Hydrolyzing the lactone ring also resulted in loss of antitumor activity, as did acetylation of the 6-hydroxyl group. Hydroxylation at the 5-position of the lactone ring resulted in partial retention of antitumor activity, but in greater toxicity to mice. Removal of the 13-hydroxyl group resulted in retention of high antitumor activity with approximately three-fold improvement in dose-potency. CI-920 is not cytotoxic to prokaryotic cells. CI-920 causes inhibition of biosynthesis of RNA and DNA in intact L1210 cells. Protein synthesis is also inhibited at higher drug concentrations. The inhibition of nucleic acid synthesis is not an antimetabolite effect, since pools of ribonucleoside triphosphates and deoxyribonucleoside triphosphates are not depleted. CI-920 does not cause DNA strand breakage, as measured by alkaline elution, and is not mutagenic in the Ames test at concentrations up to 200 micrograms/ml. CI-920 does not cause direct inhibition of RNA polymerase or DNA polymerase in permeabilized cells. It is possible that CI-920 must be metabolically activated within the target cells; alternatively it may interact with a component of chromatin other than DNA or the polymerases. Flow cytometry studies showed that growth-inhibitory levels of CI-920 caused accumulation of cells in the G2+M region. Higher drug concentrations caused an S-phase block. CI-920 is an inhibitor and irreversible inactivator of reduced folate membrane transport, and appears to enter cells by this receptor. L1210 cells selected for resistance to CI-920 are cross-resistant to methotrexate, and deficient in reduced folate transport.(ABSTRACT TRUNCATED AT 400 WORDS)


Cancer Chemotherapy and Pharmacology | 1986

Cell cycle effects of trimetrexate (CI-898)

Kenneth E. Hook; James M. Nelson; Billy J. Roberts; Daniel P. Griswold; Wilbur R. Leopold

SummaryThe cell cycle phase specificity of trimetrexate (CI-898) was examined. CHO cells synchronized by mitotic selection were exposed to 50 μM trimetrexate for 2 h at various time points after release from Colcemid block. Only S phase cells were sensitive to trimetrexate when survival was measured by a cloning assay. Comparison of plateau phase and log phase cultures indicated that plateau phase CHO cells were relatively insensitive to 5 μM trimetrexate. Exponentially growing L1210 cells were continuously exposed to either 30 nM or 3 nM trimetrexate and analyzed by DNA flow cytometry. Incubation with 30 nM trimetrexate produced cell cycle arrest in late G1 or early S phase, while exposure to 3 nM trimetrexate caused only a delay in progression through S phase. In an in vivo schedule dependence study with mice bearing approximately 3×106 P388 leukemia cells, trimetrexate was most effective with frequent administration. Mice treated on the optimal schedule, every 3 h×8 on days 1, 5, and 9 after tumor implant, had life-span increases in excess of 100%.


Investigational New Drugs | 1996

Cellular transport of CI-980

Kenneth E. Hook; Sally Przybranowski; Wilbur R. Leopold

SummaryCI-980, originally synthesized as a potential folate antagonist, is a tubulin-binding mitotic inhibitor currently in pediatric phase I and adult phase II clinical trials. Because of its extensive tissue distribution in animals and its favorable activity against multidrug resistant (MDR) cells compared with other mitotic inhibitors, such as vincristine, we examined the membrane transport properties of CI-980. CI-980 accumulated rapidly in L1210 and CHO/K1 cells, reaching intracellular levels 40- and 8-fold higher, respectively, than those in the extracellular medium. Efflux was also quite rapid, but a small fraction of drug remained associated with the cells in drug-free medium. The uptake of CI-980 was not temperature or energy dependent, nor was it saturable up to an extracellular concentration of 100 μM. Inhibitors of nucleoside transport had no effect on CI-980 uptake. A cell line deficient in the transport of reduced folate was not resistant to CI-980, nor did it exhibit reduced CI-980 uptake. A 100-fold excess of the R-enantiomer inhibited CI-980 uptake by only 50%. These results are consistent with a model of CI-980 uptake involving passive diffusion followed by significant but largely reversible binding to intracellular or membrane components.


Advances in Enzyme Regulation | 1989

Biochemical pharmacology and antitumor properties of 4-amino-8-[β--ribofuranosylamino]pyrimido-[5,4-d]pyrimidine

Robert C. Jackson; Theodore J. Boritzki; P. Dan Cook; Kenneth E. Hook; Wilbur R. Leopold; David W. Fry

Abstract 1. 1. APP is activated by adenosine kinase to its 5′-phosphate (APP-MP). 2. 2. APP-MP inhibits PRPP synthetase, and depletes cellular PRPP and purine and pyrimidine nucleotides. 3. 3. APP inhibits synthesis of DNA and RNA, and blocks cells in G1 phase of the cell cycle. 4. 4. APP retains full activity against MDR cells. 5. 5. APP is equally active against quiescent and proliferating CHO cells. 6. 6. APP has only weak activity against L1210 leukemia in vivo , but has substantial activity against mammary carcinoma 16/c. 7. 7. In vitro , APP has a relatively high ratio of solid tumor: leukemia activity.


Proceedings of the National Academy of Sciences of the United States of America | 1998

Specific, irreversible inactivation of the epidermal growth factor receptor and erbB2, by a new class of tyrosine kinase inhibitor

David W. Fry; Alexander James Bridges; William A. Denny; Annette Marian Doherty; Kenneth D. Greis; James L. Hicks; Kenneth E. Hook; Paul R. Keller; Wilbur R. Leopold; Joseph A. Loo; Dennis Joseph Mcnamara; James M. Nelson; Veronika Sherwood; Jeff B. Smaill; Susanne Trumpp-Kallmeyer; Ellen Myra Dobrusin


International Journal of Cancer | 1995

Role of a small molecular weight phosphoprotein in the mechanism of action of CI-994 (N-acetyldinaline)

Sue A. Rummel; Alan J. Kraker; Randall W. Steinkampf; Kenneth E. Hook; Wayne D. Klohs

Collaboration


Dive into the Kenneth E. Hook's collaboration.

Top Co-Authors

Avatar

David W. Fry

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wilbur R. Leopold

Charles River Laboratories

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge