Network


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

Hotspot


Dive into the research topics where Jeffrey R. Jackson is active.

Publication


Featured researches published by Jeffrey R. Jackson.


Nature Reviews Cancer | 2007

Targeted anti-mitotic therapies: can we improve on tubulin agents?

Jeffrey R. Jackson; Denis R. Patrick; Mohammed M. Dar; Pearl S. Huang

The advent of molecularly targeted drug discovery has facilitated the identification of a new generation of anti-mitotic therapies that target proteins with specific functions in mitosis. The exquisite selectivity for mitosis and the distinct ways in which these new agents interfere with mitosis provides the potential to not only overcome certain limitations of current tubulin-targeted anti-mitotic drugs, but to expand the scope of clinical efficacy that those drugs have established. The development of these new anti-mitotic drugs as targeted therapies faces significant challenges; nevertheless, these potential therapies also serve as unique tools to dissect the molecular mechanisms of the mitotic-checkpoint response.


Current Opinion in Pharmacology | 2001

Past and future of the mitotic spindle as an oncology target

Kenneth W. Wood; William D. Cornwell; Jeffrey R. Jackson

Tubulin poisons were first discovered decades ago, but the recent clinical and commercial success of Taxol has led to a renaissance in the search for novel mitotic spindle poisons to treat cancer. Many tubulin poisons have been identified, but few have demonstrated clinical utility. Recent studies have begun to identify the factors that differentiate the efficacy of these agents. In addition, promising alternative approaches to targeting the mitotic spindle have been identified from detailed studies of mitotic regulation and mechanics.


ACS Medicinal Chemistry Letters | 2010

Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin.

Steven David Knight; Nicholas D. Adams; Joelle L. Burgess; Amita M. Chaudhari; Michael G. Darcy; Carla A. Donatelli; Juan I. Luengo; Ken A. Newlander; Cynthia A. Parrish; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt; Glenn S. Van Aller; Jeffrey D. Carson; Melody Diamond; Patricia A. Elkins; Christine M. Gardiner; Eric Garver; Seth Gilbert; Richard R. Gontarek; Jeffrey R. Jackson; Kevin L. Kershner; Lusong Luo; Kaushik Raha; Christian S. Sherk; Chiu-Mei Sung; David Sutton; Peter J. Tummino; Ronald Wegrzyn; Kurt R. Auger

Phosphoinositide 3-kinase α (PI3Kα) is a critical regulator of cell growth and transformation, and its signaling pathway is the most commonly mutated pathway in human cancers. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of PI3K/AKT pathway inhibition. 2,4-Difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide (GSK2126458, 1) has been identified as a highly potent, orally bioavailable inhibitor of PI3Kα and mTOR with in vivo activity in both pharmacodynamic and tumor growth efficacy models. Compound 1 is currently being evaluated in human clinical trials for the treatment of cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Antitumor activity of an allosteric inhibitor of centromere-associated protein-E

Kenneth W. Wood; Latesh Lad; Lusong Luo; Xiangping Qian; Steven D. Knight; Neysa Nevins; Katjuša Brejc; David Sutton; Aidan G. Gilmartin; Penelope Chua; Radhika Desai; Stephen Schauer; Dean E. McNulty; Roland S. Annan; Lisa Belmont; Carlos Garcia; Yan Lee; Melody Diamond; Leo F. Faucette; Michele Giardiniere; Shu-Yun Zhang; Chiu-Mei Sun; Justin D. Vidal; Serge Lichtsteiner; William D. Cornwell; Joel Greshock; Richard Wooster; Jeffrey T. Finer; Robert A. Copeland; Pearl S. Huang

Centromere-associated protein-E (CENP-E) is a kinetochore-associated mitotic kinesin that is thought to function as the key receptor responsible for mitotic checkpoint signal transduction after interaction with spindle microtubules. We have identified GSK923295, an allosteric inhibitor of CENP-E kinesin motor ATPase activity, and mapped the inhibitor binding site to a region similar to that bound by loop-5 inhibitors of the kinesin KSP/Eg5. Unlike these KSP inhibitors, which block release of ADP and destabilize motor-microtubule interaction, GSK923295 inhibited release of inorganic phosphate and stabilized CENP-E motor domain interaction with microtubules. Inhibition of CENP-E motor activity in cultured cells and tumor xenografts caused failure of metaphase chromosome alignment and induced mitotic arrest, indicating that tight binding of CENP-E to microtubules is insufficient to satisfy the mitotic checkpoint. Consistent with genetic studies in mice suggesting that decreased CENP-E function can have a tumor-suppressive effect, inhibition of CENP-E induced tumor cell apoptosis and tumor regression.


Cancer Research | 2009

Distinct Concentration-Dependent Effects of the Polo-like Kinase 1–Specific Inhibitor GSK461364A, Including Differential Effect on Apoptosis

Aidan G. Gilmartin; Maureen R. Bleam; Mark Richter; Symon G. Erskine; Ryan G. Kruger; Lenore Madden; Daniel F. Hassler; Gary K. Smith; Richard R. Gontarek; Mary P. Courtney; David Sutton; Melody Diamond; Jeffrey R. Jackson; Sylvie Laquerre

Polo-like kinase 1 (Plk1) is a conserved serine/threonine kinase that plays an essential role in regulating the many processes involved in mitotic entry and progression. In humans, Plk1 is expressed primarily during late G(2) and M phases and, in conjunction with Cdk1/cyclin B1, acts as master regulatory kinases for the myriad protein substrates involved in mitosis. Plk1 overexpression is strongly associated with cancer and has been correlated with poor prognosis in a broad range of human tumor types. We have identified a potent, selective, reversible, ATP-competitive inhibitor of Plk1, GSK461364A, capable of inhibiting cell growth of most proliferating cancer cell lines tested. We observe distinct cell cycle effects of GSK461364A depending on the dose used. The predominant phenotype for cells treated with GSK461364A is prometaphase arrest with characteristic collapsed polar polo spindle. At high concentrations, GSK461364A delays mitotic entry in G(2) followed by gradual progression into terminal mitosis; in some cell lines, this correlates with decreased apoptosis. Cell culture growth inhibition by GSK461364A can be cytostatic or cytotoxic but leads to tumor regression in xenograft tumor models under proper dose scheduling. Finally, we describe pharmacodynamic biomarkers of GSK461364A activity (pHH3 and Plk1) that are currently being evaluated in human cancer clinical trials.


Molecular Cancer Therapeutics | 2010

Sensitivity of Cancer Cells to Plk1 Inhibitor GSK461364A Is Associated with Loss of p53 Function and Chromosome Instability

Yan Degenhardt; Joel Greshock; Sylvie Laquerre; Aidan G. Gilmartin; Junping Jing; Mark Richter; Xiping Zhang; Maureen R. Bleam; Wendy S. Halsey; Ashley M. Hughes; Christopher Moy; Nancy Liu-Sullivan; Scott Powers; Kurtis E. Bachman; Jeffrey R. Jackson; Barbara L. Weber; Richard Wooster

Polo-like kinases are a family of serine threonine kinases that are critical regulators of cell cycle progression and DNA damage response. Predictive biomarkers for the Plk1-selective inhibitor GSK461364A were identified by comparing the genomics and genetics of a panel of human cancer cell lines with their response to a drug washout followed by an outgrowth assay. In this assay, cell lines that have lost p53 expression or carry mutations in the TP53 gene tended to be more sensitive to GSK461364A. These more sensitive cell lines also had increased levels of chromosome instability, a characteristic associated with loss of p53 function. Further mechanistic studies showed that p53 wild-type (WT) and not mutant cells can activate a postmitotic tetraploidy checkpoint and arrest at pseudo-G1 state after GSK461364A treatment. RNA silencing of WT p53 increased the antiproliferative activity of GSK461364A. Furthermore, silencing of p53 or p21/CDKN1A weakened the tetraploidy checkpoint in cells that survived mitotic arrest and mitotic slippage. As many cancer therapies tend to be more effective in p53 WT patients, the higher sensitivity of p53-deficient tumors toward GSK461364A could potentially offer an opportunity to treat tumors that are refractory to other chemotherapies as well as early line therapy for these genotypes. Mol Cancer Ther; 9(7); 2079–89. ©2010 AACR.


Genome Biology | 2009

Integrated analysis of breast cancer cell lines reveals unique signaling pathways

Laura M. Heiser; Nicholas Wang; Carolyn L. Talcott; Keith R. Laderoute; Merrill Knapp; Yinghui Guan; Zhi Hu; Safiyyah Ziyad; Barbara L. Weber; Sylvie Laquerre; Jeffrey R. Jackson; Richard Wooster; Wen Lin Kuo; Joe W. Gray; Paul T. Spellman

BackgroundCancer is a heterogeneous disease resulting from the accumulation of genetic defects that negatively impact control of cell division, motility, adhesion and apoptosis. Deregulation in signaling along the EgfR-MAPK pathway is common in breast cancer, though the manner in which deregulation occurs varies between both individuals and cancer subtypes.ResultsWe were interested in identifying subnetworks within the EgfR-MAPK pathway that are similarly deregulated across subsets of breast cancers. To that end, we mapped genomic, transcriptional and proteomic profiles for 30 breast cancer cell lines onto a curated Pathway Logic symbolic systems model of EgfR-MAPK signaling. This model was composed of 539 molecular states and 396 rules governing signaling between active states. We analyzed these models and identified several subtype-specific subnetworks, including one that suggested Pak1 is particularly important in regulating the MAPK cascade when it is over-expressed. We hypothesized that Pak1 over-expressing cell lines would have increased sensitivity to Mek inhibitors. We tested this experimentally by measuring quantitative responses of 20 breast cancer cell lines to three Mek inhibitors. We found that Pak1 over-expressing luminal breast cancer cell lines are significantly more sensitive to Mek inhibition compared to those that express Pak1 at low levels. This indicates that Pak1 over-expression may be a useful clinical marker to identify patient populations that may be sensitive to Mek inhibitors.ConclusionsAll together, our results support the utility of symbolic system biology models for identification of therapeutic approaches that will be effective against breast cancer subsets.


Bioorganic & Medicinal Chemistry Letters | 2009

Design of potent thiophene inhibitors of polo-like kinase 1 with improved solubility and reduced protein binding.

Kyle Allen Emmitte; George M. Adjebang; C. Webb Andrews; Jennifer G. Badiang Alberti; Ramesh Bambal; Stanley D. Chamberlain; Ronda G. Davis-Ward; Hamilton D. Dickson; Daniel F. Hassler; Keith R. Hornberger; Jeffrey R. Jackson; Kevin Wayne Kuntz; Timothy J. Lansing; Robert A. Mook; Kristen E. Nailor; Mark Andrew Pobanz; Stephon C. Smith; Chiu-Mei Sung; Mui Cheung

A series of thiophene PLK1 inhibitors was optimized for increased solubility and reduced protein binding through the appendage of basic amine functionality. Interesting selectivity between PLK1 and PLK3 was also obtained through these modifications.


Clinical Cancer Research | 2008

Centromere-associated protein E: a motor that puts the brakes on the mitotic checkpoint.

Kenneth W. Wood; Penelope Chua; David Sutton; Jeffrey R. Jackson

Cell cycle checkpoints have long been recognized as important nodes for regulating cell proliferation and maintaining genomic integrity. These checkpoints are often altered in cancer and represent promising points for therapeutic intervention. Until recently, direct targeting of the mitotic checkpoint has been an untapped area for cancer drug discovery. Regulation of the mitotic checkpoint is complex, but many of the critical players have been identified and functionally characterized. A substantial number of these proteins can be localized to the kinetochore, a structure located at the centromeric region of each mitotic chromosome. The kinetochore mediates chromosome attachment to spindle microtubules and subsequent chromosome movement. The mitotic checkpoint monitors microtubule attachment and chromosome position on the mitotic spindle, inhibiting progression into anaphase until proper attachment and metaphase positioning is achieved. Centromere-associated protein E is a kinesin microtubule motor protein that plays an essential role in integrating the mechanics of microtubule-chromosome interactions with mitotic checkpoint signaling, and has emerged as a novel target for cancer therapy.


ACS Medicinal Chemistry Letters | 2010

Discovery of the First Potent and Selective Inhibitor of Centromere-Associated Protein E: GSK923295.

Xiangping Qian; Andrew Mcdonald; Han-Jie Zhou; Nicholas D. Adams; Cynthia A. Parrish; Kevin J. Duffy; Duke M. Fitch; Rosanna Tedesco; Luke W. Ashcraft; Bing Yao; Hong Jiang; Jennifer Kuo Chen Huang; Melchor V. Marin; Carrie E. Aroyan; Jianchao Wang; Seyed Ahmed; Joelle L. Burgess; Amita M. Chaudhari; Carla A. Donatelli; Michael G. Darcy; Lance H. Ridgers; Ken A. Newlander; Stanley J. Schmidt; Deping Chai; Mariela Colón; Michael N. Zimmerman; Latesh Lad; Roman Sakowicz; Stephen Schauer; Lisa Belmont

Inhibition of mitotic kinesins represents a novel approach for the discovery of a new generation of anti-mitotic cancer chemotherapeutics. We report here the discovery of the first potent and selective inhibitor of centromere-associated protein E (CENP-E) 3-chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide (GSK923295; 1), starting from a high-throughput screening hit, 3-chloro-4-isopropoxybenzoic acid 2. Compound 1 has demonstrated broad antitumor activity in vivo and is currently in human clinical trials.

Collaboration


Dive into the Jeffrey R. Jackson's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge