Network


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

Hotspot


Dive into the research topics where Gregory P. Donoho is active.

Publication


Featured researches published by Gregory P. Donoho.


Nature | 2007

A Transforming Mutation in the Pleckstrin Homology Domain of Akt1 in Cancer.

John D. Carpten; Andrew L. Faber; Candice Horn; Gregory P. Donoho; Stephen L. Briggs; Christiane M. Robbins; Galen Hostetter; Sophie Boguslawski; Tracy Moses; Stephanie Savage; Mark Uhlik; Aimin Lin; Jian Du; Yue-Wei Qian; Douglas J. Zeckner; Greg Tucker-Kellogg; Jeffrey W. Touchman; Ketan Patel; Spyro Mousses; Michael L. Bittner; Richard W. Schevitz; Mei-Huei T. Lai; Kerry Blanchard; James E. Thomas

Although AKT1 (v-akt murine thymoma viral oncogene homologue 1) kinase is a central member of possibly the most frequently activated proliferation and survival pathway in cancer, mutation of AKT1 has not been widely reported. Here we report the identification of a somatic mutation in human breast, colorectal and ovarian cancers that results in a glutamic acid to lysine substitution at amino acid 17 (E17K) in the lipid-binding pocket of AKT1. Lys 17 alters the electrostatic interactions of the pocket and forms new hydrogen bonds with a phosphoinositide ligand. This mutation activates AKT1 by means of pathological localization to the plasma membrane, stimulates downstream signalling, transforms cells and induces leukaemia in mice. This mechanism indicates a direct role of AKT1 in human cancer, and adds to the known genetic alterations that promote oncogenesis through the phosphatidylinositol-3-OH kinase/AKT pathway. Furthermore, the E17K substitution decreases the sensitivity to an allosteric kinase inhibitor, so this mutation may have important clinical utility for AKT drug development.


Cancer Cell | 2015

Inhibition of RAF Isoforms and Active Dimers by LY3009120 Leads to Anti-tumor Activities in RAS or BRAF Mutant Cancers

Sheng-Bin Peng; James Robert Henry; Michael Kaufman; Wei-Ping Lu; Bryan D. Smith; Subha Vogeti; Thomas J. Rutkoski; Scott C. Wise; Lawrence Chun; Youyan Zhang; Robert D. Van Horn; Tinggui Yin; Xiaoyi Zhang; Vipin Yadav; Shih-Hsun Chen; Xueqian Gong; Xiwen Ma; Yue Webster; Sean Buchanan; Igor Mochalkin; Lysiane Huber; Lisa Kays; Gregory P. Donoho; Jennie L. Walgren; Denis J. McCann; Phenil J. Patel; Ilaria Conti; Gregory D. Plowman; James J. Starling; Daniel L. Flynn

LY3009120 is a pan-RAF and RAF dimer inhibitor that inhibits all RAF isoforms and occupies both protomers in RAF dimers. Biochemical and cellular analyses revealed that LY3009120 inhibits ARAF, BRAF, and CRAF isoforms with similar affinity, while vemurafenib or dabrafenib have little or modest CRAF activity compared to their BRAF activities. LY3009120 induces BRAF-CRAF dimerization but inhibits the phosphorylation of downstream MEK and ERK, suggesting that it effectively inhibits the kinase activity of BRAF-CRAF heterodimers. Further analyses demonstrated that LY3009120 also inhibits various forms of RAF dimers including BRAF or CRAF homodimers. Due to these unique properties, LY3009120 demonstrates minimal paradoxical activation, inhibits MEK1/2 phosphorylation, and exhibits anti-tumor activities across multiple models carrying KRAS, NRAS, or BRAF mutation.


Molecular Cancer Therapeutics | 2014

A Novel CDK9 Inhibitor Shows Potent Antitumor Efficacy in Preclinical Hematologic Tumor Models

Tinggui Yin; María José Lallena; Emiko L. Kreklau; Kevin Robert Fales; Santiago Carballares; Raquel Torrres; Graham N. Wishart; Rose T. Ajamie; Damien M. Cronier; Phillip Iversen; Timothy I. Meier; Robert Foreman; Douglas J. Zeckner; Sean Sissons; Bart W. Halstead; Aimee B. Lin; Gregory P. Donoho; Yue-Wei Qian; Shuyu Li; Song Wu; Amit Aggarwal; Xiang S. Ye; James J. Starling; Richard B. Gaynor; Alfonso De Dios; Jian Du

DNA-dependent RNA polymerase II (RNAP II) largest subunit RPB1 C-terminal domain (CTD) kinases, including CDK9, are serine/threonine kinases known to regulate transcriptional initiation and elongation by phosphorylating Ser 2, 5, and 7 residues on CTD. Given the reported dysregulation of these kinases in some cancers, we asked whether inhibiting CDK9 may induce stress response and preferentially kill tumor cells. Herein, we describe a potent CDK9 inhibitor, LY2857785, that significantly reduces RNAP II CTD phosphorylation and dramatically decreases MCL1 protein levels to result in apoptosis in a variety of leukemia and solid tumor cell lines. This molecule inhibits the growth of a broad panel of cancer cell lines, and is particularly efficacious in leukemia cells, including orthotopic leukemia preclinical models as well as in ex vivo acute myeloid leukemia and chronic lymphocytic leukemia patient tumor samples. Thus, inhibition of CDK9 may represent an interesting approach as a cancer therapeutic target, especially in hematologic malignancies. Mol Cancer Ther; 13(6); 1442–56. ©2014 AACR.


Clinical Cancer Research | 2013

Inhibition of Tumor Growth and Metastasis in Non–Small Cell Lung Cancer by LY2801653, an Inhibitor of Several Oncokinases, Including MET

Wenjuan Wu; Chen Bi; Kelly M. Credille; Jason Manro; Victoria L. Peek; Gregory P. Donoho; Lei Yan; John A. Wijsman; S. Betty Yan; Richard A. Walgren

Purpose: Lung cancer is the leading cause of cancer-related death worldwide. Sustained activation, overexpression, or mutation of the MET pathway is associated with a poor prognosis in a variety of tumors, including non–small cell lung cancer (NSCLC), implicating the MET pathway as a potential therapeutic target for lung cancer. Previously, we reported on the development of LY2801653: a novel, orally bioavailable oncokinase inhibitor with MET as one of its targets. Here, we discuss the evaluation of LY2801653 in both preclinical in vitro and in vivo NSCLC models. Experimental Design/Results: Treatment with LY2801653 showed tumor growth inhibition in tumor cell lines and patient-derived tumor xenograft models as a single agent (37.4%–90.0% inhibition) or when used in combination with cisplatin, gemcitabine, or erlotinib (66.5%–86.3% inhibition). Mechanistic studies showed that treatment with LY2801653 inhibited the constitutive activation of MET pathway signaling and resulted in inhibition of NCI-H441 cell proliferation, anchorage-independent growth, migration, and invasion. These in vitro findings were confirmed in the H441 orthotopic model where LY2801653 treatment significantly inhibited both primary tumor growth (87.9% inhibition) and metastasis (64.5% inhibition of lymph node and 67.7% inhibition of chest wall). Tumor-bearing animals treated with LY2801653 had a significantly greater survival time (87% increase compared with the vehicle-treated mice). In the MET-independent NCI-H1299 orthotopic model, treatment with LY2801653 showed a significant inhibition of primary tumor growth but not metastasis. Conclusions: Collectively, these results support clinical evaluation of LY2801653 in NSCLCs and suggest that differences in the MET activation of tumors may be predictive of response. Clin Cancer Res; 19(20); 5699–710. ©2013 AACR.


Molecular Cancer Therapeutics | 2013

Efficacy of Low-Dose Oral Metronomic Dosing of the Prodrug of Gemcitabine, LY2334737, in Human Tumor Xenografts

Susan E. Pratt; Sara Durland-Busbice; Robert L. Shepard; Gregory P. Donoho; James J. Starling; Enaksha R. Wickremsinhe; Everett J. Perkins; Anne H. Dantzig

LY2334737, an oral prodrug of gemcitabine, is cleaved in vivo, releasing gemcitabine and valproic acid. Oral dosing of mice results in absorption of intact prodrug with slow systemic hydrolysis yielding higher plasma levels of LY2334737 than gemcitabine and prolonged gemcitabine exposure. Antitumor activity was evaluated in human colon and lung tumor xenograft models. The dose response for efficacy was examined using 3 metronomic schedules, once-a-day dosing for 14 doses, every other day for 7 doses, and once a day for 7 doses, 7 days rest, followed by an additional 7 days of once-a-day dosing. These schedules gave significant antitumor activity and were well tolerated. Oral gavage of 6 mg/kg LY2334737 daily for 21 days gave equivalent activity to i.v. 240 mg/kg gemcitabine. HCl administered once a week for 3 weeks to mice bearing a patient mesothelioma tumor PXF 1118 or a non–small cell lung cancer tumor LXFE 937. The LXFE 397 tumor possessed elevated expression of the equilibrative nucleoside transporter-1 (ENT1) important for gemcitabine uptake but not prodrug uptake and responded significantly better to treatment with LY2334737 than gemcitabine (P ≤ 0.001). In 3 colon xenografts, antitumor activity of LY2334737 plus a maximally tolerated dose of capecitabine, an oral prodrug of 5-fluorouracil, was significantly greater than either monotherapy. During treatment, the expression of carboxylesterase 2 (CES2) and concentrative nucleoside transporter-3 was induced in HCT-116 tumors; both are needed for the activity of the prodrugs. Thus, metronomic oral low-dose LY2334737 is efficacious, well tolerated, and easily combined with capecitabine for improved efficacy. Elevated CES2 or ENT1 expression may enhance LY2334737 tumor response. Mol Cancer Ther; 12(4); 481–90. ©2013 AACR.


Molecular Cancer Therapeutics | 2016

Characterization of LY3023414, a Novel PI3K/mTOR Dual Inhibitor Eliciting Transient Target Modulation to Impede Tumor Growth

Michele C. Smith; Mary M. Mader; James A. Cook; Philip W. Iversen; Rose T. Ajamie; Everett J. Perkins; Laura J. Bloem; Yvonne Yip; David Anthony Barda; Philip Parker Waid; Douglas J. Zeckner; Debra A. Young; Manuel Sanchez-Felix; Gregory P. Donoho; Volker Wacheck

The PI3K/AKT/mTOR pathway is among the most frequently altered pathways in cancer cell growth and survival. LY3023414 is a complex fused imidazoquinolinone with high solubility across a wide pH range designed to inhibit class I PI3K isoforms and mTOR kinase. Here, we describe the in vitro and in vivo activity of LY3023414. LY3023414 was highly soluble at pH 2–7. In biochemical testing against approximately 266 kinases, LY3023414 potently and selectively inhibited class I PI3K isoforms, mTORC1/2, and DNA-PK at low nanomolar concentrations. In vitro, inhibition of PI3K/AKT/mTOR signaling by LY3023414 caused G1 cell-cycle arrest and resulted in broad antiproliferative activity in cancer cell panel screens. In vivo, LY3023414 demonstrated high bioavailability and dose-dependent dephosphorylation of PI3K/AKT/mTOR pathway downstream substrates such as AKT, S6K, S6RP, and 4E-BP1 for 4 to 6 hours, reflecting the drugs half-life of 2 hours. Of note, equivalent total daily doses of LY3023414 given either once daily or twice daily inhibited tumor growth to similar extents in multiple xenograft models, indicating that intermittent target inhibition is sufficient for antitumor activity. In combination with standard-of-care drugs, LY3023414 demonstrated additive antitumor activity. The novel, orally bioavailable PI3K/mTOR inhibitor LY3023414 is highly soluble and exhibits potent in vivo efficacy via intermittent target inhibition. It is currently being evaluated in phase I and II trials for the treatment of human malignancies. Mol Cancer Ther; 15(10); 2344–56. ©2016 AACR.


Molecular Cancer Research | 2015

Intrinsic Resistance to Cixutumumab Is Conferred by Distinct Isoforms of the Insulin Receptor

Amelie Forest; Michael Amatulli; Dale L. Ludwig; Christopher B. Damoci; Ying Wang; Colleen A. Burns; Gregory P. Donoho; Nina Zanella; Heinz H. Fiebig; Marie Prewett; David Surguladze; James T. DeLigio; Peter J. Houghton; Malcolm A. Smith; Ruslan D. Novosiadly

Despite a recent shift away from anti–insulin-like growth factor I receptor (IGF-IR) therapy, this target has been identified as a key player in the resistance mechanisms to various conventional and targeted agents, emphasizing its value as a therapy, provided that it is used in the right patient population. Molecular markers predictive of antitumor activity of IGF-IR inhibitors remain largely unidentified. The aim of this study is to evaluate the impact of insulin receptor (IR) isoforms on the antitumor efficacy of cixutumumab, a humanized mAb against IGF-IR, and to correlate their expression with therapeutic outcome. The data demonstrate that expression of total IR rather than individual IR isoforms inversely correlates with single-agent cixutumumab efficacy in pediatric solid tumor models in vivo. Total IR, IR-A, and IR-B expression adversely affects the outcome of cixutumumab in combination with chemotherapy in patient-derived xenograft models of lung adenocarcinoma. IR-A overexpression in tumor cells confers complete resistance to cixutumumab in vitro and in vivo, whereas IR-B results in a partial resistance. Resistance in IR-B–overexpressing cells is fully reversed by anti–IGF-II antibodies, suggesting that IGF-II is a driver of cixutumumab resistance in this setting. The present study links IR isoforms, IGF-II, and cixutumumab efficacy mechanistically and identifies total IR as a biomarker predictive of intrinsic resistance to anti–IGF-IR antibody. Implications: This study identifies total IR as a biomarker predictive of primary resistance to IGF-IR antibodies and provides a rationale for new clinical trials enriched for patients whose tumors display low IR expression. Mol Cancer Res; 13(12); 1615–26. ©2015 AACR.


Molecular Cancer Therapeutics | 2015

A Novel Eg5 Inhibitor (LY2523355) Causes Mitotic Arrest and Apoptosis in Cancer Cells and Shows Potent Antitumor Activity in Xenograft Tumor Models

Xiang S. Ye; Li Fan; Robert D. Van Horn; Ryuichiro Nakai; Yoshihisa Ohta; Shiro Akinaga; Chikara Murakata; Yoshinori Yamashita; Tinggui Yin; Kelly M. Credille; Gregory P. Donoho; Farhana F. Merzoug; Heng Li; Amit Aggarwal; Kerry Blanchard; Eric Westin

Intervention of cancer cell mitosis by antitubulin drugs is among the most effective cancer chemotherapies. However, antitubulin drugs have dose-limiting side effects due to important functions of microtubules in resting normal cells and are often rendered ineffective by rapid emergence of resistance. Antimitotic agents with different mechanisms of action and improved safety profiles are needed as new treatment options. Mitosis-specific kinesin Eg5 represents an attractive anticancer target for discovering such new antimitotic agents, because Eg5 is essential only in mitotic progression and has no roles in resting, nondividing cells. Here, we show that a novel selective Eg5 inhibitor, LY2523355, has broad target-mediated anticancer activity in vitro and in vivo. LY2523355 arrests cancer cells at mitosis and causes rapid cell death that requires sustained spindle-assembly checkpoint (SAC) activation with a required threshold concentration. In vivo efficacy of LY2523355 is highly dose/schedule-dependent, achieving complete remission in a number of xenograft tumor models, including patient-derived xenograft (PDX) tumor models. We further establish that histone-H3 phosphorylation of tumor and proliferating skin cells is a promising pharmacodynamic biomarker for in vivo anticancer activity of LY2523355. Mol Cancer Ther; 14(11); 2463–72. ©2015 AACR.


Clinical Cancer Research | 2017

Mouse PDX Trial Suggests Synergy of concurrent Inhibition of RAF and EGFR in Colorectal Cancer with BRAF or KRAS mutations

Yung Mae M. Yao; Gregory P. Donoho; Philip W. Iversen; Youyan Zhang; Robert D. Van Horn; Amelie Forest; Ruslan D. Novosiadly; Yue Webster; Philip J. Ebert; Steven M. Bray; Jason C. Ting; Amit Aggarwal; James Robert Henry; Ramon V. Tiu; Gregory D. Plowman; Sheng Bin Peng

Purpose: To evaluate the antitumor efficacy of cetuximab in combination with LSN3074753, an analog of LY3009120 and pan-RAF inhibitor in 79 colorectal cancer patient-derived xenograft (PDX) models. Experimental Design: Seventy-nine well-characterized colorectal cancer PDX models were employed to conduct a single mouse per treatment group (n = 1) trial. Results: Consistent with clinical results, cetuximab was efficacious in wild-type KRAS and BRAF PDX models, with an overall response rate of 6.3% and disease control rate (DCR) of 20.3%. LSN3074753 was active in a small subset of PDX models that harbored KRAS or BRAF mutations. However, the combination treatment displayed the enhanced antitumor activity with DCR of 35.4%. Statistical analysis revealed that BRAF and KRAS mutations were the best predictors of the combinatorial activity and were significantly associated with synergistic effect with a P value of 0.01 compared with cetuximab alone. In 12 models with BRAF mutations, the combination therapy resulted in a DCR of 41.7%, whereas either monotherapy had a DCR of 8.3%. Among 44 KRAS mutation models, cetuximab or LSN3074753 monotherapy resulted in a DCR of 13.6% or 11.4%, respectively, and the combination therapy increased DCR to 34.1%. Molecular analysis suggests that EGFR activation is a potential feedback and resistant mechanism of pan-RAF inhibition. Conclusions: MAPK and EGFR pathway activations are two major molecular hallmarks of colorectal cancer. This mouse PDX trial recapitulated clinical results of cetuximab. Concurrent EGFR and RAF inhibition demonstrated synergistic antitumor activity for colorectal cancer PDX models with a KRAS or BRAF mutation. Clin Cancer Res; 23(18); 5547–60. ©2017 AACR.


Molecular Cancer Therapeutics | 2009

Abstract A62: A novel Eg5 inhibitor that causes mitotic arrest leading to rapid cancer cell death shows broad‐spectrum antitumor activity in preclinical xenograft tumor models

Xiang S. Ye; Li Fan; Robert D. Van Horn; Tinggui Yin; Ryuichiro Nakai; Yoshihisa Ohta; Kelly M. Credille; Gregory P. Donoho; Shiro Akinaga; Chikara Murakata; Everett J. Perkins; Scott Ocheltree; Yoshinori Yamashita; Kerry Blanchard; Eric Westin

Antitubulin agents including taxanes and vincas that target mitosis of rapidly dividing cancer cells are among the most effective cancer therapies in current clinical use. However, these antitubulin agents also have debilitating side effects that are dose‐limiting, such as neuropathy, due to their disruption of the normal microtubule functions in resting cells including neuronal cells. Eg5 is an evolutionarily conserved mitosis‐specific kinesin essential for bipolar mitotic spindle formation and has no roles in microtubule functions of resting cells. Inactivation of Eg5 causes mitotic arrest of proliferating cells, resulting in formation of monopolar spindles. Targeting Eg5 for cancer treatment thus represents an attractive strategy that has the potential to maximize the anticancer efficacy by inhibiting cancer cell mitosis while minimizing debilitating side effects associated with antitubulins. Here we describe a selective ATP‐non competitive small molecule inhibitor of human Eg5 kinesin. The Eg5 inhibitor shows no effects on microtubule dynamics in cell‐free assays and arrests cells specifically at mitosis with monopolar spindles, resulting in rapid cancer cell death. Growth inhibition assays against a panel of 21 cancer cell lines shows that the Eg5 inhibitor has potent and broad‐spectrum activity with IC50 values ranged from 0.55 nM to 14.2 nM. Quantitative live cell imaging and high content imaging reveal that the Eg5 inhibitor has a threshold concentration activity and kills cancer cells specifically at mitosis in a time/cell cycle, but not concentration above the threshold,‐dependent manner. Consistent with the in vitro activities, the Eg5 inhibitor shows broad‐spectrum antitumor activity in preclinical xenograft tumor models representing major human cancer histologies also including drug resistant tumors and demonstrates superiority as compared to several chemotherapeutic agents targeting G2/M. Furthermore, its in vivo antitumor activity is highly schedule‐dependent with a clear threshold dose effect, as expected from in vitro observations. Indeed, the Eg5 inhibitor exhibits a robust PK/PD relationship in antitumor activity and its antitumor activity is associated with mitotic arrest of cancer cells and subsequent cell death. The Eg5 inhibitor is currently being evaluated in Phase I studies. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A62.

Collaboration


Dive into the Gregory P. Donoho'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge