Network


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

Hotspot


Dive into the research topics where Deborah Mortensen is active.

Publication


Featured researches published by Deborah Mortensen.


Cancer | 2015

A phase I dose-escalation study to assess safety, tolerability, pharmacokinetics, and preliminary efficacy of the dual mTORC1/mTORC2 kinase inhibitor CC-223 in patients with advanced solid tumors or multiple myeloma

Johanna C. Bendell; Kent C. Shih; Jennifer A. Grabowsky; Emily K. Bergsland; Suzanne F. Jones; Thomas G. Martin; Jeffrey R. Infante; Paul S. Mischel; Tomoo Matsutani; Shuichan Xu; Lilly Wong; Yong Liu; Xiaoling Wu; Deborah Mortensen; Rajesh Chopra; Kristen Hege; Pamela N. Munster

The mammalian target of rapamycin (mTOR) pathway is essential for tumor development, yet mTOR inhibitors have yielded modest results. This phase 1 study investigated the mTORC1/mTORC2 inhibitor CC‐223 in patients with advanced cancer.


Clinical Cancer Research | 2013

The mTOR Kinase Inhibitors, CC214-1 and CC214-2, Preferentially Block the Growth of EGFRvIII-Activated Glioblastomas

Beatrice Gini; Ciro Zanca; Deliang Guo; Tomoo Matsutani; Kenta Masui; Shiro Ikegami; Huijun Yang; David Nathanson; Genaro R. Villa; David Shackelford; Shaojun Zhu; Kazuhiro Tanaka; Ivan Babic; David Akhavan; Kelly Y. Lin; Alvaro Assuncao; Yuchao Gu; Bruno Bonetti; Deborah Mortensen; Shuichan Xu; Heather Raymon; Webster K. Cavenee; Frank Furnari; C. David James; Guido Kroemer; James R. Heath; Kristen Hege; Rajesh Chopra; Timothy F. Cloughesy; Paul S. Mischel

Purpose: mTOR pathway hyperactivation occurs in approximately 90% of glioblastomas, but the allosteric mTOR inhibitor rapamycin has failed in the clinic. Here, we examine the efficacy of the newly discovered ATP-competitive mTOR kinase inhibitors CC214-1 and CC214-2 in glioblastoma, identifying molecular determinants of response and mechanisms of resistance, and develop a pharmacologic strategy to overcome it. Experimental Design: We conducted in vitro and in vivo studies in glioblastoma cell lines and an intracranial model to: determine the potential efficacy of the recently reported mTOR kinase inhibitors CC214-1 (in vitro use) and CC214-2 (in vivo use) at inhibiting rapamycin-resistant signaling and blocking glioblastoma growth and a novel single-cell technology—DNA Encoded Antibody Libraries—was used to identify mechanisms of resistance. Results: Here, we show that CC214-1 and CC214-2 suppress rapamycin-resistant mTORC1 signaling, block mTORC2 signaling, and significantly inhibit the growth of glioblastomas in vitro and in vivo. EGFRvIII expression and PTEN loss enhance sensitivity to CC214 compounds, consistent with enhanced efficacy in strongly mTOR-activated tumors. Importantly, CC214 compounds potently induce autophagy, preventing tumor cell death. Genetic or pharmacologic inhibition of autophagy greatly sensitizes glioblastoma cells and orthotopic xenografts to CC214-1- and CC214-2–induced cell death. Conclusions: These results identify CC214-1 and CC214-2 as potentially efficacious mTOR kinase inhibitors in glioblastoma, and suggest a strategy for identifying patients most likely to benefit from mTOR inhibition. In addition, this study also shows a central role for autophagy in preventing mTOR-kinase inhibitor-mediated tumor cell death, and suggests a pharmacologic strategy for overcoming it. Clin Cancer Res; 19(20); 5722–32. ©2013 AACR.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery and SAR exploration of a novel series of imidazo[4,5-b]pyrazin-2-ones as potent and selective mTOR kinase inhibitors.

Deborah Mortensen; Sophie Perrin-Ninkovic; Roy Harris; Branden Lee; Graziella I. Shevlin; Matt Hickman; Gody Khambatta; René R. Bisonette; Kimberly Elizabeth Fultz; Sabita Sankar

We report here the discovery of a novel series of selective mTOR kinase inhibitors. A series of imidazo[4,5-b]pyrazin-2-ones, represented by screening hit 1, was developed into lead compounds with excellent mTOR potency and exquisite kinase selectivity. Potent compounds from this series show >1000-fold selectivity over the related PI3Kα lipid kinase. Further, compounds such as 2 achieve mTOR pathway inhibition, blocking both mTORC1 and mTORC2 signaling, in PC3 cancer cells as measured by inhibition of pS6 and pAkt (S473).


Molecular Cancer Therapeutics | 2015

CC-223, a Potent and Selective Inhibitor of mTOR Kinase: In Vitro and In Vivo Characterization.

Deborah Mortensen; Kimberly Elizabeth Fultz; Shuichan Xu; Weiming Xu; Garrick Packard; Godrej Khambatta; James C. Gamez; Jim Leisten; Jingjing Zhao; Julius Apuy; Kamran Ghoreishi; Matt Hickman; Rama Krishna Narla; Rene Bissonette; Samantha J. Richardson; Sophie X. Peng; Sophie Perrin-Ninkovic; Tam Tran; Tao Shi; Wen Qing Yang; Zeen Tong; Brian E. Cathers; Mehran F. Moghaddam; Stacie S. Canan; Peter Worland; Sabita Sankar; Heather Raymon

mTOR is a serine/threonine kinase that regulates cell growth, metabolism, proliferation, and survival. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are critical mediators of the PI3K–AKT pathway, which is frequently mutated in many cancers, leading to hyperactivation of mTOR signaling. Although rapamycin analogues, allosteric inhibitors that target only the mTORC1 complex, have shown some clinical activity, it is hypothesized that mTOR kinase inhibitors, blocking both mTORC1 and mTORC2 signaling, will have expanded therapeutic potential. Here, we describe the preclinical characterization of CC-223. CC-223 is a potent, selective, and orally bioavailable inhibitor of mTOR kinase, demonstrating inhibition of mTORC1 (pS6RP and p4EBP1) and mTORC2 [pAKT(S473)] in cellular systems. Growth inhibitory activity was demonstrated in hematologic and solid tumor cell lines. mTOR kinase inhibition in cells, by CC-223, resulted in more complete inhibition of the mTOR pathway biomarkers and improved antiproliferative activity as compared with rapamycin. Growth inhibitory activity and apoptosis was demonstrated in a panel of hematologic cancer cell lines. Correlative analysis revealed that IRF4 expression level associates with resistance, whereas mTOR pathway activation seems to associate with sensitivity. Treatment with CC-223 afforded in vivo tumor biomarker inhibition in tumor-bearing mice, after a single oral dose. CC-223 exhibited dose-dependent tumor growth inhibition in multiple solid tumor xenografts. Significant inhibition of mTOR pathway markers pS6RP and pAKT in CC-223–treated tumors suggests that the observed antitumor activity of CC-223 was mediated through inhibition of both mTORC1 and mTORC2. CC-223 is currently in phase I clinical trials. Mol Cancer Ther; 14(6); 1295–305. ©2015 AACR.


Journal of Medicinal Chemistry | 2015

Optimization of a Series of Triazole Containing Mammalian Target of Rapamycin (mTOR) Kinase Inhibitors and the Discovery of CC-115

Deborah Mortensen; Sophie Perrin-Ninkovic; Graziella I. Shevlin; Jan Elsner; Jingjing Zhao; Brandon Wade Whitefield; Lida Tehrani; John Sapienza; Jennifer Riggs; Jason Parnes; Patrick Papa; Garrick Packard; Branden Lee; Roy Harris; Matthew Correa; Sogole Bahmanyar; Samantha J. Richardson; Sophie X. Peng; Jim Leisten; Godrej Khambatta; Matt Hickman; James C. Gamez; René R. Bisonette; Julius L. Apuy; Brian E. Cathers; Stacie S. Canan; Mehran F. Moghaddam; Heather Raymon; Peter J. Worland; Rama Krishna Narla

We report here the synthesis and structure-activity relationship (SAR) of a novel series of triazole containing mammalian target of rapamycin (mTOR) kinase inhibitors. SAR studies examining the potency, selectivity, and PK parameters for a series of triazole containing 4,6- or 1,7-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones resulted in the identification of triazole containing mTOR kinase inhibitors with improved PK properties. Potent compounds from this series were found to block both mTORC1(pS6) and mTORC2(pAktS473) signaling in PC-3 cancer cells, in vitro and in vivo. When assessed in efficacy models, analogs exhibited dose-dependent efficacy in tumor xenograft models. This work resulted in the selection of CC-115 for clinical development.


Bioorganic & Medicinal Chemistry Letters | 2013

Use of core modification in the discovery of CC214-2, an orally available, selective inhibitor of mTOR kinase

Deborah Mortensen; John Sapienza; Branden Lee; Sophie Perrin-Ninkovic; Roy Harris; Graziella I. Shevlin; Jason Parnes; Brandon Wade Whitefield; Matt Hickman; Gody Khambatta; René R. Bisonette; Sophie X. Peng; Jim Gamez; Jim Leisten; Rama Krishna Narla; Kimberly Elizabeth Fultz; Sabita Sankar

We report here the discovery of a novel series of selective mTOR kinase inhibitors and the identification of CC214-2, a compound with demonstrated anti-tumor activity upon oral dosing in a PC3 prostate cancer xenograft model. A series of 4,6-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones were discovered through a core modification of our original compound series. Analogs from this series have excellent mTOR potency and maintain selectivity over the related PI3Kα lipid kinase. Compounds such as CC214-2 were found to block both mTORC1(pS6) and mTORC2(pAktS473) signaling in PC3 cancer cells, in vitro and in vivo.


Journal of Medicinal Chemistry | 2015

Discovery of Mammalian Target of Rapamycin (mTOR) Kinase Inhibitor CC-223

Deborah Mortensen; Sophie Perrin-Ninkovic; Graziella I. Shevlin; Jingjing Zhao; Garrick Packard; Sogole Bahmanyar; Matthew Correa; Jan Elsner; Roy Harris; Branden Lee; Patrick Papa; Jason Parnes; Jennifer Riggs; John Sapienza; Lida Tehrani; Brandon Wade Whitefield; Julius L. Apuy; René R. Bisonette; James C. Gamez; Matt Hickman; Godrej Khambatta; Jim Leisten; Sophie X. Peng; Samantha J. Richardson; Brian E. Cathers; Stacie S. Canan; Mehran F. Moghaddam; Heather Raymon; Peter J. Worland; Rama Krishna Narla

We report here the synthesis and structure-activity relationship (SAR) of a novel series of mammalian target of rapamycin (mTOR) kinase inhibitors. A series of 4,6- or 1,7-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones were optimized for in vivo efficacy. These efforts resulted in the identification of compounds with excellent mTOR kinase inhibitory potency, with exquisite kinase selectivity over the related lipid kinase PI3K. The improved PK properties of this series allowed for exploration of in vivo efficacy and ultimately the selection of CC-223 for clinical development.


Oncotarget | 2017

CC-115, a dual inhibitor of mTOR Kinase and DNA-PK, blocks DNA damage repair pathways and selectively inhibits ATM-deficient cell growth in vitro

Toshiya Tsuji; Lisa M. Sapinoso; Tam Tran; Bonny Gaffney; Lilly Wong; Sabita Sankar; Heather Raymon; Deborah Mortensen; Shuichan Xu

CC-115, a selective dual inhibitor of the mammalian target of rapamycin (mTOR) kinase and DNA-dependent protein kinase (DNA-PK), is undergoing Phase 1 clinical studies. Here we report the characterization of DNA-PK inhibitory activity of CC-115 in cancer cell lines. CC-115 inhibits auto-phosphorylation of the catalytic subunit of DNA-PK (DNA-PKcs) at the S2056 site (pDNA-PK S2056), leading to blockade of DNA-PK-mediated non-homologous end joining (NHEJ). CC-115 also indirectly reduces the phosphorylation of ataxia-telangiectasia mutated kinase (ATM) at S1981 and its substrates as well as homologous recombination (HR). The mTOR kinase and DNA-PK inhibitory activity of CC-115 leads to not only potent anti-tumor activity against a large panel of hematopoietic and solid cancer cell lines but also strong induction of apoptosis in a subset of cancer lines. Mechanistically, CC-115 prevents NHEJ by inhibiting the dissociation of DNA-PKcs, X-ray repair cross-complementing protein 4 (XRCC4), and DNA ligase IV from DNA ends. CC-115 inhibits colony formation of ATM-deficient cells more potently than ATM-proficient cells, indicating that inhibition of DNA-PK is synthetically lethal with the loss of functional ATM. In conclusion, CC-115 inhibits both mTOR signaling and NHEJ and HR by direct inhibition of DNA-PK. The mechanistic data not only provide selection of potential pharmacodynamic (PD) markers but also support CC-115 clinical development in patients with ATM-deficient tumors.


Molecular Cancer Therapeutics | 2014

Genetic and Pharmacologic Evidence That mTOR Targeting Outweighs mTORC1 Inhibition as an Antimyeloma Strategy

Xi Chen; Elena Díaz-Rodríguez; Enrique M. Ocio; Bruno Paiva; Deborah Mortensen; Antonia Lopez-Girona; Rajesh Chopra; Jes us San Miguel; Atanasio Pandiella

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth, proliferation, metabolism, and cell survival, and plays those roles by forming two functionally distinct multiprotein complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Deregulation of the mTOR pathway has been found in different cancers, including multiple myeloma. Agents acting on mTORC1, such as rapamycin and derivatives, are being explored as antitumoral strategies. However, whether targeting mTOR would be a more effective antimyeloma strategy than exclusively acting on the mTORC1 branch remains to be established. In this report, we explored the activation status of mTOR routes in malignant plasma cells, and analyzed the contribution of mTOR and its two signaling branches to the proliferation of myeloma cells. Gene expression profiling demonstrated deregulation of mTOR pathway–related genes in myeloma plasma cells from patients. Activation of the mTOR pathway in myelomatous plasma cells was corroborated by flow cytometric analyses. RNA interference (RNAi) experiments indicated that mTORC1 predominated over mTORC2 in the control of myeloma cell proliferation. However, mTOR knockdown had a superior antiproliferative effect than acting only on mTORC1 or mTORC2. Pharmacologic studies corroborated that the neutralization of mTOR has a stronger antimyeloma effect than the individual inhibition of mTORC1 or mTORC2. Together, our data support the clinical development of agents that widely target mTOR, instead of agents, such as rapamycin or its derivatives, that solely act on mTORC1. Mol Cancer Ther; 13(2); 504–16. ©2014 AACR.


ACS Medicinal Chemistry Letters | 2018

Discovery of a Stress-Activated Protein Kinase Inhibitor for Lymphatic Filariasis

Sreedhar Reddy Tummalapalli; Rohit Bhat; Agnieska Chojnowski; Monika Prorok; Tamara Kreiss; Ronald Goldberg; Stacie S. Canan; Natalie Hawryluk; Deborah Mortensen; Vikram Khetani; Jerome B. Zeldis; John J. Siekierka; David P. Rotella

Lymphatic filariasis infects over 120 million people worldwide and can lead to significant disfigurement and disease. Resistance is emerging with current treatments, and these therapies have dose limiting adverse events; consequently new targets are needed. One approach to achieve this goal is inhibition of parasitic protein kinases involved in circumventing host defense mechanisms. This report describes structure-activity relationships leading to the identification of a potent, orally bioavailable stress activated protein kinase inhibitor that may be used to investigate this hypothesis.

Collaboration


Dive into the Deborah Mortensen's collaboration.

Researchain Logo
Decentralizing Knowledge