Jim Leisten
Celgene
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jim Leisten.
Journal of Medicinal Chemistry | 2009
Hon-Wah Man; Peter H. Schafer; Lu Min Wong; Rebecca T. Patterson; Laura G. Corral; Heather Raymon; Kate Blease; Jim Leisten; Michael A. Shirley; Yang Tang; Darius Babusis; Roger Shen-Chu Chen; Dave Stirling; George W. Muller
In this communication, we report the discovery of 1S (apremilast), a novel potent and orally active phosphodiesterase 4 (PDE4) and tumor necrosis factor-alpha inhibitor. The optimization of previously reported 3-(1,3-dioxo-1,3-dihydroisoindol-2-yl)-3-(3,4-dimethoxyphenyl)propionic acid PDE4 inhibitors led to this series of sulfone analogues. Evaluation of the structure-activity relationship of substitutions on the phthalimide group led to the discovery of an acetylamino analogue 1S, which is currently in clinical trials.
British Journal of Haematology | 2014
Soraya Carrancio; Jennifer Markovics; Piu Wong; Jim Leisten; Paola Castiglioni; Matthew C. Groza; Heather Raymon; Carla Heise; Tom Daniel; Rajesh Chopra; Victoria Sung
Sotatercept (ACE‐011), a recombinant human fusion protein containing the extracellular domain of the human Activin receptor IIA, binds to and inhibits activin and other members of the transforming growth factor ‐β (TGF‐β) superfamily. Administration of sotatercept led to a rapid and sustained increase in red blood cell (RBC) count and haemoglobin (Hb) in healthy volunteers (phase I clinical trials), but the mechanism is not fully understood. Mice treated with RAP‐011 (murine ortholog of ACE‐011) respond with a rapid (within 24 h) increase in haematocrit, Hb, and RBC count. These effects are accompanied by an equally rapid stimulation of late‐stage erythroid precursors in the bone marrow (BM). RAP‐011 also induces a significant increase in erythroid burst‐forming units and erythropoietin, which could contribute to additional, sustained effects on RBC production. Further in vitro co‐culture studies demonstrate that BM accessory cells are required for RAP‐011 effects. To better understand which TGF‐β family ligand(s) mediate RAP‐011 effects, we evaluated the impact of several of these ligands on erythroid differentiation. Our data suggest that RAP‐011 may act to rescue growth differentiation factor 11/Activin A‐induced inhibition of late‐stage erythropoiesis. These data define the mechanism of action of a novel agent that regulates RBC differentiation and provide the rationale to develop sotatercept for the treatment of anaemia and ineffective erythropoiesis.
Molecular Cancer Therapeutics | 2015
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
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
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.
Bioorganic & Medicinal Chemistry Letters | 2012
Garrick Packard; Patrick Papa; Jennifer Riggs; Paul E. Erdman; Lida Tehrani; Dale Robinson; Roy Harris; Graziella I. Shevlin; Sophie Perrin-Ninkovic; Robert Hilgraf; Margaret A. McCarrick; Tam Tran; Yuedi W. Fleming; April Bai; Samantha J. Richardson; Jason Katz; Yang Tang; Jim Leisten; Mehran F. Moghaddam; Brian E. Cathers; Dan Zhu; Steven T. Sakata
The serine/threonine specific protein kinase B-Raf is part of the MAPK pathway and is an interesting oncology target. We have identified thieno[2,3-d]pyrimidines as a core scaffold of small molecule B-Raf inhibitors. The SAR of analogs in this series will be described.
Journal of Medicinal Chemistry | 2015
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.
British Journal of Haematology | 2016
Emily Rychak; Derek Mendy; Tao Shi; Yuhong Ning; Jim Leisten; Ling Lu; Karen Miller; Rama Krishna Narla; Robert Z. Orlowski; Heather Raymon; Chad C. Bjorklund; Anjan Thakurta; Anita Gandhi; Brian E. Cathers; Rajesh Chopra; Thomas O. Daniel; Antonia Lopez-Girona
Pomalidomide is an IMiD® immunomodulatory agent, which has shown clinically significant benefits in relapsed and/or refractory multiple myeloma (rrMM) patients when combined with dexamethasone, regardless of refractory status to lenalidomide or bortezomib. (Schey et al, ; San Miguel et al, 2013; Richardson et al, 2014; Scott, ) In this work, we present preclinical data showing that the combination of pomalidomide with dexamethasone (PomDex) demonstrates potent anti‐proliferative and pro‐apoptotic activity in both lenalidomide‐sensitive and lenalidomide‐resistant MM cell lines. PomDex also synergistically inhibited tumour growth compared with single‐agent treatment in xenografts of lenalidomide‐resistant H929 R10‐1 cells. Typical hallmarks of IMiD compound activity, including IKZF3 (Aiolos) degradation, and the downregulation of interferon regulatory factor (IRF) 4 and MYC, seen in lenalidomide‐sensitive H929 MM cell lines, were also observed in PomDex‐treated lenalidomide‐resistant H929 MM cells. Remarkably, this resulted in strong, synergistic effects on the induction of apoptosis in both lenalidomide‐sensitive and resistant MM cells. Furthermore, gene expression profiling revealed a unique differential gene expression pattern in PomDex‐treated samples, highlighted by the modulation of pro‐apoptotic pathways in lenalidomide‐resistant cells. These results provide key insights into molecular mechanisms of PomDex in the lenalidomide‐resistant setting.
Cancer Research | 2017
Rama Krishna Narla; Hardik Modi; Lilly Wong; Mahan Abassian; Daniel Bauer; Pragnya Desai; Bonny Gaffney; Pilgrim Jackson; Jim Leisten; Jing Liu; Antonia Lopez-Girona; Maria S. Romero; WenQing Yang; Brendan P. Eckelman; Quinn Deveraux; Laurie Phillips; Heather Raymon; Laure Escoubet; John Boylan; Kandasamy Hariharan
Cluster of differentiation 47 (CD47) is a transmembrane protein ubiquitously expressed on human cells but overexpressed on many different tumor cells. The interaction of CD47 with signal-regulatory protein alpha (SIRPα) expressed on macrophages results in the inhibition of phagocytosis. Thus, tumor cell overexpression of CD47 enables escape from immune surveillance via the blockade of phagocytic mechanisms. We report here the characterization of CC-90002, a humanized anti-CD47 monoclonal antibody (mAb) that is being developed as a potential therapeutic for hematologic malignancies and solid tumors. CC-90002 has a high affinity for binding to CD47 with a subnanomolar dissociation constant (Kd) value. The IC50 required for the blockade of the CD47-SIRPα interaction was also in the subnanomolar range. CC-90002 is unique among previously reported anti-CD47 antibodies for its inability to promote hemagglutination while maintaining high affinity binding to CD47 and inhibition of the CD47-SIRPα interaction. CC-90002 enabled antibody-mediated phagocytosis of a panel of hematological cancer cell lines in vitro, including ALL, multiple myeloma (MM) and acute myeloid leukemia (AML) cells, and primary AML patient samples. The phagocytosis index for this panel (at 1 μg/mL) ranged from approximately 20% to 60% for all the cell lines tested. Antibody concentration-response studies indicated that the CC-90002 effect was concentration-dependent in the ALL and AML cell lines. CC-90002 treatment significantly enabled the phagocytosis of two lenalidomide-resistant MM cell lines and primary AML cells from three patients. Additionally, CC-90002 treatment elicited the phagocytosis of solid tumor lines, including those from ovarian, breast, head and neck, lung, and pancreatic cancers. The in vivo efficacy of CC-90002 was evaluated across cell line-derived and patient-derived xenograft models. Significant dose-dependent antitumor activity was observed with CC-90002 treatment in the MM cell line-derived xenograft models, RPMI 8226 and the parental NCI-H929 and lenalidomide-resistant NCI-H929 (H929/R1). Treatment with CC-90002 demonstrated significant tumor regression in solid tumor xenografts including a cell line-derived model of triple negative breast cancer (TNBC), MDA-MB-231, and a patient-derived TNBC model, AA1126 and significantly prolonged survival in the HL-60-disseminated AML tumor model. Mechanistic studies in the RPMI 8226 xenograft model confirmed binding of CC-90002 to tumor cells and recruitment of F4-80-positive macrophages into the tumor. RPMI 8226 tumor lysates from CC-90002-treated animals demonstrated an increase in select chemokines and cytokines of murine origin. Taken together, the in vitro and in vivo data demonstrate the potential for activity of CC-90002 across both hematological malignancies and solid tumors. CC-90002 is currently in early clinical development. Citation Format: Rama Krishna Narla, Hardik Modi, Lilly Wong, Mahan Abassian, Daniel Bauer, Pragnya Desai, Bonny Gaffney, Pilgrim Jackson, Jim Leisten, Jing Liu, Antonia Lopez-Girona, Maria Romero, WenQing Yang, Brendan P. Eckelman, Quinn Deveraux, Laurie Phillips, Heather K. Raymon, Laure Escoubet, John Boylan, Kandasamy Hariharan. The humanized anti-CD47 monclonal antibody, CC-90002, has antitumor activity in vitro and in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4694. doi:10.1158/1538-7445.AM2017-4694
Bioorganic & Medicinal Chemistry | 2011
Alexander L. Ruchelman; Hon-Wah Man; Roger Shen-Chu Chen; Wei Liu; Ling Lu; Dorota Cedzik; Ling Zhang; Jim Leisten; Alice Collette; Rama Krishna Narla; Heather Raymon; George W. Muller