Network


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

Hotspot


Dive into the research topics where Mehran F. Moghaddam is active.

Publication


Featured researches published by Mehran F. Moghaddam.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of CC-930, an orally active anti-fibrotic JNK inhibitor.

Plantevin Krenitsky; Lisa Nadolny; Mercedes Delgado; Leticia Ayala; Steven S. Clareen; Robert Hilgraf; Ronald J. Albers; S Hegde; N D'Sidocky; John Sapienza; Jonathan Wright; Margaret A. McCarrick; Sogole Bahmanyar; Philip Chamberlain; S.L Delker; Jeff Muir; David Giegel; Li Xu; Maria Celeridad; J Lachowitzer; Brydon L. Bennett; Mehran F. Moghaddam; Oleg Khatsenko; Jason Katz; R Fan; April Bai; Yang Tang; Michael A. Shirley; B Benish; T Bodine

In this Letter we describe the discovery of potent, selective, and orally active aminopurine JNK inhibitors. Improving the physico-chemical properties as well as increasing the potency and selectivity of a subseries with rat plasma exposure, led to the identification of four structurally diverse inhibitors. Differentiation based on PK profiles in multiple species as well as activity in a chronic efficacy model led to the identification of 1 (CC-930) as a development candidate, which is currently in Phase II clinical trial for IPF.


PLOS ONE | 2013

Pomalidomide Shows Significant Therapeutic Activity against CNS Lymphoma with a Major Impact on the Tumor Microenvironment in Murine Models

Zhimin Li; Yushi Qiu; Peng Huang; Brandy Edenfield; Jason Katz; Darius Babusis; Yang Tang; Michael A. Shirely; Mehran F. Moghaddam; John A. Copland; Han W. Tun

Primary CNS lymphoma carries a poor prognosis. Novel therapeutic agents are urgently needed. Pomalidomide (POM) is a novel immunomodulatory drug with anti-lymphoma activity. CNS pharmacokinetic analysis was performed in rats to assess the CNS penetration of POM. Preclinical evaluation of POM was performed in two murine models to assess its therapeutic activity against CNS lymphoma. The impact of POM on the CNS lymphoma immune microenvironment was evaluated by immunohistochemistry and immunofluorescence. In vitro cell culture experiments were carried out to further investigate the impact of POM on the biology of macrophages. POM crosses the blood brain barrier with CNS penetration of ~ 39%. Preclinical evaluations showed that it had significant therapeutic activity against CNS lymphoma with significant reduction in tumor growth rate and prolongation of survival, that it had a major impact on the tumor microenvironment with an increase in macrophages and natural killer cells, and that it decreased M2-polarized tumor-associated macrophages and increased M1-polarized macrophages when macrophages were evaluated based on polarization status. In vitro studies using various macrophage models showed that POM converted the polarization status of IL4-stimulated macrophages from M2 to M1, that M2 to M1 conversion by POM in the polarization status of lymphoma-associated macrophages is dependent on the presence of NK cells, that POM induced M2 to M1 conversion in the polarization of macrophages by inactivating STAT6 signaling and activating STAT1 signaling, and that POM functionally increased the phagocytic activity of macrophages. Based on our findings, POM is a promising therapeutic agent for CNS lymphoma with excellent CNS penetration, significant preclinical therapeutic activity, and a major impact on the tumor microenvironment. It can induce significant biological changes in tumor-associated macrophages, which likely play a major role in its therapeutic activity against CNS lymphoma. POM should be further evaluated in clinical trials.


Bioorganic & Medicinal Chemistry Letters | 2013

Isosteric analogs of lenalidomide and pomalidomide: synthesis and biological activity.

Alexander L. Ruchelman; Hon-Wah Man; Weihong Zhang; Roger Shen-Chu Chen; Lori Capone; Jian Kang; Anastasia Parton; Laura G. Corral; Peter H. Schafer; Darius Babusis; Mehran F. Moghaddam; Yang Tang; Michael A. Shirley; George W. Muller

A series of analogs of the immunomodulary drugs lenalidomide (1) and pomalidomide (2), in which the amino group is replaced with various isosteres, was prepared and assayed for immunomodulatory activity and activity against cancer cell lines. The 4-methyl and 4-chloro analogs 4 and 15, respectively, displayed potent inhibition of tumor necrosis factor-α (TNF-α) in LPS-stimulated hPBMC, potent stimulation of IL-2 in a human T cell co-stimulation assay, and anti-proliferative activity against the Namalwa lymphoma cell line. Both of these analogs displayed oral bioavailability in rat.


Bioorganic & Medicinal Chemistry Letters | 2012

Aminopurine based JNK inhibitors for the prevention of ischemia reperfusion injury.

Véronique Plantevin Krenitsky; Mercedes Delgado; Lisa Nadolny; Kiran Sahasrabudhe; Leticia Ayala; Steven S. Clareen; Robert Hilgraf; Ronald J. Albers; Adam Kois; Kevin S. Hughes; Jonathan Wright; Jacek Nowakowski; Elise A. Sudbeck; Sutapa Ghosh; Sogole Bahmanyar; Philip Chamberlain; Jeff Muir; Brian E. Cathers; David Giegel; Li Xu; Maria Celeridad; Mehran F. Moghaddam; Oleg Khatsenko; Paul Omholt; Jason Katz; Sema Pai; Rachel Fan; Yang Tang; Michael A. Shirley; Brent Benish

In this Letter we describe the optimization of an aminopurine lead (1) with modest potency and poor overall kinase selectivity which led to the identification of a series of potent, selective JNK inhibitors. Improvement in kinase selectivity was enabled by introduction of an aliphatic side chain at the C-2 position. CC-359 (2) was selected as a potential clinical candidate for diseases manifested by ischemia reperfusion injury.


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 | 2012

Discovery and optimization of thieno[2,3-d]pyrimidines as B-Raf inhibitors.

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.


Drug Metabolism Letters | 2016

Efficiency in Drug Discovery: Liver S9 Fraction Assay As a Screen for Metabolic Stability.

Samantha J. Richardson; April Bai; Ashutosh Kulkarni; Mehran F. Moghaddam

Background: A rapid and comprehensive metabolic stability screen at the top of a drug discovery flow chart serves as an effective gate in eliminating low value compounds. This imparts a significant level of efficiency and saves valuable resources. While microsomes are amenable to high throughput automation and are cost effective, their enzymatic make-up is limited to that which is contained in endoplasmic reticulum, thereby informing only on Phase I metabolism. Lack of Phase II metabolism data can become a potential liability later in the process, adversely affecting discovery projects’ timelines and budget. Hepatocytes offer a full complement of metabolic enzymes and retain their cellular compartments, better representing liver metabolic function. However, hepatocyte screens are relatively expensive, labor intensive, and not easily automatable. Liver S9 fractions include Phase I and II metabolic enzymes, are relatively inexpensive, easy to use, and amenable to automation, making them a more appropriate screening system. We compare the data from the three systems and present the results. Results: Liver S9 and hepatocyte stability assays binned into the same category 70-84% of the time. Microsome and hepatocyte data were in agreement 73-82% of the time. The true rate for stability versus plasma clearance was 45% for hepatocytes and 43% for S9. Conclusion: In our opinion, replacing liver microsome and hepatocyte assays with S9 assay for high throughput metabolic screening purposes provides the combined benefit of comprehensive and high quality data at a reasonable expense for drug discovery programs.


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.


Drug Metabolism Letters | 2014

A Proposed Screening Paradigm for Discovery of Covalent Inhibitor Drugs

Mehran F. Moghaddam; Yang Tang; Zhihong O’Brien; Samantha J. Richardson; Maria Bacolod; Prasoon Chaturedi; Julius Apuy; Ashutosh Kulkarni

The in vitro and in vivo preclinical ADME properties of 10 clinically late stage or marketed covalent inhibitors were evaluated in order to define advancement criteria for discovery of future drugs in this arena. Our studies revealed the following: After incubating with S9 fractions for 30 minutes, the rat and human in vitro stability for these compounds ranged from 1% to 100%. The blood stability ranged from 30% to 100%. There was a broad range of CYP inhibition with prevalence for time-dependent inhibition of at least one enzyme. The Caco-2 permeability (A→B) ranged from negligible (0.6 x 10(-6) cm/s) to highly permeable (31 x 10(-6) cm/s) and the efflux ratio also varied widely (0.2-30). Most of the compounds were highly protein bound in both rat and human with binding ≥ 90%. Rat plasma clearance for the 10 compounds ranged from slow (11 mL/min/kg) to very rapid (350 mL/min/kg). The Vss ranged from low (0.67 L/kg) to very high (115 L/kg). MRTs also ranged from short (0.5 hr) to long (7.4 hr). The oral exposures also showed a very broad range with CMaxs ranging from 0.01-77 μM and exposure levels ranging from 0.03-106 μM.hr. In conclusion, the wide range in in vitro and in vivo ADME data makes these particular ADME assays non-discriminatory in the selection of promising compounds. In our opinion, non-traditional assays such as target mass modification, target confirmation by amino acid sequencing, cellular target occupancy, and target turnover rate data in combination with the pharmacokinetic profiles are the critical considerations for progression of irreversible compounds in early discovery.

Collaboration


Dive into the Mehran F. Moghaddam's collaboration.

Researchain Logo
Decentralizing Knowledge