Network


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

Hotspot


Dive into the research topics where Makonen Belema is active.

Publication


Featured researches published by Makonen Belema.


Journal of General Virology | 2011

The effects of NS5A inhibitors on NS5A phosphorylation, polyprotein processing and localization

Dike Qiu; Julie A. Lemm; Donald R. O’Boyle; Jin-Hua Sun; Peter T. Nower; Van N. Nguyen; Lawrence G. Hamann; Lawrence B. Snyder; Daniel H. Deon; Edward H. Ruediger; Nicholas A. Meanwell; Makonen Belema; Min Gao; Robert A. Fridell

Hepatitis C virus (HCV) non-structural protein 5A (NS5A) is a multi-functional protein that is expressed in basally phosphorylated (p56) and in hyperphosphorylated (p58) forms. NS5A phosphorylation has been implicated in regulating multiple aspects of HCV replication. We recently reported the identification of a class of compounds that potently inhibit HCV RNA replication by targeting NS5A. Although the precise mechanism of inhibition of these compounds is not well understood, one activity that has been described is their ability to block expression of the hyperphosphorylated form of NS5A. Here, we report that an NS5A inhibitor impaired hyperphosphorylation without affecting basal phosphorylation at the C-terminal region of NS5A. This inhibitor activity did not require NS5A domains II and III and was distinct from that of a cellular kinase inhibitor that also blocked NS5A hyperphosphorylation, results that are consistent with an inhibitor-binding site within the N-terminal region of NS5A. In addition, we observed that an NS5A inhibitor promoted the accumulation of an HCV polyprotein intermediate, suggesting that inhibitor binding to NS5A may occur prior to the completion of polyprotein processing. Finally, we observed that NS5A p56 and p58 separated into different membrane fractions during discontinuous sucrose gradient centrifugation, consistent with these NS5A phosphoforms performing distinct replication functions. The p58 localization pattern was disrupted by an NS5A inhibitor. Collectively, our results suggest that NS5A inhibitors probably impact several aspects of HCV expression and regulation. These findings may help to explain the exceptional potency of this class of HCV replication complex inhibitors.


Virology | 2013

Characterizations of HCV NS5A replication complex inhibitors

Donald R. O'Boyle; Jin-Hua Sun; Peter T. Nower; Julie A. Lemm; Robert A. Fridell; Chunfu Wang; Jeffrey L. Romine; Makonen Belema; Van N. Nguyen; Denis R. St. Laurent; Michael H. Serrano-Wu; Lawrence B. Snyder; Nicholas A. Meanwell; David R. Langley; Min Gao

The hepatitis C virus NS5A protein is an established and clinically validated target for antiviral intervention by small molecules. Characterizations are presented of compounds identified as potent inhibitors of HCV replication to provide insight into structural elements that interact with the NS5A protein. UV-activated cross linking and affinity isolation was performed with one series to probe the physical interaction between the inhibitors and the NS5A protein expressed in HCV replicon cells. Resistance mapping with the second series was used to determine the functional impact of specific inhibitor subdomains on the interaction with NS5A. The data provide evidence for a direct high-affinity interaction between these inhibitors and the NS5A protein, with the interaction dependent on inhibitor stereochemistry. The functional data supports a model of inhibition that implicates inhibitor binding by covalently combining distinct pharmacophores across an NS5A dimer interface to achieve maximal inhibition of HCV replication.


Bioorganic & Medicinal Chemistry Letters | 2012

HCV NS5A replication complex inhibitors. Part 2: investigation of stilbene prolinamides.

Denis R. St. Laurent; Makonen Belema; Min Gao; Jason Goodrich; Ramesh Kakarla; Jay O. Knipe; Julie A. Lemm; Mengping Liu; Omar D. Lopez; Van N. Nguyen; Peter T. Nower; Donald R. O’Boyle; Yuping Qiu; Jeffrey L. Romine; Michael H. Serrano-Wu; Jin-Hua Sun; Lourdes Valera; Fukang Yang; Xuejie Yang; Nicholas A. Meanwell; Lawrence B. Snyder

In a previous disclosure,(1) we reported the dimerization of an iminothiazolidinone to form 1, a contributor to the observed inhibition of HCV genotype 1b replicon activity. The dimer was isolated via bioassay-guided fractionation experiments and shown to be a potent inhibitor of genotype 1b HCV replication for which resistance mapped to the NS5A protein. The elements responsible for governing HCV inhibitory activity were successfully captured in the structurally simplified stilbene prolinamide 2. We describe herein the early SAR and profiling associated with stilbene prolinamides that culminated in the identification of analogs with PK properties sufficient to warrant continued commitment to this chemotype. These studies represent the key initial steps toward the discovery of daclatasvir (BMS-790052), a compound that has demonstrated clinical proof-of-concept for inhibiting the NS5A replication complex in the treatment of HCV infection.


Bioorganic & Medicinal Chemistry Letters | 2013

HCV NS5A replication complex inhibitors. Part 3: discovery of potent analogs with distinct core topologies

Omar D. Lopez; Van N. Nguyen; Denis R. St. Laurent; Makonen Belema; Michael H. Serrano-Wu; Jason Goodrich; Fukang Yang; Yuping Qiu; Amy Ripka; Peter T. Nower; Lourdes Valera; Mengping Liu; Donald R. O’Boyle; Jin-Hua Sun; Robert A. Fridell; Julie A. Lemm; Min Gao; Andrew C. Good; Nicholas A. Meanwell; Lawrence B. Snyder

In a recent disclosure, we described the discovery of dimeric, prolinamide-based NS5A replication complex inhibitors exhibiting excellent potency towards an HCV genotype 1b replicon. That disclosure dealt with the SAR exploration of the peripheral region of our lead chemotype, and herein is described the SAR uncovered from a complementary effort that focused on the central core region. From this effort, the contribution of the core region to the overall topology of the pharmacophore, primarily vector orientation and planarity, was determined, with a set of analogs exhibiting <10 nM EC(50) in a genotype 1b replicon assay.


Bioorganic & Medicinal Chemistry Letters | 2013

HCV NS5A replication complex inhibitors. Part 5: discovery of potent and pan-genotypic glycinamide cap derivatives.

Makonen Belema; Van N. Nguyen; Denis R. St. Laurent; Omar D. Lopez; Yuping Qiu; Andrew C. Good; Peter T. Nower; Lourdes Valera; Donald R. O’Boyle; Jin-Hua Sun; Mengping Liu; Robert A. Fridell; Julie A. Lemm; Min Gao; Jay O. Knipe; Nicholas A. Meanwell; Lawrence B. Snyder

The isoquinolinamide series of HCV NS5A inhibitors exemplified by compounds 2b and 2c provided the first dual genotype-1a/1b (GT-1a/1b) inhibitor class that demonstrated a significant improvement in potency toward GT-1a replicons compared to that of the initial program lead, stilbene 2a. Structure-activity relationship (SAR) studies that uncovered an alternate phenylglycine-based cap series that exhibit further improvements in virology profile, along with some insights into the pharmacophoric elements associated with the GT-1a potency, are described.


Annual Reports in Medicinal Chemistry | 2011

Hepatitis C Virus—Progress Toward Inhibiting the Nonenzymatic Viral Proteins

Nicholas A. Meanwell; Makonen Belema

Publisher Summary This chapter focuses on the progress made toward the identification of potent and effective inhibitors of hepatitis C virus (HCV) that do not target the key enzymes NS3 protease and NS5B polymerase. The HCV NS3 protease and NS5B RNA-dependent RNA polymerase were inevitably the initial targets of focus because these enzymes were readily recapitulated functionally using biochemical assays, an effort considerably facilitated by the solving of X-ray crystallographic structures that informed structure-based drug design campaigns. The genomic arrangement of HCV is depicted in the chapter. The viral genome is translated as a single polyprotein that is cleaved by a combination of host cell proteases and the viral proteases NS2 and NS3. All viral proteins are plausible targets, and since many play multiple roles in replication, temporal selectivity based on unique protein function at different stages of the virus life cycle may be possible. Inhibitors of HCV NS5A are the most clinically advanced and appear to offer promise both as adjunct therapy and in combination with mechanistically complementary direct-acting antiviral agents.


Antimicrobial Agents and Chemotherapy | 2016

Synergistic Activity of Combined NS5A Inhibitors

Donald R. O'Boyle; Peter T. Nower; Min Gao; Robert A. Fridell; Chunfu Wang; Piyasena Hewawasam; Omar D. Lopez; Yong Tu; Nicholas A. Meanwell; Makonen Belema; Susan B. Roberts; Mark Cockett; Jin-Hua Sun

ABSTRACT Daclatasvir (DCV) is a first-in-class hepatitis C virus (HCV) nonstructural 5A replication complex inhibitor (NS5A RCI) that is clinically effective in interferon-free combinations with direct-acting antivirals (DAAs) targeting alternate HCV proteins. Recently, we reported NS5A RCI combinations that enhance HCV inhibitory potential in vitro, defining a new class of HCV inhibitors termed NS5A synergists (J. Sun, D. R. O’Boyle II, R. A. Fridell, D. R. Langley, C. Wang, S. Roberts, P. Nower, B. M. Johnson F. Moulin, M. J. Nophsker, Y. Wang, M. Liu, K. Rigat, Y. Tu, P. Hewawasam, J. Kadow, N. A. Meanwell, M. Cockett, J. A. Lemm, M. Kramer, M. Belema, and M. Gao, Nature 527:245–248, 2015, doi:10.1038/nature15711). To extend the characterization of NS5A synergists, we tested new combinations of DCV and NS5A synergists against genotype (gt) 1 to 6 replicons and gt 1a, 2a, and 3a viruses. The kinetics of inhibition in HCV-infected cells treated with DCV, an NS5A synergist (NS5A-Syn), or a combination of DCV and NS5A-Syn were distinctive. Similar to activity observed clinically, DCV caused a multilog drop in HCV, followed by rebound due to the emergence of resistance. DCV–NS5A-Syn combinations were highly efficient at clearing cells of viruses, in line with the trend seen in replicon studies. The retreatment of resistant viruses that emerged using DCV monotherapy with DCV–NS5A-Syn resulted in a multilog drop and rebound in HCV similar to the initial decline and rebound observed with DCV alone on wild-type (WT) virus. A triple combination of DCV, NS5A-Syn, and a DAA targeting the NS3 or NS5B protein cleared the cells of viruses that are highly resistant to DCV. Our data support the observation that the cooperative interaction of DCV and NS5A-Syn potentiates both the genotype coverage and resistance barrier of DCV, offering an additional DAA option for combination therapy and tools for explorations of NS5A function.


Drug Metabolism and Disposition | 2016

Biotransformation of Daclatasvir In Vitro and in Nonclinical Species: Formation of the Main Metabolite by Pyrrolidine δ-Oxidation and Rearrangement

Wenying Li; Weiping Zhao; Xiaohong Liu; Omar D. Lopez; John E. Leet; R. Marcus Fancher; Van N. Nguyen; Jason Goodrich; John A. Easter; Yang Hong; Janet Caceres-Cortes; Shu Y. Chang; Li Ma; Makonen Belema; Lawrence G. Hamann; Min Gao; Mingshe Zhu; Yue-Zhong Shu; W. Griffith Humphreys; Benjamin M. Johnson

Daclatasvir is a first-in-class, potent, and selective inhibitor of the hepatitis C virus nonstructural protein 5A replication complex. In support of nonclinical studies during discovery and exploratory development, liquid chromatography–tandem mass spectrometry and nuclear magnetic resonance were used in connection with synthetic and radiosynthetic approaches to investigate the biotransformation of daclatasvir in vitro and in cynomolgus monkeys, dogs, mice, and rats. The results of these studies indicated that disposition of daclatasvir was accomplished mainly by the release of unchanged daclatasvir into bile and feces and, secondarily, by oxidative metabolism. Cytochrome P450s were the main enzymes involved in the metabolism of daclatasvir. Oxidative pathways included δ-oxidation of the pyrrolidine moiety, resulting in ring opening to an aminoaldehyde intermediate followed by an intramolecular reaction between the aldehyde and the proximal imidazole nitrogen atom. Despite robust formation of the resulting metabolite in multiple systems, rates of covalent binding to protein associated with metabolism of daclatasvir were modest (55.2–67.8 pmol/mg/h) in nicotinamide adenine dinucleotide phosphate (reduced form)–supplemented liver microsomes (human, monkey, rat), suggesting that intramolecular rearrangement was favored over intermolecular binding in the formation of this metabolite. This biotransformation profile supported the continued development of daclatasvir, which is now marketed for the treatment of chronic hepatitis C virus infection.


Bioorganic & Medicinal Chemistry Letters | 2007

Synthesis and biological evaluation of 4-amino derivatives of benzimidazoquinoxaline, benzimidazoquinoline, and benzopyrazoloquinazoline as potent IKK inhibitors

Francis Beaulieu; Carl Ouellet; Edward H. Ruediger; Makonen Belema; Yuping Qiu; Xuejie Yang; Jacques Banville; James R. Burke; Kurt R. Gregor; John F. MacMaster; Alain Martel; Kim W. McIntyre; Mark A. Pattoli; F. Christopher Zusi; Dolatrai M. Vyas


Bioorganic & Medicinal Chemistry Letters | 2007

Synthesis and structure-activity relationship of imidazo (1,2 -a )thieno (3,2 -e )pyrazines as IKK-β inhibitors

Makonen Belema; Amy Bunker; Van N. Nguyen; Francis Beaulieu; Carl Ouellet; Yuping Qiu; Yunhui Zhang; Alain Martel; James R. Burke; Kim W. McIntyre; Mark A. Pattoli; Connie Daloisio; Kathleen M. Gillooly; Wendy Clarke; Patrick J. Brassil; F. Chris Zusi; Dolatrai M. Vyas

Collaboration


Dive into the Makonen Belema'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

Min Gao

Bristol-Myers Squibb

View shared research outputs
Researchain Logo
Decentralizing Knowledge