Network


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

Hotspot


Dive into the research topics where Chudi Ndubaku is active.

Publication


Featured researches published by Chudi Ndubaku.


ACS Medicinal Chemistry Letters | 2014

Discovery of a Potent and Selective BCL-XL Inhibitor with in Vivo Activity

Zhi-Fu Tao; Lisa A. Hasvold; Le Wang; Xilu Wang; Andrew M. Petros; Chang H. Park; Erwin R. Boghaert; Nathaniel D. Catron; Jun Chen; Peter M. Colman; Peter E. Czabotar; Kurt Deshayes; Wayne J. Fairbrother; John A. Flygare; Sarah G. Hymowitz; Sha Jin; Russell A. Judge; Michael F. T. Koehler; Peter Kovar; Guillaume Lessene; Michael J. Mitten; Chudi Ndubaku; Paul Nimmer; Hans E. Purkey; Anatol Oleksijew; Darren C. Phillips; Brad E. Sleebs; Brian J. Smith; Morey L. Smith; Stephen K. Tahir

A-1155463, a highly potent and selective BCL-XL inhibitor, was discovered through nuclear magnetic resonance (NMR) fragment screening and structure-based design. This compound is substantially more potent against BCL-XL-dependent cell lines relative to our recently reported inhibitor, WEHI-539, while possessing none of its inherent pharmaceutical liabilities. A-1155463 caused a mechanism-based and reversible thrombocytopenia in mice and inhibited H146 small cell lung cancer xenograft tumor growth in vivo following multiple doses. A-1155463 thus represents an excellent tool molecule for studying BCL-XL biology as well as a productive lead structure for further optimization.


ACS Chemical Biology | 2009

Antagonism of c-IAP and XIAP proteins is required for efficient induction of cell death by small-molecule IAP antagonists.

Chudi Ndubaku; Eugene Varfolomeev; Lan Wang; Kerry Zobel; Kevin Lau; Linda O. Elliott; Brigitte Maurer; Anna V. Fedorova; Jasmin N. Dynek; Michael F. T. Koehler; Sarah G. Hymowitz; Vickie Tsui; Kurt Deshayes; Wayne J. Fairbrother; John A. Flygare; Domagoj Vucic

The inhibitor of apoptosis (IAP) proteins are critical regulators of cancer cell survival, which makes them attractive targets for therapeutic intervention in cancers. Herein, we describe the structure-based design of IAP antagonists with high affinities and selectivity (>2000-fold) for c-IAP1 over XIAP and their functional characterization as activators of apoptosis in tumor cells. Although capable of inducing cell death and preventing clonogenic survival, c-IAP-selective antagonists are significantly less potent in promoting apoptosis when compared to pan-selective compounds. However, both pan-IAP- and c-IAP-selective antagonists stimulate c-IAP1 and c-IAP2 degradation and activation of NF-kappaB pathways with comparable potencies. Therefore, although compounds that specifically target c-IAP1 and c-IAP2 are capable of inducing apoptosis, antagonism of the c-IAP proteins and XIAP is required for efficient induction of cancer cell death by IAP antagonists.


Journal of Medicinal Chemistry | 2013

Discovery of 2-{3-[2-(1-Isopropyl-3-methyl-1H-1,2–4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl]-1H-pyrazol-1-yl}-2-methylpropanamide (GDC-0032): A β-Sparing Phosphoinositide 3-Kinase Inhibitor with High Unbound Exposure and Robust in Vivo Antitumor Activity

Chudi Ndubaku; Timothy P. Heffron; Steven Staben; Matthew Baumgardner; Nicole Blaquiere; Erin K. Bradley; Richard James Bull; Steven Do; Jennafer Dotson; Danette Dudley; Kyle A. Edgar; Lori Friedman; Richard Goldsmith; Robert Heald; Aleksandr Kolesnikov; Leslie Lee; Cristina Lewis; Michelle Nannini; Jim Nonomiya; Jodie Pang; Steve Price; Wei Wei Prior; Laurent Salphati; Steve Sideris; Jeffery J. Wallin; Lan Wang; Binqing Wei; Deepak Sampath; Alan G. Olivero

Dysfunctional signaling through the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway leads to uncontrolled tumor proliferation. In the course of the discovery of novel benzoxepin PI3K inhibitors, we observed a strong dependency of in vivo antitumor activity on the free-drug exposure. By lowering the intrinsic clearance, we derived a set of imidazobenzoxazepin compounds that showed improved unbound drug exposure and effectively suppressed growth of tumors in a mouse xenograft model at low drug dose levels. One of these compounds, GDC-0032 (11l), was progressed to clinical trials and is currently under phase I evaluation as a potential treatment for human malignancies.


Nature | 2015

MAP4K4 regulates integrin-FERM binding to control endothelial cell motility

Philip Vitorino; Stacey Yeung; Ailey Crow; Jesse Bakke; Tanya Smyczek; Kristina West; Erin McNamara; Jeffrey Eastham-Anderson; Stephen Jay Gould; Seth F. Harris; Chudi Ndubaku; Weilan Ye

Cell migration is a stepwise process that coordinates multiple molecular machineries. Using in vitro angiogenesis screens with short interfering RNA and chemical inhibitors, we define here a MAP4K4–moesin–talin–β1-integrin molecular pathway that promotes efficient plasma membrane retraction during endothelial cell migration. Loss of MAP4K4 decreased membrane dynamics, slowed endothelial cell migration, and impaired angiogenesis in vitro and in vivo. In migrating endothelial cells, MAP4K4 phosphorylates moesin in retracting membranes at sites of focal adhesion disassembly. Epistasis analyses indicated that moesin functions downstream of MAP4K4 to inactivate integrin by competing with talin for binding to β1-integrin intracellular domain. Consequently, loss of moesin (encoded by the MSN gene) or MAP4K4 reduced adhesion disassembly rate in endothelial cells. Additionally, α5β1-integrin blockade reversed the membrane retraction defects associated with loss of Map4k4 in vitro and in vivo. Our study uncovers a novel aspect of endothelial cell migration. Finally, loss of MAP4K4 function suppressed pathological angiogenesis in disease models, identifying MAP4K4 as a potential therapeutic target.


Journal of Medicinal Chemistry | 2015

Inhibiting the deubiquitinating enzymes (DUBs).

Chudi Ndubaku; Vickie Tsui

The diverse roles of deubiquitinating enzymes, or DUBs, in determining the fate of specific proteins continue to unfold. Concurrent with the revelation of DUBs as potential therapeutic targets are publications of small molecule inhibitors of these enzymes. In this review, we summarize these molecules and their associated data and suggest additional experiments to further validate and characterize these compounds. We believe the field of drug discovery against DUBs is still in its infancy, but advances in assay development, biophysical techniques, and screening libraries hold promise for identifying suitable agents that could advance into the clinic.


Nature | 2017

USP7 small-molecule inhibitors interfere with ubiquitin binding

Lorna Kategaya; Paola Di Lello; Lionel Rouge; Richard Pastor; Kevin R. Clark; Jason Drummond; Tracy Kleinheinz; Eva Lin; John-Paul Upton; Sumit Prakash; Johanna Heideker; Mark L. McCleland; Maria Stella Ritorto; Dario R. Alessi; Matthias Trost; Travis W. Bainbridge; Michael C. M. Kwok; Taylur P. Ma; Zachary Stiffler; Bradley Brasher; Yinyan Tang; Priyadarshini Jaishankar; Brian R. Hearn; Adam R. Renslo; Michelle R. Arkin; Fred E. Cohen; Kebing Yu; Frank Peale; Florian Gnad; Matthew T. Chang

The ubiquitin system regulates essential cellular processes in eukaryotes. Ubiquitin is ligated to substrate proteins as monomers or chains and the topology of ubiquitin modifications regulates substrate interactions with specific proteins. Thus ubiquitination directs a variety of substrate fates including proteasomal degradation. Deubiquitinase enzymes cleave ubiquitin from substrates and are implicated in disease; for example, ubiquitin-specific protease-7 (USP7) regulates stability of the p53 tumour suppressor and other proteins critical for tumour cell survival. However, developing selective deubiquitinase inhibitors has been challenging and no co-crystal structures have been solved with small-molecule inhibitors. Here, using nuclear magnetic resonance-based screening and structure-based design, we describe the development of selective USP7 inhibitors GNE-6640 and GNE-6776. These compounds induce tumour cell death and enhance cytotoxicity with chemotherapeutic agents and targeted compounds, including PIM kinase inhibitors. Structural studies reveal that GNE-6640 and GNE-6776 non-covalently target USP7 12 Å distant from the catalytic cysteine. The compounds attenuate ubiquitin binding and thus inhibit USP7 deubiquitinase activity. GNE-6640 and GNE-6776 interact with acidic residues that mediate hydrogen-bond interactions with the ubiquitin Lys48 side chain, suggesting that USP7 preferentially interacts with and cleaves ubiquitin moieties that have free Lys48 side chains. We investigated this idea by engineering di-ubiquitin chains containing differential proximal and distal isotopic labels and measuring USP7 binding by nuclear magnetic resonance. This preferential binding protracted the depolymerization kinetics of Lys48-linked ubiquitin chains relative to Lys63-linked chains. In summary, engineering compounds that inhibit USP7 activity by attenuating ubiquitin binding suggests opportunities for developing other deubiquitinase inhibitors and may be a strategy more broadly applicable to inhibiting proteins that require ubiquitin binding for full functional activity.


Journal of Medicinal Chemistry | 2014

Discovery of Selective 4-Amino-pyridopyrimidine Inhibitors of MAP4K4 Using Fragment-Based Lead Identification and Optimization.

Terry D. Crawford; Chudi Ndubaku; Huifen Chen; Jason Boggs; Brandon J. Bravo; Kelly DeLaTorre; Anthony M. Giannetti; Stephen E. Gould; Seth F. Harris; Steven Magnuson; Erin McNamara; Lesley J. Murray; Jim Nonomiya; Amy Sambrone; Stephen Schmidt; Tanya Smyczek; Mark S. Stanley; Philip Vitorino; Lan Wang; Kristina West; Ping Wu; Weilan Ye

Mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) is a serine/threonine kinase implicated in the regulation of many biological processes. A fragment-based lead discovery approach was used to generate potent and selective MAP4K4 inhibitors. The fragment hit pursued in this article had excellent ligand efficiency (LE), an important attribute for subsequent successful optimization into drug-like lead compounds. The optimization efforts eventually led us to focus on the pyridopyrimidine series, from which 6-(2-fluoropyridin-4-yl)pyrido[3,2-d]pyrimidin-4-amine (29) was identified. This compound had low nanomolar potency, excellent kinase selectivity, and good in vivo exposure, and demonstrated in vivo pharmacodynamic effects in a human tumor xenograft model.


Journal of Medicinal Chemistry | 2016

The Rational Design of Selective Benzoxazepin Inhibitors of the α-Isoform of Phosphoinositide 3-Kinase Culminating in the Identification of (S)-2-((2-(1-Isopropyl-1H-1,2,4-triazol-5-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)oxy)propanamide (GDC-0326)

Timothy P. Heffron; Robert Heald; Chudi Ndubaku; Binqing Wei; Martin Augistin; Steven Do; Kyle A. Edgar; Charles Eigenbrot; Lori Friedman; Emanuela Gancia; Philip Stephen Jackson; G. Jones; Aleksander Kolesnikov; Leslie Lee; John D. Lesnick; Cristina Lewis; Neville McLean; Mario Mörtl; Jim Nonomiya; Jodie Pang; Steve Price; Wei Wei Prior; Laurent Salphati; Steve Sideris; Steven Staben; Stefan Steinbacher; Vickie Tsui; Jeffrey Wallin; Deepak Sampath; Alan G. Olivero

Inhibitors of the class I phosphoinositide 3-kinase (PI3K) isoform PI3Kα have received substantial attention for their potential use in cancer therapy. Despite the particular attraction of targeting PI3Kα, achieving selectivity for the inhibition of this isoform has proved challenging. Herein we report the discovery of inhibitors of PI3Kα that have selectivity over the other class I isoforms and all other kinases tested. In GDC-0032 (3, taselisib), we previously minimized inhibition of PI3Kβ relative to the other class I insoforms. Subsequently, we extended our efforts to identify PI3Kα-specific inhibitors using PI3Kα crystal structures to inform the design of benzoxazepin inhibitors with selectivity for PI3Kα through interactions with a nonconserved residue. Several molecules selective for PI3Kα relative to the other class I isoforms, as well as other kinases, were identified. Optimization of properties related to drug metabolism then culminated in the identification of the clinical candidate GDC-0326 (4).


Future Medicinal Chemistry | 2009

Targeting inhibitor of apoptosis proteins for therapeutic intervention

Chudi Ndubaku; Fred E. Cohen; Eugene Varfolomeev; Domagoj Vucic

The inhibitors of apoptosis (IAP) proteins have emerged over the last decade as important targets for therapeutic intervention in human malignancies. Overexpression of IAPs has been implicated in cell survival and resistance against stress-induced apoptosis brought on by radiation and/or chemotherapeutics (currently the standard-of-care in a variety of different cancer diseases). In addition, evasion from death receptor-mediated apoptosis and regulation of NF-κB pathways and cell division have also been associated with IAP proteins. Efforts to target IAP proteins in tumors have focused mainly on designing small molecules that mimic the IAP-binding motif of the endogenous IAP antagonist, second mitochondrial activator of caspases. In addition, several other IAP-targeting strategies, including antisense oligonucleotides and transcriptional repression, have also been initiated, with the hope of providing therapeutic benefit to cancer patients.


ACS Medicinal Chemistry Letters | 2015

Design of Selective PAK1 Inhibitor G-5555: Improving Properties by Employing an Unorthodox Low-pK a Polar Moiety.

Chudi Ndubaku; James J. Crawford; Joy Drobnick; Ignacio Aliagas; D Campbell; Ping Dong; Laura M. Dornan; S Duron; Jennifer Epler; Lewis J. Gazzard; Christopher E. Heise; Klaus P. Hoeflich; Diana Jakubiak; Hank La; Wendy Lee; B Lin; J.P Lyssikatos; J Maksimoska; R Marmorstein; Lesley J. Murray; T O'Brien; Angela Oh; Sreemathy Ramaswamy; Weiru Wang; Xianrui Zhao; Yu Zhong; Elizabeth Blackwood; Joachim Rudolph

Signaling pathways intersecting with the p21-activated kinases (PAKs) play important roles in tumorigenesis and cancer progression. By recognizing that the limitations of FRAX1036 (1) were chiefly associated with the highly basic amine it contained, we devised a mitigation strategy to address several issues such as hERG activity. The 5-amino-1,3-dioxanyl moiety was identified as an effective means of reducing pK a and logP simultaneously. When positioned properly within the scaffold, this group conferred several benefits including potency, pharmacokinetics, and selectivity. Mouse xenograft PK/PD studies were carried out using an advanced compound, G-5555 (12), derived from this approach. These studies concluded that dose-dependent pathway modulation was achievable and paves the way for further in vivo investigations of PAK1 function in cancer and other diseases.

Collaboration


Dive into the Chudi Ndubaku'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
Researchain Logo
Decentralizing Knowledge