Network


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

Hotspot


Dive into the research topics where Stefan O. Ochiana is active.

Publication


Featured researches published by Stefan O. Ochiana.


Journal of Medicinal Chemistry | 2013

Experimental and structural testing module to analyze paralogue-specificity and affinity in the Hsp90 inhibitors series.

Tony Taldone; Pallav D. Patel; Maulik R. Patel; Hardik J. Patel; Christopher E. Evans; Anna Rodina; Stefan O. Ochiana; Smit K. Shah; Mohammad Uddin; Daniel T. Gewirth; Gabriela Chiosis

We here describe the first reported comprehensive analysis of Hsp90 paralogue affinity and selectivity in the clinical Hsp90 inhibitor chemotypes. This has been possible through the development of a versatile experimental assay based on a new FP-probe (16a) that we both describe here. The assay can test rapidly and accurately the binding affinity of all major Hsp90 chemotypes and has a testing range that spans low nanomolar to millimolar binding affinities. We couple this assay with a computational analysis that allows for rationalization of paralogue selectivity and defines not only the major binding modes that relay pan-paralogue binding or, conversely, paralogue selectivity, but also identifies molecular characteristics that impart such features. The methods developed here provide a blueprint for parsing out the contribution of the four Hsp90 paralogues to the perceived biological activity with the current Hsp90 chemotypes and set the ground for the development of paralogue selective inhibitors.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Ligand deconstruction: Why some fragment binding positions are conserved and others are not

Dima Kozakov; David R. Hall; Stefan Jehle; Lingqi Luo; Stefan O. Ochiana; Elizabeth V. Jones; Karen N. Allen; Adrian Whitty; Sandor Vajda

Significance Fragment-based drug discovery (FBDD), in which initial screening is done with low-molecular-weight compounds called fragments, relies on the premise that the fragment binding mode will be conserved on subsequent expansion to a larger ligand. We describe a remarkably simple condition for fragment binding conservation that can be tested computationally. The condition can be used for detecting whether a protein is suitable for FBDD, for predicting the size of fragments required for screening, and for determining if a fragment hit can be extended into a higher affinity ligand. The findings also reveal general properties of binding sites, highlighting the role that critical interactions between anchor sites and anchor fragments play in protein–ligand interactions in general. Fragment-based drug discovery (FBDD) relies on the premise that the fragment binding mode will be conserved on subsequent expansion to a larger ligand. However, no general condition has been established to explain when fragment binding modes will be conserved. We show that a remarkably simple condition can be developed in terms of how fragments coincide with binding energy hot spots—regions of the protein where interactions with a ligand contribute substantial binding free energy—the locations of which can easily be determined computationally. Because a substantial fraction of the free energy of ligand binding comes from interacting with the residues in the energetically most important hot spot, a ligand moiety that sufficiently overlaps with this region will retain its location even when other parts of the ligand are removed. This hypothesis is supported by eight case studies. The condition helps identify whether a protein is suitable for FBDD, predicts the size of fragments required for screening, and determines whether a fragment hit can be extended into a higher affinity ligand. Our results show that ligand binding sites can usefully be thought of in terms of an anchor site, which is the top-ranked hot spot and dominates the free energy of binding, surrounded by a number of weaker satellite sites that confer improved affinity and selectivity for a particular ligand and that it is the intrinsic binding potential of the protein surface that determines whether it can serve as a robust binding site for a suitably optimized ligand.


European Journal of Medicinal Chemistry | 2013

The human Aurora kinase inhibitor danusertib is a lead compound for anti-trypanosomal drug discovery via target repurposing

Stefan O. Ochiana; Vidya Pandarinath; Zhouxi Wang; Rishika Kapoor; Mary Jo Ondrechen; Larry Ruben

New drugs for neglected tropical diseases such as human African trypanosomiasis (HAT) are needed, yet drug discovery efforts are not often focused on this area due to cost. Target repurposing, achieved by the matching of essential parasite enzymes to those human enzymes that have been successfully inhibited by small molecule drugs, provides an attractive means by which new drug optimization programs can be pragmatically initiated. In this report we describe our results in repurposing an established class of human Aurora kinase inhibitors, typified by danusertib (1), which we have observed to be an inhibitor of trypanosomal Aurora kinase 1 (TbAUK1) and effective in parasite killing in vitro. Informed by homology modeling and docking, a series of analogs of 1 were prepared that explored the scope of the chemotype and provided a nearly 25-fold improvement in cellular selectivity for parasite cells over human cells.


Journal of Medicinal Chemistry | 2011

Pharmacological Validation of Trypanosoma brucei Phosphodiesterases B1 and B2 as Druggable Targets for African Sleeping Sickness

Nicholas D. Bland; Cuihua Wang; Craig Tallman; Alden Gustafson; Zhouxi Wang; Trent D. Ashton; Stefan O. Ochiana; Gregory McAllister; Kristina Cotter; Anna P. Fang; Lara Gechijian; Norman Garceau; Rajiv Gangurde; Ron Ortenberg; Mary Jo Ondrechen; Robert K. Campbell

Neglected tropical disease drug discovery requires application of pragmatic and efficient methods for development of new therapeutic agents. In this report, we describe our target repurposing efforts for the essential phosphodiesterase (PDE) enzymes TbrPDEB1 and TbrPDEB2 of Trypanosoma brucei , the causative agent for human African trypanosomiasis (HAT). We describe protein expression and purification, assay development, and benchmark screening of a collection of 20 established human PDE inhibitors. We disclose that the human PDE4 inhibitor piclamilast, and some of its analogues, show modest inhibition of TbrPDEB1 and B2 and quickly kill the bloodstream form of the subspecies T. brucei brucei . We also report the development of a homology model of TbrPDEB1 that is useful for understanding the compound-enzyme interactions and for comparing the parasitic and human enzymes. Our profiling and early medicinal chemistry results strongly suggest that human PDE4 chemotypes represent a better starting point for optimization of TbrPDEB inhibitors than those that target any other human PDEs.


Bioorganic & Medicinal Chemistry Letters | 2012

Synthesis and evaluation of human phosphodiesterases (PDE) 5 inhibitor analogs as trypanosomal PDE inhibitors. Part 1. Sildenafil analogs

Cuihua Wang; Trent D. Ashton; Alden Gustafson; Nicholas D. Bland; Stefan O. Ochiana; Robert K. Campbell

Parasitic diseases, such as African sleeping sickness, have a significant impact on the health and well-being in the poorest regions of the world. Pragmatic drug discovery efforts are needed to find new therapeutic agents. In this Letter we describe target repurposing efforts focused on trypanosomal phosphodiesterases. We outline the synthesis and biological evaluation of analogs of sildenafil (1), a human PDE5 inhibitor, for activities against trypanosomal PDEB1 (TbrPDEB1). We find that, while low potency analogs can be prepared, this chemical class is a sub-optimal starting point for further development of TbrPDE inhibitors.


Journal of Medicinal Chemistry | 2015

Structure–Activity Relationship in a Purine-Scaffold Compound Series with Selectivity for the Endoplasmic Reticulum Hsp90 Paralog Grp94

Hardik J. Patel; Pallav D. Patel; Stefan O. Ochiana; Pengrong Yan; Weilin Sun; Maulik R. Patel; Smit K. Shah; Elisa Tramentozzi; James C. Brooks; Alexander Bolaender; Liza Shrestha; Ralph Stephani; Paola Finotti; Cynthia A. Leifer; Zihai Li; Daniel T. Gewirth; Tony Taldone; Gabriela Chiosis

Grp94 is involved in the regulation of a restricted number of proteins and represents a potential target in a host of diseases, including cancer, septic shock, autoimmune diseases, chronic inflammatory conditions, diabetes, coronary thrombosis, and stroke. We have recently identified a novel allosteric pocket located in the Grp94 N-terminal binding site that can be used to design ligands with a 2-log selectivity over the other Hsp90 paralogs. Here we perform extensive SAR investigations in this ligand series and rationalize the affinity and paralog selectivity of choice derivatives by molecular modeling. We then use this to design 18c, a derivative with good potency for Grp94 (IC50 = 0.22 μM) and selectivity over other paralogs (>100- and 33-fold for Hsp90α/β and Trap-1, respectively). The paralog selectivity and target-mediated activity of 18c was confirmed in cells through several functional readouts. Compound 18c was also inert when tested against a large panel of kinases. We show that 18c has biological activity in several cellular models of inflammation and cancer and also present here for the first time the in vivo profile of a Grp94 inhibitor.


Blood | 2015

Tumor-specific HSP90 inhibition as a therapeutic approach in JAK-mutant acute lymphoblastic leukemias.

Nicole Kucine; Sachie Marubayashi; Neha Bhagwat; Efthymia Papalexi; Priya Koppikar; Marta Sanchez Martin; Lauren Dong; Marty S. Tallman; Elisabeth Paietta; Kai Wang; Jie He; Doron Lipson; Phil Stephens; V.A. Miller; Jacob M. Rowe; Julie Teruya-Feldstein; Charles G. Mullighan; Adolfo A. Ferrando; Andrei V. Krivtsov; Scott A. Armstrong; Laura Leung; Stefan O. Ochiana; Gabriela Chiosis; Ross L. Levine; Maria Kleppe

The development of the dual Janus kinase 1/2 (JAK1/2) inhibitor ruxolitinib for the treatment of myeloproliferative neoplasms (MPNs) has led to studies of ruxolitinib in other clinical contexts, including JAK-mutated acute lymphoblastic leukemia (ALL). However, the limited ability of JAK inhibition to induce molecular or clinicopathological responses in MPNs suggests a need for development of better therapies for JAK kinase-dependent malignancies. Here, we demonstrate that heat shock protein 90 (HSP90) inhibition using a purine-scaffold HSP90 inhibitor in early clinical development is an effective therapeutic approach in JAK-dependent ALL and can overcome persistence to JAK-inhibitor therapy in ALL cells.


ACS Chemical Biology | 2014

Affinity Purification Probes of Potential Use To Investigate the Endogenous Hsp70 Interactome in Cancer

Anna Rodina; Tony Taldone; Yanlong Kang; Pallav D. Patel; John Koren; Pengrong Yan; Erica DaGama Gomes; Chenghua Yang; Maulik R. Patel; Liza Shrestha; Stefan O. Ochiana; Cristina Santarossa; Ronnie Maharaj; Alexander Gozman; Marc B. Cox; Hediye Erdjument-Bromage; Ronald C. Hendrickson; Leandro Cerchietti; Ari Melnick; Monica L. Guzman; Gabriela Chiosis

Heat shock protein 70 (Hsp70) is a family of proteins with key roles in regulating malignancy. Cancer cells rely on Hsp70 to inhibit apoptosis, regulate senescence and autophagy, and maintain the stability of numerous onco-proteins. Despite these important biological functions in cancer, robust chemical tools that enable the analysis of the Hsp70-regulated proteome in a tumor-by-tumor manner are yet unavailable. Here we take advantage of a recently reported Hsp70 ligand to design and develop an affinity purification chemical toolset for potential use in the investigation of the endogenous Hsp70-interacting proteome in cancer. We demonstrate that these tools lock Hsp70 in complex with onco-client proteins and effectively isolate Hsp70 complexes for identification through biochemical techniques. Using these tools we provide proof-of-concept analyses that glimpse into the complex roles played by Hsp70 in maintaining a multitude of cell-specific malignancy-driving proteins.


Journal of Labelled Compounds and Radiopharmaceuticals | 2016

Radiosynthesis of the iodine-124 labeled Hsp90 inhibitor PU-H71

Tony Taldone; Danuta Zatorska; Stefan O. Ochiana; Peter Smith-Jones; Jacek Koziorowski; Mark Dunphy; Pat Zanzonico; Alexander Bolaender; Jason S. Lewis; Steven M. Larson; Gabriela Chiosis; Naga Vara Kishore Pillarsetty

Heat shock protein 90 (Hsp90) is an ATP dependent molecular chaperone protein whose function is critical for maintaining several key proteins involved in survival and proliferation of cancer cells. PU-H71 (1), is a potent purine-scaffold based ATP pocket binding Hsp90 inhibitor which has been shown to have potent activity in a broad range of in vivo cancer models and is currently in Phase I clinical trials in patients with advanced solid malignancies, lymphomas, and myeloproliferative neoplasms. In this report, we describe the radiosynthesis of [(124)I]-PU-H71(5); this was synthesized from the corresponding Boc-protected stannane precursor 3 by iododestannylation with [(124)I]-NaI using chloramine-T as an oxidant for 2 min, followed by Boc deprotection with 6 N HCl at 50 °C for 30 min to yield the final compound. The final product 5 was purified using HPLC and was isolated with an overall yield of 55 ± 6% (n = 6, isolated) from 3, and >98% purity and an average specific activity of 980 mCi/µmol. Our report sets the stage for the introduction of [(124)I]-PU-H71 as a potential non-invasive probe for understanding biodistribution and pharmacokinetics of PU-H71 in living subjects using positron emission tomography imaging.


Chemical Biology & Drug Design | 2015

Repurposing Human PDE4 Inhibitors for Neglected Tropical Diseases. Evaluation of Analogs of the Human PDE4 Inhibitor GSK‐256066 as Inhibitors of PDEB1 of Trypanosoma brucei

Stefan O. Ochiana; Nicholas D. Bland; Luca Settimo; Robert K. Campbell

Cyclic nucleotide phosphodiesterases (PDEs) have been identified as important enzyme targets for drug development in both humans and Trypanosoma brucei, the causative agent of human African trypanosomiasis. With this in mind, we recently reported the profiling of a range of human phosphodiesterase inhibitors, showing that human PDE4 inhibitors tend to display the best potency against the trypanosomal phosphodiesterase TbrPDEB1. Among these was GSK‐256066, a potent inhibitor of human PDE4 and a weak inhibitor of TbrPDEB1. In this report, we describe the results of a structure–activity relationship study of this chemotype, leading to the discovery of analogs with improved potency against TbrPDEB1 and micromolar inhibition of T. brucei cellular growth. We rationalize the potency trends via molecular docking of the new inhibitors into a recently reported apo structure of TbrPDEB1. The studies in this article will inform future efforts in repurposing human PDE inhibitors as antitrypanosomal agents.

Collaboration


Dive into the Stefan O. Ochiana's collaboration.

Top Co-Authors

Avatar

Gabriela Chiosis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tony Taldone

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pallav D. Patel

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hardik J. Patel

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Maulik R. Patel

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pengrong Yan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anna Rodina

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Liza Shrestha

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nicholas D. Bland

Marine Biological Laboratory

View shared research outputs
Top Co-Authors

Avatar

Robert K. Campbell

Marine Biological Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge