Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Nat Commun ; 12(1): 4669, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344873

ABSTRACT

Diseases are a manifestation of how thousands of proteins interact. In several diseases, such as cancer and Alzheimer's disease, proteome-wide disturbances in protein-protein interactions are caused by alterations to chaperome scaffolds termed epichaperomes. Epichaperome-directed chemical probes may be useful for detecting and reversing defective chaperomes. Here we provide structural, biochemical, and functional insights into the discovery of epichaperome probes, with a focus on their use in central nervous system diseases. We demonstrate on-target activity and kinetic selectivity of a radiolabeled epichaperome probe in both cells and mice, together with a proof-of-principle in human patients in an exploratory single group assignment diagnostic study (ClinicalTrials.gov Identifier: NCT03371420). The clinical study is designed to determine the pharmacokinetic parameters and the incidence of adverse events in patients receiving a single microdose of the radiolabeled probe administered by intravenous injection. In sum, we introduce a discovery platform for brain-directed chemical probes that specifically modulate epichaperomes and provide proof-of-principle applications in their use in the detection, quantification, and modulation of the target in complex biological systems.


Subject(s)
Central Nervous System/metabolism , Molecular Chaperones/metabolism , Protein Interaction Mapping/instrumentation , Proteome/metabolism , Animals , Biomarkers, Tumor/metabolism , Blood-Brain Barrier/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Survival/drug effects , Central Nervous System/drug effects , Glioblastoma/diagnosis , Glioblastoma/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Molecular Probes/chemistry , Molecular Probes/pharmacokinetics , Molecular Probes/pharmacology , Molecular Probes/therapeutic use , Positron-Emission Tomography
2.
Clin Cancer Res ; 26(19): 5178-5187, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32366671

ABSTRACT

PURPOSE: 124I-PU-H71 is an investigational first-in-class radiologic agent specific for imaging tumor epichaperome formations. The intracellular epichaperome forms under cellular stress and is a clinically validated oncotherapeutic target. We conducted a first-in-human study of microdose 124I-PU-H71 for PET to study in vivo biodistribution, pharmacokinetics, metabolism, and safety; and the feasibility of epichaperome-targeted tumor imaging. EXPERIMENTAL DESIGN: Adult patients with cancer (n = 30) received 124I-PU-H71 tracer (201±12 MBq, <25 µg) intravenous bolus followed by PET/CT scans and blood radioassays. RESULTS: 124I-PU-H71 PET detected tumors of different cancer types (breast, lymphoma, neuroblastoma, genitourinary, gynecologic, sarcoma, and pancreas). 124I-PU-H71 was retained by tumors for several days while it cleared rapidly from bones, healthy soft tissues, and blood. Radiation dosimetry is favorable and patients suffered no adverse effects. CONCLUSIONS: Our first-in-human results demonstrate the safety and feasibility of noninvasive in vivo detection of tumor epichaperomes using 124I-PU-H71 PET, supporting clinical development of PU-H71 and other epichaperome-targeted therapeutics.


Subject(s)
Benzodioxoles/administration & dosage , HSP90 Heat-Shock Proteins/genetics , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Purines/administration & dosage , Adult , Aged , Benzodioxoles/adverse effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Iodine Radioisotopes/administration & dosage , Male , Middle Aged , Neoplasms/genetics , Neoplasms/pathology , Positron Emission Tomography Computed Tomography/methods , Purines/adverse effects , Tissue Distribution/radiation effects
3.
J Labelled Comp Radiopharm ; 59(3): 129-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26806023

ABSTRACT

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.


Subject(s)
Benzodioxoles/chemistry , Iodine Radioisotopes/chemistry , Purines/chemistry , Radiopharmaceuticals/chemical synthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors
4.
Cell Rep ; 13(10): 2159-73, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26628369

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous and fatal disease with an urgent need for improved therapeutic regimens given that most patients die from relapsed disease. Irrespective of mutation status, the development of aggressive leukemias is enabled by increasing dependence on signaling networks. We demonstrate that a hyperactive signalosome drives addiction of AML cells to a tumor-specific Hsp90 species (teHsp90). Through genetic, environmental, and pharmacologic perturbations, we demonstrate a direct and quantitative link between hyperactivated signaling pathways and apoptotic sensitivity of AML to teHsp90 inhibition. Specifically, we find that hyperactive JAK-STAT and PI3K-AKT signaling networks are maintained by teHsp90 and, in fact, gradual activation of these networks drives tumors increasingly dependent on teHsp90. Thus, although clinically aggressive AML survives via signalosome activation, this addiction creates a vulnerability that can be exploited with Hsp90-directed therapy.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Signal Transduction/physiology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Benzodioxoles/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Mice , Mice, Nude , Purines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Blood ; 126(22): 2479-83, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26443624

ABSTRACT

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.


Subject(s)
Benzodioxoles/pharmacology , HSP90 Heat-Shock Proteins , Janus Kinase 1 , Janus Kinase 2 , Neoplasm Proteins , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Purines/pharmacology , Animals , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mice , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Xenograft Model Antitumor Assays
6.
J Med Chem ; 58(9): 3922-43, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25901531

ABSTRACT

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/ß 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.


Subject(s)
Adenine/analogs & derivatives , Endoplasmic Reticulum/metabolism , HSP90 Heat-Shock Proteins/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Purines/chemistry , Adenine/chemistry , Adenine/pharmacokinetics , Adenine/pharmacology , Allosteric Site , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Differentiation , Cell Line , Cell Line, Tumor , Female , Insulin-Like Growth Factor II/metabolism , Ligands , Membrane Glycoproteins/metabolism , Mice, Nude , Molecular Docking Simulation , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Protein Binding , Purines/pharmacokinetics , Purines/pharmacology , Receptor, ErbB-2/metabolism , Structure-Activity Relationship , Tissue Distribution , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
7.
Proc Natl Acad Sci U S A ; 112(20): E2585-94, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25918377

ABSTRACT

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.


Subject(s)
Drug Discovery/methods , Ligands , Models, Biological , Peptide Fragments/metabolism , Binding Sites/genetics , Conserved Sequence/genetics , Peptide Fragments/genetics , Protein Binding
8.
Chem Biol Drug Des ; 85(5): 549-64, 2015 May.
Article in English | MEDLINE | ID: mdl-25283372

ABSTRACT

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.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Aminoquinolines/therapeutic use , Binding Sites , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Repositioning , Humans , Molecular Docking Simulation , Neglected Diseases/drug therapy , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Protein Structure, Tertiary , Protozoan Proteins/metabolism , Quinolines/chemistry , Quinolines/metabolism , Quinolines/pharmacology , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology , Sulfones/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy
9.
Trends Pharmacol Sci ; 35(11): 592-603, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25262919

ABSTRACT

Normal cellular function is maintained by coordinated proteome machinery that performs a vast array of activities. Helping the proteome in such roles is the chaperome, a network of molecular chaperones and folding enzymes. The stressed cell contains, at any time, a complex mixture of chaperome complexes; a majority performs 'housekeeping functions' similarly to non-stressed, normal cells, but a finely-tuned fraction buffers the proteome altered by chronic stress. The stress chaperome is epigenetically distinct from its normal, housekeeping counterpart, providing a basis for its selective targeting by small molecules. We discuss here the development of chaperome inhibitors, and how agents targeting chaperome members in stressed cells are in fact being directed towards chaperome complexes, and their effect is therefore determined by their ability to sample and engage such complexes. A new approach is needed to target and implement chaperome modulators in the investigation of diseases, and we propose that the classical thinking in drug discovery needs adjustment when developing chaperome-targeting drugs.


Subject(s)
Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/metabolism , Animals , Heat-Shock Proteins/chemistry , Humans , Models, Molecular , Molecular Chaperones/chemistry , Molecular Targeted Therapy
10.
ACS Chem Biol ; 9(8): 1698-705, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24934503

ABSTRACT

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.


Subject(s)
Affinity Labels , Chromatography, Affinity/methods , HSP70 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Cell Line, Tumor , HSP70 Heat-Shock Proteins/isolation & purification , Humans , Protein Binding
11.
Expert Opin Investig Drugs ; 23(5): 611-28, 2014 May.
Article in English | MEDLINE | ID: mdl-24669860

ABSTRACT

INTRODUCTION: Heat shock protein 90 (HSP90) serves as a critical facilitator for oncogene addiction. There has been augmenting enthusiasm in pursuing HSP90 as an anticancer strategy. In fact, since the initial serendipitous discovery that geldanamycin (GM) inhibits HSP90, the field has rapidly moved from proof-of-concept clinical studies with GM derivatives to novel second-generation inhibitors. AREAS COVERED: The authors highlight the current status of the second-generation HSP90 inhibitors in clinical development. Herein, the authors note the lessons learned from the completed clinical trials of first- and second-generation inhibitors and describe various assays attempting to serve for a more rational implementation of these agents to cancer treatment. Finally, the authors discuss the future perspectives for this promising class of agents. EXPERT OPINION: The knowledge gained thus far provides perhaps only a glimpse at the potential of HSP90 for which there is still much work to be done. Lessons from the clinical trials suggest that HSP90 therapy would advance at a faster pace if patient selection and tumor pharmacokinetics of these drugs were better understood and applied to their clinical development. It is also evident that combining HSP90 inhibitors with other potent anticancer therapies holds great promise not only due to synergistic antitumor activity but also due to the potential of prolonging or preventing the development of drug resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Forecasting , HSP90 Heat-Shock Proteins/metabolism , Humans , Neoplasms/metabolism , Treatment Outcome
12.
Bioorg Med Chem Lett ; 23(21): 5971-4, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24042005

ABSTRACT

Human African trypanosomiasis (HAT) is a parasitic neglected tropical disease that affects 10,000 patients each year. Current treatments are sub-optimal, and the disease is fatal if not treated. Herein, we report our continuing efforts to repurpose the human phosphodiesterase 4 (hPDE4) inhibitor piclamilast to target trypanosomal phosphodiesterase TbrPDEB1. We prepared a range of substituted heterocyclic replacements for the 4-amino-3,5-dichloro-pyridine headgroup of piclamilast, and found that these compounds exhibited weak inhibitory activity of TbrPDEB1.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Catechols/chemistry , Catechols/pharmacology , Protozoan Proteins/antagonists & inhibitors , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Benzamides/chemistry , Benzamides/pharmacology , Drug Discovery , Humans , Models, Molecular , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Protozoan Proteins/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology
13.
Eur J Med Chem ; 62: 777-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22889561

ABSTRACT

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.


Subject(s)
Benzamides/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma/drug effects , Aurora Kinases , Benzamides/chemical synthesis , Benzamides/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Trypanosomiasis/drug therapy
14.
Bioorg Med Chem Lett ; 22(7): 2582-4, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22377518

ABSTRACT

In this Letter we describe our ongoing target repurposing efforts focused on discovery of inhibitors of the essential trypanosomal phosphodiesterase TbrPDEB1. This enzyme has been implicated in virulence of Trypanosoma brucei, the causative agent of human African trypanosomiasis (HAT). We outline the synthesis and biological evaluation of analogs of tadalafil, a human PDE5 inhibitor currently utilized for treatment of erectile dysfunction, and report that these analogs are weak inhibitors of TbrPDEB1.


Subject(s)
Carbolines/chemical synthesis , Phosphodiesterase 5 Inhibitors/chemical synthesis , Phosphoric Diester Hydrolases/chemistry , Protozoan Proteins/antagonists & inhibitors , Trypanocidal Agents/chemical synthesis , Trypanosoma brucei brucei/enzymology , Animals , Biological Assay , Carbolines/chemistry , Humans , Phosphodiesterase 5 Inhibitors/chemistry , Protozoan Proteins/chemistry , Solutions , Structure-Activity Relationship , Tadalafil , Trypanocidal Agents/chemistry
15.
Bioorg Med Chem Lett ; 22(7): 2579-81, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22370268

ABSTRACT

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.


Subject(s)
Phosphodiesterase 5 Inhibitors/chemical synthesis , Phosphoric Diester Hydrolases/chemistry , Piperazines/chemical synthesis , Protozoan Proteins/antagonists & inhibitors , Sulfones/chemical synthesis , Trypanocidal Agents/chemical synthesis , Trypanosoma brucei brucei/enzymology , Animals , Biological Assay , Humans , Phosphodiesterase 5 Inhibitors/chemistry , Piperazines/chemistry , Protozoan Proteins/chemistry , Purines/chemical synthesis , Purines/chemistry , Sildenafil Citrate , Solutions , Structure-Activity Relationship , Sulfones/chemistry , Trypanocidal Agents/chemistry
16.
J Med Chem ; 54(23): 8188-94, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22023548

ABSTRACT

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.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Benzamides/chemical synthesis , Benzamides/chemistry , Benzamides/pharmacology , Catalytic Domain , Humans , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL