Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Bioorg Med Chem ; 44: 116275, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34314938

ABSTRACT

Bruton's tyrosine kinase (BTK) is an essential node on the BCR signaling in B cells, which are clinically validated to play a critical role in B-cell lymphomas and various auto-immune diseases such as Multiple Sclerosis (MS), Pemphigus, and rheumatoid arthritis (RA). Although non-selective irreversible BTK inhibitors have been approved for oncology, due to the emergence of drug resistance in B-cell lymphoma associated with covalent inhibitor, there an unmet medical need to identify reversible, selective, potent BTK inhibitor as viable therapeutics for patients. Herein, we describe the identification of Hits and subsequence optimization to improve the physicochemical properties, potency and kinome selectivity leading to the discovery of a novel class of BTK inhibitors. Utilizing Met ID and structure base design inhibitors were synthesized with increased in vivo metabolic stability and oral exposure in rodents suitable for advancing to lead optimization.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Drug Discovery , Protein Kinase Inhibitors/pharmacokinetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Structure-Activity Relationship
2.
Bioorg Med Chem ; 27(13): 2905-2913, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31138459

ABSTRACT

Since the approval of ibrutinib for the treatment of B-cell malignancies in 2012, numerous clinical trials have been reported using covalent inhibitors to target Bruton's tyrosine kinase (BTK) for oncology indications. However, a formidable challenge for the pharmaceutical industry has been the identification of reversible, selective, potent molecules for inhibition of BTK. Herein, we report application of Tethering-fragment-based screens to identify low molecular weight fragments which were further optimized to improve on-target potency and ADME properties leading to the discovery of reversible, selective, potent BTK inhibitors suitable for pre-clinical proof-of-concept studies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Humans , Protein Kinase Inhibitors/pharmacology
3.
Molecules ; 21(7)2016 Jul 16.
Article in English | MEDLINE | ID: mdl-27438812

ABSTRACT

Pacific, which is derived from the Latin pac, means peaceful. [...].


Subject(s)
Drug Discovery/methods , Humans , Quantitative Structure-Activity Relationship
4.
Circulation ; 128(8): 845-53, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23825361

ABSTRACT

BACKGROUND: Proton pump inhibitors (PPIs) are gastric acid-suppressing agents widely prescribed for the treatment of gastroesophageal reflux disease. Recently, several studies in patients with acute coronary syndrome have raised the concern that use of PPIs in these patients may increase their risk of major adverse cardiovascular events. The mechanism of this possible adverse effect is not known. Whether the general population might also be at risk has not been addressed. METHODS AND RESULTS: Plasma asymmetrical dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthase. Elevated plasma ADMA is associated with increased risk for cardiovascular disease, likely because of its attenuation of the vasoprotective effects of endothelial nitric oxide synthase. We find that PPIs elevate plasma ADMA levels and reduce nitric oxide levels and endothelium-dependent vasodilation in a murine model and ex vivo human tissues. PPIs increase ADMA because they bind to and inhibit dimethylarginine dimethylaminohydrolase, the enzyme that degrades ADMA. CONCLUSIONS: We present a plausible biological mechanism to explain the association of PPIs with increased major adverse cardiovascular events in patients with unstable coronary syndromes. Of concern, this adverse mechanism is also likely to extend to the general population using PPIs. This finding compels additional clinical investigations and pharmacovigilance directed toward understanding the cardiovascular risk associated with the use of the PPIs in the general population.


Subject(s)
Arginine/analogs & derivatives , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/metabolism , Gastroesophageal Reflux/drug therapy , Proton Pump Inhibitors/adverse effects , Proton Pump Inhibitors/therapeutic use , Amidohydrolases/metabolism , Animals , Arginine/blood , Biomarkers/blood , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Esomeprazole/adverse effects , Esomeprazole/pharmacology , Esomeprazole/therapeutic use , Humans , Lansoprazole/adverse effects , Lansoprazole/pharmacology , Lansoprazole/therapeutic use , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Proton Pump Inhibitors/pharmacology , Risk Factors , Vasodilation/drug effects
5.
J Pharmacol Exp Ther ; 348(1): 69-76, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24135074

ABSTRACT

PD 404182 [6H-6-imino-(2,3,4,5-tetrahydropyrimido)[1,2-c]-[1,3]benzothiazine], a heterocyclic iminobenzothiazine derivative, is a member of the Library of Pharmacologically Active Compounds (LOPAC) that is reported to possess antimicrobial and anti-inflammatory properties. In this study, we used biochemical assays to screen LOPAC against human dimethylarginine dimethylaminohydrolase isoform 1 (DDAH1), an enzyme that physiologically metabolizes asymmetric dimethylarginine (ADMA), an endogenous and competitive inhibitor of nitric oxide (NO) synthase. We discovered that PD 404182 directly and dose-dependently inhibits DDAH. Moreover, PD 404182 significantly increased intracellular levels of ADMA in cultured primary human vascular endothelial cells (ECs) and reduced lipopolysaccharide-induced NO production in these cells, suggesting its therapeutic potential in septic shock-induced vascular collapse. In addition, PD 404182 abrogated the formation of tube-like structures by ECs in an in vitro angiogenesis assay, indicating its antiangiogenic potential in diseases characterized by pathologically excessive angiogenesis. Furthermore, we investigated the potential mechanism of inhibition of DDAH by this small molecule and found that PD 404182, which has striking structural similarity to ADMA, could be competed by a DDAH substrate, suggesting that it is a competitive inhibitor. Finally, our enzyme kinetics assay showed time-dependent inhibition, and our inhibitor dilution assay showed that the enzymatic activity of DDAH did not recover significantly after dilution, suggesting that PD 404182 might be a tightly bound, covalent, or an irreversible inhibitor of human DDAH1. This proposal is supported by mass spectrometry studies with PD 404182 and glutathione.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Endothelium, Vascular/metabolism , Imines/chemistry , Imines/pharmacology , Neovascularization, Pathologic/drug therapy , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Thiazines/pharmacology , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Female , Humans , Hydroxamic Acids , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Thiazines/chemistry , Vorinostat
6.
J Med Chem ; 67(4): 2287-2304, 2024 02 22.
Article in English | MEDLINE | ID: mdl-38289623

ABSTRACT

This Perspective is the eighth in an annual series that summarizes successful fragment-to-lead (F2L) case studies published each year. A tabulated summary of relevant articles published in 2022 is provided, and features such as target class, screening methods, and ligand efficiency are discussed both for the 2022 examples and for the combined examples over the years 2015-2022. In addition, trends and new developments in the field are summarized. In 2022, 18 publications described successful fragment-to-lead studies, including the development of three clinical compounds (MTRX1719, MK-8189, and BI-823911).


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery , Pyrimidines , Sulfur Compounds , Drug Discovery/methods , Publications , Ligands
7.
Bioorg Med Chem Lett ; 23(10): 2844-52, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23562240

ABSTRACT

In the past 15 years, fragment-based lead discovery (FBLD) has been adopted widely throughout academia and industry. The approach entails discovering very small molecular fragments and growing, merging, or linking them to produce drug leads. Because the affinities of the initial fragments are often low, detection methods are pushed to their limits, leading to a variety of artifacts, false positives, and false negatives that too often go unrecognized. This Digest discusses some of these problems and offers suggestions to avoid them. Although the primary focus is on FBLD, many of the lessons also apply to more established approaches such as high-throughput screening.


Subject(s)
Drug Discovery , High-Throughput Screening Assays , Learning , Small Molecule Libraries/chemistry , High-Throughput Screening Assays/methods , Molecular Structure
8.
J Med Chem ; 66(2): 1137-1156, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36622056

ABSTRACT

This Perspective is the seventh in an annual series that summarizes successful Fragment-to-Lead (F2L) case studies published in a given year. A tabulated summary of relevant articles published in 2021 is provided, and features such as target class, screening methods, and ligand efficiency are discussed, both for the 2021 examples and for the combined examples over the years 2015-2021. In addition, trends and new developments in the field are summarized. In particular, the use of structural information in fragment-based drug discovery is discussed.


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery , Drug Discovery/methods , Publications , Ligands , Drug Design
9.
Top Curr Chem ; 317: 1-32, 2012.
Article in English | MEDLINE | ID: mdl-21695633

ABSTRACT

Fragment-based drug discovery (FBDD) has emerged in the past decade as a powerful tool for discovering drug leads. The approach first identifies starting points: very small molecules (fragments) that are about half the size of typical drugs. These fragments are then expanded or linked together to generate drug leads. Although the origins of the technique date back some 30 years, it was only in the mid-1990s that experimental techniques became sufficiently sensitive and rapid for the concept to be become practical. Since that time, the field has exploded: FBDD has played a role in discovery of at least 18 drugs that have entered the clinic, and practitioners of FBDD can be found throughout the world in both academia and industry. Literally dozens of reviews have been published on various aspects of FBDD or on the field as a whole, as have three books (Jahnke and Erlanson, Fragment-based approaches in drug discovery, 2006; Zartler and Shapiro, Fragment-based drug discovery: a practical approach, 2008; Kuo, Fragment based drug design: tools, practical approaches, and examples, 2011). However, this chapter will assume that the reader is approaching the field with little prior knowledge. It will introduce some of the key concepts, set the stage for the chapters to follow, and demonstrate how X-ray crystallography plays a central role in fragment identification and advancement.


Subject(s)
Drug Discovery , Small Molecule Libraries/chemistry , Computational Biology , Crystallography, X-Ray , High-Throughput Screening Assays , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Surface Plasmon Resonance
10.
RSC Med Chem ; 13(12): 1439, 2022 Dec 14.
Article in English | MEDLINE | ID: mdl-36561073

ABSTRACT

Guest editors David Rees, Anna Hirsch and Daniel Erlanson introduce the RSC Medicinal Chemistry themed collection on fragment-based drug discovery.

11.
J Med Chem ; 65(1): 84-99, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34928151

ABSTRACT

Fragment-based drug discovery (FBDD) continues to evolve and make an impact in the pharmaceutical sciences. We summarize successful fragment-to-lead studies that were published in 2020. Having systematically analyzed annual scientific outputs since 2015, we discuss trends and best practices in terms of fragment libraries, target proteins, screening technologies, hit-optimization strategies, and the properties of hit fragments and the leads resulting from them. As well as the tabulated Fragment-to-Lead (F2L) programs, our 2020 literature review identifies several trends and innovations that promise to further increase the success of FBDD. These include developing structurally novel screening fragments, improving fragment-screening technologies, using new computer-aided design and virtual screening approaches, and combining FBDD with other innovative drug-discovery technologies.


Subject(s)
Chemistry, Pharmaceutical/trends , Drug Design , Drug Discovery/trends , Publications/trends , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Humans
12.
Bioorg Med Chem Lett ; 21(10): 3078-83, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21459573

ABSTRACT

We report the use of a fragment-based lead discovery method, Tethering with extenders, to discover a pyridinone fragment that binds in an adaptive site of the protein PDK1. With subsequent medicinal chemistry, this led to the discovery of a potent and highly selective inhibitor of PDK1, which binds in the 'DFG-out' conformation.


Subject(s)
Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , Inhibitory Concentration 50 , Models, Biological , Molecular Structure , Pyridones/chemistry , Pyridones/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
13.
Curr Opin Chem Biol ; 62: 101-108, 2021 06.
Article in English | MEDLINE | ID: mdl-33838397

ABSTRACT

The protein KRAS has for decades been considered a holy grail of cancer drug discovery. For most of that time, it has also been considered undruggable. Since 2018, five compounds have entered the clinic targeting a single mutant form of KRAS, G12C. Here, we review each of these compounds along with additional approaches to targeting this and other mutants. Remaining challenges include expanding the identification of inhibitors to a broader range of known mutants and to conformations of the protein more likely to avoid development of resistance.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Mutant Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Acetonitriles/chemistry , Acetonitriles/pharmacology , Animals , Antineoplastic Agents/pharmacology , Drug Design , Drug Resistance, Neoplasm , Enzyme Inhibitors/metabolism , Humans , Mutant Proteins/metabolism , Mutation/genetics , Piperazines/chemistry , Piperazines/pharmacology , Precision Medicine , Protein Binding , Protein Conformation , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Structure-Activity Relationship
15.
J Med Chem ; 63(24): 15494-15507, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33226222

ABSTRACT

Fragment-based drug discovery (FBDD) has grown and matured to a point where it is valuable to keep track of its extent and details of application. This Perspective summarizes successful fragment-to-lead stories published in 2019. It is the fifth in a series that started with literature published in 2015. The analysis of screening methods, optimization strategies, and molecular properties of hits and leads are presented in the hope of informing best practices for FBDD. Moreover, FBDD is constantly evolving, and the latest technologies and emerging trends are summarized. These include covalent FBDD, FBDD for the stabilization of proteins or protein-protein interactions, FBDD for enzyme activators, new screening technologies, and advances in library design and chemical synthesis.


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery , Publications , Chemistry, Pharmaceutical/trends , Humans , Protein Interaction Domains and Motifs , Protein Stability , Proteins/chemistry , Proteins/metabolism
16.
J Med Chem ; 63(9): 4430-4444, 2020 05 14.
Article in English | MEDLINE | ID: mdl-31913033

ABSTRACT

This Perspective, the fourth in an annual series, summarizes fragment-to-lead (F2L) success stories published during 2018. Topics such as target class, screening methods, physicochemical properties, and ligand efficiency are discussed for the 2018 examples as well as for the combined 111 F2L examples covering 2015-2018. While the overall properties of fragments and leads have remained constant, a number of new trends are noted, for example, broadening of target class coverage and application of FBDD to covalent inhibitors. Moreover, several studies make use of fragment hits that were previously described in the literature, illustrating that fragments are versatile starting points that can be optimized to structurally diverse leads. By focusing on success stories, the hope is that this Perspective will identify and inform best practices in fragment-based drug discovery.


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery/methods , Chemistry, Pharmaceutical/trends , Drug Discovery/trends , Drug Evaluation, Preclinical/methods , Publications
17.
Nat Struct Mol Biol ; 11(8): 730-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258570

ABSTRACT

Obesity and type II diabetes are closely linked metabolic syndromes that afflict >100 million people worldwide. Although protein tyrosine phosphatase 1B (PTP1B) has emerged as a promising target for the treatment of both syndromes, the discovery of pharmaceutically acceptable inhibitors that bind at the active site remains a substantial challenge. Here we describe the discovery of an allosteric site in PTP1B. Crystal structures of PTP1B in complex with allosteric inhibitors reveal a novel site located approximately 20 A from the catalytic site. We show that allosteric inhibitors prevent formation of the active form of the enzyme by blocking mobility of the catalytic loop, thereby exploiting a general mechanism used by tyrosine phosphatases. Notably, these inhibitors exhibit selectivity for PTP1B and enhance insulin signaling in cells. Allosteric inhibition is a promising strategy for targeting PTP1B and constitutes a mechanism that may be applicable to other tyrosine phosphatases.


Subject(s)
Protein Tyrosine Phosphatases/chemistry , Allosteric Site , Animals , Binding Sites , Binding, Competitive , CHO Cells , Catalysis , Catalytic Domain , Cloning, Molecular , Cricetinae , Crystallography, X-Ray , DNA/chemistry , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Kinetics , Ligands , Models, Chemical , Models, Molecular , Obesity , Phosphoric Monoester Hydrolases/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Time Factors , Transfection , Tyrosine/chemistry
18.
Cell Chem Biol ; 26(1): 9-15, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30482678

ABSTRACT

Fragment-based drug discovery typically requires an interplay between screening methods, structural methods, and medicinal chemistry. X-ray crystallography is generally the method of choice to obtain three-dimensional structures of the bound ligand/protein complex, but this can sometimes be difficult, particularly for early, low-affinity fragment hits. In this Perspective, we discuss strategies to advance and evolve fragments in the absence of crystal structures of protein-fragment complexes, although the structure of the unliganded protein may be available. The strategies can involve other structural techniques, such as NMR spectroscopy, molecular modeling, or a variety of chemical approaches. Often, these strategies are aimed at guiding evolution of initial fragment hits to a stage where crystal structures can be obtained for further structure-based optimization.


Subject(s)
Drug Discovery , Pharmaceutical Preparations/chemistry , Animals , Crystallography, X-Ray , Humans , Models, Molecular , Proteins/chemistry
19.
J Med Chem ; 62(8): 3857-3872, 2019 04 25.
Article in English | MEDLINE | ID: mdl-30462504

ABSTRACT

This Miniperspective is the third in a series reviewing fragment-to-lead publications from a given year. Following our reviews for 2015 and 2016, this Miniperspective provides tabulated summaries of relevant articles published in 2017 along with some general observations. In addition, we discuss insights obtained from analysis of the combined data set of 85 examples from all three years of publications.


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery/methods , Chemistry, Pharmaceutical/trends , Drug Discovery/trends , Drug Evaluation, Preclinical/methods
20.
ACS Med Chem Lett ; 10(9): 1302-1308, 2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31531201

ABSTRACT

KRAS regulates many cellular processes including proliferation, survival, and differentiation. Point mutants of KRAS have long been known to be molecular drivers of cancer. KRAS p.G12C, which occurs in approximately 14% of lung adenocarcinomas, 3-5% of colorectal cancers, and low levels in other solid tumors, represents an attractive therapeutic target for covalent inhibitors. Herein, we disclose the discovery of a class of novel, potent, and selective covalent inhibitors of KRASG12C identified through a custom library synthesis and screening platform called Chemotype Evolution and structure-based design. Identification of a hidden surface groove bordered by H95/Y96/Q99 side chains was key to the optimization of this class of molecules. Best-in-series exemplars exhibit a rapid covalent reaction with cysteine 12 of GDP-KRASG12C with submicromolar inhibition of downstream signaling in a KRASG12C-specific manner.

SELECTION OF CITATIONS
SEARCH DETAIL