Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Am J Hum Genet ; 104(4): 651-664, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30929736

ABSTRACT

Pheochromocytomas and paragangliomas (PPGLs) provide some of the clearest genetic evidence for the critical role of metabolism in the tumorigenesis process. Approximately 40% of PPGLs are caused by driver germline mutations in 16 known susceptibility genes, and approximately half of these genes encode members of the tricarboxylic acid (TCA) cycle. Taking as a starting point the involvement of the TCA cycle in PPGL development, we aimed to identify unreported mutations that occurred in genes involved in this key metabolic pathway and that could explain the phenotypes of additional individuals who lack mutations in known susceptibility genes. To accomplish this, we applied a targeted sequencing of 37 TCA-cycle-related genes to DNA from 104 PPGL-affected individuals with no mutations in the major known predisposing genes. We also performed omics-based analyses, TCA-related metabolite determination, and 13C5-glutamate labeling assays. We identified five germline variants affecting DLST in eight unrelated individuals (∼7%); all except one were diagnosed with multiple PPGLs. A recurrent variant, c.1121G>A (p.Gly374Glu), found in four of the eight individuals triggered accumulation of 2-hydroxyglutarate, both in tumors and in a heterologous cell-based assay designed to functionally evaluate DLST variants. p.Gly374Glu-DLST tumors exhibited loss of heterozygosity, and their methylation and expression profiles are similar to those of EPAS1-mutated PPGLs; this similarity suggests a link between DLST disruption and pseudohypoxia. Moreover, we found positive DLST immunostaining exclusively in tumors carrying TCA-cycle or EPAS1 mutations. In summary, this study reveals DLST as a PPGL-susceptibility gene and further strengthens the relevance of the TCA cycle in PPGL development.


Subject(s)
Acyltransferases/genetics , Adrenal Gland Neoplasms/genetics , Germ-Line Mutation , Paraganglioma/genetics , Pheochromocytoma/genetics , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinogenesis , Catalytic Domain , Citric Acid Cycle , DNA Methylation , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Loss of Heterozygosity , Male , Middle Aged
3.
Nat Commun ; 10(1): 97, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626872

ABSTRACT

Squalene epoxidase (SQLE), also known as squalene monooxygenase, catalyzes the stereospecific conversion of squalene to 2,3(S)-oxidosqualene, a key step in cholesterol biosynthesis. SQLE inhibition is targeted for the treatment of hypercholesteremia, cancer, and fungal infections. However, lack of structure-function understanding has hindered further progression of its inhibitors. We have determined the first three-dimensional high-resolution crystal structures of human SQLE catalytic domain with small molecule inhibitors (2.3 Å and 2.5 Å). Comparison with its unliganded state (3.0 Å) reveals conformational rearrangements upon inhibitor binding, thus allowing deeper interpretation of known structure-activity relationships. We use the human SQLE structure to further understand the specificity of terbinafine, an approved agent targeting fungal SQLE, and to provide the structural insights into terbinafine-resistant mutants encountered in the clinic. Collectively, these findings elucidate the structural basis for the specificity of the epoxidation reaction catalyzed by SQLE and enable further rational development of next-generation inhibitors.


Subject(s)
Squalene Monooxygenase/chemistry , Squalene Monooxygenase/metabolism , Animals , Catalytic Domain , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Humans , Insecta , Protein Conformation , Protein Domains , Squalene/metabolism , Squalene Monooxygenase/antagonists & inhibitors
4.
Nat Commun ; 10(1): 96, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626880

ABSTRACT

Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Screening Assays, Antitumor , Squalene Monooxygenase/antagonists & inhibitors , Squalene Monooxygenase/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cholesterol/biosynthesis , Gene Deletion , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans
5.
ACS Med Chem Lett ; 9(4): 300-305, 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29670690

ABSTRACT

Somatic point mutations at a key arginine residue (R132) within the active site of the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) confer a novel gain of function in cancer cells, resulting in the production of d-2-hydroxyglutarate (2-HG), an oncometabolite. Elevated 2-HG levels are implicated in epigenetic alterations and impaired cellular differentiation. IDH1 mutations have been described in an array of hematologic malignancies and solid tumors. Here, we report the discovery of AG-120 (ivosidenib), an inhibitor of the IDH1 mutant enzyme that exhibits profound 2-HG lowering in tumor models and the ability to effect differentiation of primary patient AML samples ex vivo. Preliminary data from phase 1 clinical trials enrolling patients with cancers harboring an IDH1 mutation indicate that AG-120 has an acceptable safety profile and clinical activity.

6.
Blood ; 130(11): 1347-1356, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28760888

ABSTRACT

Pyruvate kinase (PK) deficiency is a rare genetic disease that causes chronic hemolytic anemia. There are currently no targeted therapies for PK deficiency. Here, we describe the identification and characterization of AG-348, an allosteric activator of PK that is currently in clinical trials for the treatment of PK deficiency. We demonstrate that AG-348 can increase the activity of wild-type and mutant PK enzymes in biochemical assays and in patient red blood cells treated ex vivo. These data illustrate the potential for AG-348 to restore the glycolytic pathway activity in patients with PK deficiency and ultimately lead to clinical benefit.


Subject(s)
Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Erythrocytes/enzymology , Pyruvate Kinase/deficiency , Pyruvate Kinase/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Anemia, Hemolytic, Congenital Nonspherocytic , Animals , Enzyme Activation/drug effects , Enzyme Activators/chemistry , Erythrocytes/drug effects , Humans , Kinetics , Mice , Piperazines , Pyruvate Kinase/drug effects , Pyruvate Metabolism, Inborn Errors , Quinolines/chemistry , Recombinant Proteins/metabolism , Sulfonamides/chemistry , Tissue Donors
7.
Cancer Discov ; 7(5): 478-493, 2017 05.
Article in English | MEDLINE | ID: mdl-28193778

ABSTRACT

Somatic gain-of-function mutations in isocitrate dehydrogenases (IDH) 1 and 2 are found in multiple hematologic and solid tumors, leading to accumulation of the oncometabolite (R)-2-hydroxyglutarate (2HG). 2HG competitively inhibits α-ketoglutarate-dependent dioxygenases, including histone demethylases and methylcytosine dioxygenases of the TET family, causing epigenetic dysregulation and a block in cellular differentiation. In vitro studies have provided proof of concept for mutant IDH inhibition as a therapeutic approach. We report the discovery and characterization of AG-221, an orally available, selective, potent inhibitor of the mutant IDH2 enzyme. AG-221 suppressed 2HG production and induced cellular differentiation in primary human IDH2 mutation-positive acute myeloid leukemia (AML) cells ex vivo and in xenograft mouse models. AG-221 also provided a statistically significant survival benefit in an aggressive IDH2R140Q-mutant AML xenograft mouse model. These findings supported initiation of the ongoing clinical trials of AG-221 in patients with IDH2 mutation-positive advanced hematologic malignancies.Significance: Mutations in IDH1/2 are identified in approximately 20% of patients with AML and contribute to leukemia via a block in hematopoietic cell differentiation. We have shown that the targeted inhibitor AG-221 suppresses the mutant IDH2 enzyme in multiple preclinical models and induces differentiation of malignant blasts, supporting its clinical development. Cancer Discov; 7(5); 478-93. ©2017 AACR.See related commentary by Thomas and Majeti, p. 459See related article by Shih et al., p. 494This article is highlighted in the In This Issue feature, p. 443.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/genetics , Triazines/pharmacology , Animals , Cell Line, Tumor , Humans , Isocitrate Dehydrogenase/genetics , Mice , Mutation , Xenograft Model Antitumor Assays
8.
Cell Rep ; 17(3): 876-890, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732861

ABSTRACT

Although aberrant metabolism in tumors has been well described, the identification of cancer subsets with particular metabolic vulnerabilities has remained challenging. Here, we conducted an siRNA screen focusing on enzymes involved in the tricarboxylic acid (TCA) cycle and uncovered a striking range of cancer cell dependencies on OGDH, the E1 subunit of the alpha-ketoglutarate dehydrogenase complex. Using an integrative metabolomics approach, we identified differential aspartate utilization, via the malate-aspartate shuttle, as a predictor of whether OGDH is required for proliferation in 3D culture assays and for the growth of xenograft tumors. These findings highlight an anaplerotic role of aspartate and, more broadly, suggest that differential nutrient utilization patterns can identify subsets of cancers with distinct metabolic dependencies for potential pharmacological intervention.


Subject(s)
Aspartic Acid/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Respiration/drug effects , Citric Acid Cycle/drug effects , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Humans , RNA, Small Interfering/metabolism
10.
ACS Med Chem Lett ; 6(1): 53-7, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25589930

ABSTRACT

The first allosteric, type III inhibitor of LIM-kinase 2 (LIMK2) is reported. A series of molecules that feature both an N-phenylsulfonamide and tertiary amide were not only very potent at LIMK2 but also were extremely selective against a panel of other kinases. Enzymatic kinetic studies showed these molecules to be noncompetitive with ATP, suggesting allosteric inhibition. X-ray crystallography confirmed that these sulfonamides are a rare example of a type III kinase inhibitor that binds away from the highly conserved hinge region and instead resides in the hydrophobic pocket formed in the DFG-out conformation of the kinase, thus accounting for the high level of selectivity observed.

11.
Chem Biol ; 21(9): 1143-61, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25237859

ABSTRACT

Cancer cells must carefully regulate their metabolism to maintain growth and division under varying nutrient and oxygen levels. Compelling data support the investigation of numerous enzymes as therapeutic targets to exploit metabolic vulnerabilities common to several cancer types. We discuss the rationale for developing such drugs and review three targets with central roles in metabolic pathways crucial for cancer cell growth: pyruvate kinase muscle isozyme splice variant 2 (PKM2) in glycolysis, glutaminase in glutaminolysis, and mutations in isocitrate dehydrogenase 1 and 2 isozymes (IDH1/2) in the tricarboxylic acid cycle. These targets exemplify the drugging approach to cancer metabolism, with allosteric modulation being the common theme. The first glutaminase and mutant IDH1/2 inhibitors have entered clinical testing, and early data are promising. Cancer metabolism provides a wealth of novel targets, and targeting allosteric sites promises to yield selective drugs with the potential to transform clinical outcomes across many cancer types.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Antineoplastic Agents/therapeutic use , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Citric Acid Cycle/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Glycolysis/drug effects , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
12.
Chem Biol ; 19(9): 1187-98, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22999886

ABSTRACT

Proliferating tumor cells use aerobic glycolysis to support their high metabolic demands. Paradoxically, increased glycolysis is often accompanied by expression of the lower activity PKM2 isoform, effectively constraining lower glycolysis. Here, we report the discovery of PKM2 activators with a unique allosteric binding mode. Characterization of how these compounds impact cancer cells revealed an unanticipated link between glucose and amino acid metabolism. PKM2 activation resulted in a metabolic rewiring of cancer cells manifested by a profound dependency on the nonessential amino acid serine for continued cell proliferation. Induction of serine auxotrophy by PKM2 activation was accompanied by reduced carbon flow into the serine biosynthetic pathway and increased expression of high affinity serine transporters. These data support the hypothesis that PKM2 expression confers metabolic flexibility to cancer cells that allows adaptation to nutrient stress.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Serine/metabolism , Small Molecule Libraries/pharmacology , Thyroid Hormones/metabolism , Allosteric Site/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Thyroid Hormone-Binding Proteins
13.
Curr Chem Genomics ; 4: 19-26, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20556201

ABSTRACT

Trytophan Hydroxylase Type I (TPH1), most abundantly expressed in the gastrointestinal tract, initiates the synthesis of serotonin by catalyzing hydroxylation of tryptophan in the presence of biopterin and oxygen. We have previously described three series of novel, periphery-specific TPH1 inhibitors that selectively deplete serotonin in the gastrointestinal tract. We have now determined co-crystal structures of TPH1 with three of these inhibitors at high resolution. Analysis of the structural data showed that each of the three inhibitors fills the tryptophan binding pocket of TPH1 without reaching into the binding site of the cofactor pterin, and induces major conformational changes of the enzyme. The enzyme-inhibitor complexes assume a compact conformation that is similar to the one in tryptophan complex. Kinetic analysis showed that all three inhibitors are competitive versus the substrate tryptophan, consistent with the structural data that the compounds occupy the tryptophan binding site. On the other hand, all three inhibitors appear to be uncompetitive versus the cofactor 6-methyltetrahydropterin, which is not only consistent with the structural data but also indicate that the hydroxylation reaction follows an ordered binding mechanism in which a productive complex is formed only if tryptophan binds only after pterin, similar to the kinetic mechanisms of tyrosine and phenylalanine hydroxylase.

14.
Bioorg Med Chem Lett ; 19(17): 5229-32, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19631532

ABSTRACT

Tryptophan hydroxylase (TPH) is a key enzyme in the synthesis of serotonin. As a neurotransmitter, serotonin plays important physiological roles both peripherally and centrally. Here we describe the discovery of substituted triazines as a novel class of tryptophan hydroxylase inhibitors. This class of TPH inhibitors can selectively reduce serotonin levels in murine intestine after oral administration without affecting levels in the brain. These TPH inhibitors may provide novel treatments for gastrointestinal disorders associated with dysregulation of the serotonergic system, such as chemotherapy-induced emesis and irritable bowel syndrome.


Subject(s)
Enzyme Inhibitors/chemistry , Pyrazines/chemistry , Tryptophan Hydroxylase/antagonists & inhibitors , Animals , Binding Sites , Cell Line , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Molecular Conformation , Pyrazines/chemical synthesis , Pyrazines/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Serotonin/biosynthesis , Structure-Activity Relationship , Tryptophan Hydroxylase/metabolism
15.
Magn Reson Chem ; 47(1): 38-52, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18991323

ABSTRACT

Since the introduction of NMR prediction software, medicinal chemists have imagined submitting their compounds to corporate compound registration systems that would ultimately display a simplified pass/fail result. We initially implemented such a system based on HPLC and liquid chromatography mass spectrometry (LCMS) data that is embedded within our industry standard sample submission and registration process. By using gradient-heteronuclear single quantum coherence (HSQC) experiments, we have extended this concept to NMR data through a comparison of experimentally acquired data against predicted (1)H and (13)C NMR data. Integration of our compound registration system with our analytical instruments now provides our chemists unattended and automated NMR verification for collections of submitted compounds. The benefits achieved from automated processing and interpretation of results produced enhanced confidence in our compound library and released the chemists from the tedium of manipulating large amounts of data. This allows scientists to focus more of their attention to the drug discovery process.


Subject(s)
Drug Discovery/methods , Magnetic Resonance Spectroscopy/methods , Automation , Pharmaceutical Preparations/analysis , Technology, Pharmaceutical
16.
J Med Chem ; 50(9): 2117-26, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17425298

ABSTRACT

Ligand-based virtual screening approaches were applied to search for new chemotype KCOs activating Kir6.2/SUR1 KATP channels. A total of 65 208 commercially available compounds, extracted from the ZINC archive, served as database for screening. In a first step, pharmacokinetic filtering via VolSurf reduced the initial database to 1913 compounds. Afterward, six molecules were selected as templates for similarity searches: similarity scores, obtained toward these templates, were calculated with the GRIND, FLAP, and TOPP approaches, which differently encode structural information into potential pharmacophores. In this way, we obtained 32 hit candidates, 16 via GRIND and eight each via FLAP and TOPP. For biological testing of the hit candidates, their effects on membrane potentials in HEK 293 cells expressing Kir6.2/SUR1 were studied. GRIND, FLAP, and TOPP all yielded hits, but no method top-ranked all the actives. Thus, parallel application of different approaches probably improves hit detection.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Insulin-Secreting Cells/metabolism , Ion Channel Gating , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels/chemistry , Receptors, Drug/chemistry , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/physiology , Cell Line , Databases, Factual , Humans , Insulin/metabolism , Insulin Secretion , Membrane Potentials/drug effects , Models, Molecular , Potassium Channels/drug effects , Potassium Channels/physiology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/physiology , Quantitative Structure-Activity Relationship , Receptors, Drug/drug effects , Receptors, Drug/physiology , Sulfonylurea Receptors
17.
Bioorg Med Chem Lett ; 15(15): 3637-42, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15978804

ABSTRACT

We report here a general method for the prediction of hERG potassium channel blockers using computational models generated from correlation analyses of a large dataset and pharmacophore-based GRIND descriptors. These 3D-QSAR models are compared favorably with other traditional and chemometric based HQSAR methods.


Subject(s)
Models, Molecular , Potassium Channel Blockers/chemistry , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Software , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/pharmacology , Binding Sites , Humans , Inhibitory Concentration 50 , Isomerism , Potassium Channel Blockers/pharmacology , Quantitative Structure-Activity Relationship
18.
J Med Chem ; 48(8): 2927-35, 2005 Apr 21.
Article in English | MEDLINE | ID: mdl-15828831

ABSTRACT

Trying to understand the complex interactions that substrates and inhibitors have with the efflux transporter P-glycoprotein has been the subject of various publications. In this work, we have confined our study to substrates by picking a diverse set of 129 compounds based on the efflux ratios from Caco-2 permeability measurements. These compounds were then evaluated for P-glycoprotein inhibition using a calcein-AM assay. The subsequent data was used in a 3D-QSAR analysis using GRIND pharmacophore-based and physicochemical descriptors. Pharmacophore-based descriptors produced a much more robust model than the one obtained from physicochemical-based descriptors. This supports the process proposed by Seelig and co-workers previously published whereby the substrate enters the membrane as the first step and is then recognized by P-glycoprotein in a second step. The strong correlation, highlighted by PLS statistical analysis, between pharmacophoric descriptors and inhibition values suggests that substrate interaction, with perhaps the mouth of the protein or another binding site, plays a key role in the efflux process, yielding a model in which diffusion across the membrane is less important than substrate-protein interaction. One pharmacophore emerged from the analysis of the model. We pose that the recognition elements, at least determined by the molecules used in this study, are two hydrophobic groups 16.5 A apart and two hydrogen-bond-acceptor groups 11.5 A apart and that the dimensions of the molecule also plays a role in its recognition as a substrate.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Biological Transport , Caco-2 Cells , Fluoresceins , Fluorescent Dyes , Humans , Models, Molecular , Multivariate Analysis , Permeability , Quantitative Structure-Activity Relationship
19.
J Med Chem ; 47(12): 3193-201, 2004 Jun 03.
Article in English | MEDLINE | ID: mdl-15163198

ABSTRACT

An extended VolSurf approach, that additionally includes SHAPE descriptors, was applied to a dataset of 55 quinolones. Bactericidal activity was measured at Bayer AG, Germany, for Gram-negative (Escherichia coli and Pseudomonas aeruginosa) and Gram-positive bacteria (Staphylococcus aureus and Enterococcus faecalis). Chemometric analysis was first approached via a classical VolSurf approach. The following descriptors were found most important: bactericidal activity particularly increases with high values of the best volume (BV11(OH2)) and the minimum energy (Emin1(OH2)) of the water probe, high values of the integy moment (ID(DRY)) of the lipophilic probe, and high values of the hydrophilic region (W(O)) of the hydrogen bond acceptor probe. Best volume (BV31(OH2)) of the water probe and best volume (BV12(DRY)) and lipophilic regions (D(DRY)) of the lipophilic probe as well as H-bonding capacity derived with the CO probe (HB(O)) are inversely related to activity. PLS analysis yields a five-component model with an r(2) of 0.83 and a q(2) of 0.43 after variable selection via fractional factorial design (FFD). Chemometric modeling could be improved by including newly derived SHAPE descriptors, which were merged with the VolSurf descriptors and subjected to PLS analysis. The global model of this extended VolSurf approach is optimal with two components and exhibits a significantly improved statistical quality; a marginally reduced r(2) (0.75 versus 0.83) is more than compensated by a highly improved predictivity with a q(2) of 0.63 versus 0.43. To prove model quality, external prediction of seven test set quinolones was performed. The precise prediction of all test set molecules nicely demonstrates the robustness and statistical significance of the obtained chemometric model using the extended VolSurf approach.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Quinolones/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Enterococcus faecalis/drug effects , Escherichia coli/drug effects , Models, Molecular , Pseudomonas aeruginosa/drug effects , Quantitative Structure-Activity Relationship , Quinolones/chemistry , Quinolones/pharmacology , Staphylococcus aureus/drug effects , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...