Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Sci Data ; 11(1): 363, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605048

ABSTRACT

Translational research requires data at multiple scales of biological organization. Advancements in sequencing and multi-omics technologies have increased the availability of these data, but researchers face significant integration challenges. Knowledge graphs (KGs) are used to model complex phenomena, and methods exist to construct them automatically. However, tackling complex biomedical integration problems requires flexibility in the way knowledge is modeled. Moreover, existing KG construction methods provide robust tooling at the cost of fixed or limited choices among knowledge representation models. PheKnowLator (Phenotype Knowledge Translator) is a semantic ecosystem for automating the FAIR (Findable, Accessible, Interoperable, and Reusable) construction of ontologically grounded KGs with fully customizable knowledge representation. The ecosystem includes KG construction resources (e.g., data preparation APIs), analysis tools (e.g., SPARQL endpoint resources and abstraction algorithms), and benchmarks (e.g., prebuilt KGs). We evaluated the ecosystem by systematically comparing it to existing open-source KG construction methods and by analyzing its computational performance when used to construct 12 different large-scale KGs. With flexible knowledge representation, PheKnowLator enables fully customizable KGs without compromising performance or usability.


Subject(s)
Biological Science Disciplines , Knowledge Bases , Pattern Recognition, Automated , Algorithms , Translational Research, Biomedical
2.
ACS Cent Sci ; 10(3): 494-510, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38559298

ABSTRACT

The ENPKG framework organizes large heterogeneous metabolomics data sets as a knowledge graph, offering exciting opportunities for drug discovery and chemodiversity characterization.

3.
Bioinform Adv ; 4(1): vbae036, 2024.
Article in English | MEDLINE | ID: mdl-38577542

ABSTRACT

Motivation: Graph representation learning is a family of related approaches that learn low-dimensional vector representations of nodes and other graph elements called embeddings. Embeddings approximate characteristics of the graph and can be used for a variety of machine-learning tasks such as novel edge prediction. For many biomedical applications, partial knowledge exists about positive edges that represent relationships between pairs of entities, but little to no knowledge is available about negative edges that represent the explicit lack of a relationship between two nodes. For this reason, classification procedures are forced to assume that the vast majority of unlabeled edges are negative. Existing approaches to sampling negative edges for training and evaluating classifiers do so by uniformly sampling pairs of nodes. Results: We show here that this sampling strategy typically leads to sets of positive and negative examples with imbalanced node degree distributions. Using representative heterogeneous biomedical knowledge graph and random walk-based graph machine learning, we show that this strategy substantially impacts classification performance. If users of graph machine-learning models apply the models to prioritize examples that are drawn from approximately the same distribution as the positive examples are, then performance of models as estimated in the validation phase may be artificially inflated. We present a degree-aware node sampling approach that mitigates this effect and is simple to implement. Availability and implementation: Our code and data are publicly available at https://github.com/monarch-initiative/negativeExampleSelection.

4.
Nucleic Acids Res ; 52(D1): D938-D949, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38000386

ABSTRACT

Bridging the gap between genetic variations, environmental determinants, and phenotypic outcomes is critical for supporting clinical diagnosis and understanding mechanisms of diseases. It requires integrating open data at a global scale. The Monarch Initiative advances these goals by developing open ontologies, semantic data models, and knowledge graphs for translational research. The Monarch App is an integrated platform combining data about genes, phenotypes, and diseases across species. Monarch's APIs enable access to carefully curated datasets and advanced analysis tools that support the understanding and diagnosis of disease for diverse applications such as variant prioritization, deep phenotyping, and patient profile-matching. We have migrated our system into a scalable, cloud-based infrastructure; simplified Monarch's data ingestion and knowledge graph integration systems; enhanced data mapping and integration standards; and developed a new user interface with novel search and graph navigation features. Furthermore, we advanced Monarch's analytic tools by developing a customized plugin for OpenAI's ChatGPT to increase the reliability of its responses about phenotypic data, allowing us to interrogate the knowledge in the Monarch graph using state-of-the-art Large Language Models. The resources of the Monarch Initiative can be found at monarchinitiative.org and its corresponding code repository at github.com/monarch-initiative/monarch-app.


Subject(s)
Databases, Factual , Disease , Genes , Phenotype , Humans , Internet , Databases, Factual/standards , Software , Genes/genetics , Disease/genetics
5.
Bioinformatics ; 39(7)2023 07 01.
Article in English | MEDLINE | ID: mdl-37389415

ABSTRACT

MOTIVATION: Knowledge graphs (KGs) are a powerful approach for integrating heterogeneous data and making inferences in biology and many other domains, but a coherent solution for constructing, exchanging, and facilitating the downstream use of KGs is lacking. RESULTS: Here we present KG-Hub, a platform that enables standardized construction, exchange, and reuse of KGs. Features include a simple, modular extract-transform-load pattern for producing graphs compliant with Biolink Model (a high-level data model for standardizing biological data), easy integration of any OBO (Open Biological and Biomedical Ontologies) ontology, cached downloads of upstream data sources, versioned and automatically updated builds with stable URLs, web-browsable storage of KG artifacts on cloud infrastructure, and easy reuse of transformed subgraphs across projects. Current KG-Hub projects span use cases including COVID-19 research, drug repurposing, microbial-environmental interactions, and rare disease research. KG-Hub is equipped with tooling to easily analyze and manipulate KGs. KG-Hub is also tightly integrated with graph machine learning (ML) tools which allow automated graph ML, including node embeddings and training of models for link prediction and node classification. AVAILABILITY AND IMPLEMENTATION: https://kghub.org.


Subject(s)
Biological Ontologies , COVID-19 , Humans , Pattern Recognition, Automated , Rare Diseases , Machine Learning
6.
EBioMedicine ; 87: 104413, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36563487

ABSTRACT

BACKGROUND: Stratification of patients with post-acute sequelae of SARS-CoV-2 infection (PASC, or long COVID) would allow precision clinical management strategies. However, long COVID is incompletely understood and characterised by a wide range of manifestations that are difficult to analyse computationally. Additionally, the generalisability of machine learning classification of COVID-19 clinical outcomes has rarely been tested. METHODS: We present a method for computationally modelling PASC phenotype data based on electronic healthcare records (EHRs) and for assessing pairwise phenotypic similarity between patients using semantic similarity. Our approach defines a nonlinear similarity function that maps from a feature space of phenotypic abnormalities to a matrix of pairwise patient similarity that can be clustered using unsupervised machine learning. FINDINGS: We found six clusters of PASC patients, each with distinct profiles of phenotypic abnormalities, including clusters with distinct pulmonary, neuropsychiatric, and cardiovascular abnormalities, and a cluster associated with broad, severe manifestations and increased mortality. There was significant association of cluster membership with a range of pre-existing conditions and measures of severity during acute COVID-19. We assigned new patients from other healthcare centres to clusters by maximum semantic similarity to the original patients, and showed that the clusters were generalisable across different hospital systems. The increased mortality rate originally identified in one cluster was consistently observed in patients assigned to that cluster in other hospital systems. INTERPRETATION: Semantic phenotypic clustering provides a foundation for assigning patients to stratified subgroups for natural history or therapy studies on PASC. FUNDING: NIH (TR002306/OT2HL161847-01/OD011883/HG010860), U.S.D.O.E. (DE-AC02-05CH11231), Donald A. Roux Family Fund at Jackson Laboratory, Marsico Family at CU Anschutz.


Subject(s)
COVID-19 , Post-Acute COVID-19 Syndrome , Humans , Disease Progression , SARS-CoV-2
7.
Nat Comput Sci ; 3(6): 552-568, 2023 Jun.
Article in English | MEDLINE | ID: mdl-38177435

ABSTRACT

Graph representation learning methods opened new avenues for addressing complex, real-world problems represented by graphs. However, many graphs used in these applications comprise millions of nodes and billions of edges and are beyond the capabilities of current methods and software implementations. We present GRAPE (Graph Representation Learning, Prediction and Evaluation), a software resource for graph processing and embedding that is able to scale with big graphs by using specialized and smart data structures, algorithms, and a fast parallel implementation of random-walk-based methods. Compared with state-of-the-art software resources, GRAPE shows an improvement of orders of magnitude in empirical space and time complexity, as well as competitive edge- and node-label prediction performance. GRAPE comprises approximately 1.7 million well-documented lines of Python and Rust code and provides 69 node-embedding methods, 25 inference models, a collection of efficient graph-processing utilities, and over 80,000 graphs from the literature and other sources. Standardized interfaces allow a seamless integration of third-party libraries, while ready-to-use and modular pipelines permit an easy-to-use evaluation of graph-representation-learning methods, therefore also positioning GRAPE as a software resource that performs a fair comparison between methods and libraries for graph processing and embedding.


Subject(s)
Libraries , Vitis , Algorithms , Software , Learning
8.
BMC Bioinformatics ; 23(Suppl 2): 154, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36510125

ABSTRACT

BACKGROUND: Cis-regulatory regions (CRRs) are non-coding regions of the DNA that fine control the spatio-temporal pattern of transcription; they are involved in a wide range of pivotal processes such as the development of specific cell-lines/tissues and the dynamic cell response to physiological stimuli. Recent studies showed that genetic variants occurring in CRRs are strongly correlated with pathogenicity or deleteriousness. Considering the central role of CRRs in the regulation of physiological and pathological conditions, the correct identification of CRRs and of their tissue-specific activity status through Machine Learning methods plays a major role in dissecting the impact of genetic variants on human diseases. Unfortunately, the problem is still open, though some promising results have been already reported by (deep) machine-learning based methods that predict active promoters and enhancers in specific tissues or cell lines by encoding epigenetic or spectral features directly extracted from DNA sequences. RESULTS: We present the experiments we performed to compare two Deep Neural Networks, a Feed-Forward Neural Network model working on epigenomic features, and a Convolutional Neural Network model working only on genomic sequence, targeted to the identification of enhancer- and promoter-activity in specific cell lines. While performing experiments to understand how the experimental setup influences the prediction performance of the methods, we particularly focused on (1) automatic model selection performed by Bayesian optimization and (2) exploring different data rebalancing setups for reducing negative unbalancing effects. CONCLUSIONS: Results show that (1) automatic model selection by Bayesian optimization improves the quality of the learner; (2) data rebalancing considerably impacts the prediction performance of the models; test set rebalancing may provide over-optimistic results, and should therefore be cautiously applied; (3) despite working on sequence data, convolutional models obtain performance close to those of feed forward models working on epigenomic information, which suggests that also sequence data carries informative content for CRR-activity prediction. We therefore suggest combining both models/data types in future works.


Subject(s)
Deep Learning , Humans , Bayes Theorem , Regulatory Sequences, Nucleic Acid , Neural Networks, Computer , Machine Learning
9.
medRxiv ; 2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35665012

ABSTRACT

Accurate stratification of patients with post-acute sequelae of SARS-CoV-2 infection (PASC, or long COVID) would allow precision clinical management strategies. However, the natural history of long COVID is incompletely understood and characterized by an extremely wide range of manifestations that are difficult to analyze computationally. In addition, the generalizability of machine learning classification of COVID-19 clinical outcomes has rarely been tested. We present a method for computationally modeling PASC phenotype data based on electronic healthcare records (EHRs) and for assessing pairwise phenotypic similarity between patients using semantic similarity. Our approach defines a nonlinear similarity function that maps from a feature space of phenotypic abnormalities to a matrix of pairwise patient similarity that can be clustered using unsupervised machine learning procedures. Using k-means clustering of this similarity matrix, we found six distinct clusters of PASC patients, each with distinct profiles of phenotypic abnormalities. There was a significant association of cluster membership with a range of pre-existing conditions and with measures of severity during acute COVID-19. Two of the clusters were associated with severe manifestations and displayed increased mortality. We assigned new patients from other healthcare centers to one of the six clusters on the basis of maximum semantic similarity to the original patients. We show that the identified clusters were generalizable across different hospital systems and that the increased mortality rate was consistently observed in two of the clusters. Semantic phenotypic clustering can provide a foundation for assigning patients to stratified subgroups for natural history or therapy studies on PASC.

10.
Virol J ; 19(1): 84, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35570298

ABSTRACT

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used to reduce pain, fever, and inflammation but have been associated with complications in community-acquired pneumonia. Observations shortly after the start of the COVID-19 pandemic in 2020 suggested that ibuprofen was associated with an increased risk of adverse events in COVID-19 patients, but subsequent observational studies failed to demonstrate increased risk and in one case showed reduced risk associated with NSAID use. METHODS: A 38-center retrospective cohort study was performed that leveraged the harmonized, high-granularity electronic health record data of the National COVID Cohort Collaborative. A propensity-matched cohort of 19,746 COVID-19 inpatients was constructed by matching cases (treated with NSAIDs at the time of admission) and 19,746 controls (not treated) from 857,061 patients with COVID-19 available for analysis. The primary outcome of interest was COVID-19 severity in hospitalized patients, which was classified as: moderate, severe, or mortality/hospice. Secondary outcomes were acute kidney injury (AKI), extracorporeal membrane oxygenation (ECMO), invasive ventilation, and all-cause mortality at any time following COVID-19 diagnosis. RESULTS: Logistic regression showed that NSAID use was not associated with increased COVID-19 severity (OR: 0.57 95% CI: 0.53-0.61). Analysis of secondary outcomes using logistic regression showed that NSAID use was not associated with increased risk of all-cause mortality (OR 0.51 95% CI: 0.47-0.56), invasive ventilation (OR: 0.59 95% CI: 0.55-0.64), AKI (OR: 0.67 95% CI: 0.63-0.72), or ECMO (OR: 0.51 95% CI: 0.36-0.7). In contrast, the odds ratios indicate reduced risk of these outcomes, but our quantitative bias analysis showed E-values of between 1.9 and 3.3 for these associations, indicating that comparatively weak or moderate confounder associations could explain away the observed associations. CONCLUSIONS: Study interpretation is limited by the observational design. Recording of NSAID use may have been incomplete. Our study demonstrates that NSAID use is not associated with increased COVID-19 severity, all-cause mortality, invasive ventilation, AKI, or ECMO in COVID-19 inpatients. A conservative interpretation in light of the quantitative bias analysis is that there is no evidence that NSAID use is associated with risk of increased severity or the other measured outcomes. Our results confirm and extend analogous findings in previous observational studies using a large cohort of patients drawn from 38 centers in a nationally representative multicenter database.


Subject(s)
Acute Kidney Injury , COVID-19 , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , COVID-19 Testing , Cohort Studies , Humans , Pandemics , Retrospective Studies
11.
NAR Genom Bioinform ; 3(4): lqab113, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34888523

ABSTRACT

Inhibiting protein kinases (PKs) that cause cancers has been an important topic in cancer therapy for years. So far, almost 8% of >530 PKs have been targeted by FDA-approved medications, and around 150 protein kinase inhibitors (PKIs) have been tested in clinical trials. We present an approach based on natural language processing and machine learning to investigate the relations between PKs and cancers, predicting PKs whose inhibition would be efficacious to treat a certain cancer. Our approach represents PKs and cancers as semantically meaningful 100-dimensional vectors based on word and concept neighborhoods in PubMed abstracts. We use information about phase I-IV trials in ClinicalTrials.gov to construct a training set for random forest classification. Our results with historical data show that associations between PKs and specific cancers can be predicted years in advance with good accuracy. Our tool can be used to predict the relevance of inhibiting PKs for specific cancers and to support the design of well-focused clinical trials to discover novel PKIs for cancer therapy.

12.
medRxiv ; 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-33907758

ABSTRACT

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used to reduce pain, fever, and inflammation but have been associated with complications in community-acquired pneumonia. Observations shortly after the start of the COVID-19 pandemic in 2020 suggested that ibuprofen was associated with an increased risk of adverse events in COVID-19 patients, but subsequent observational studies failed to demonstrate increased risk and in one case showed reduced risk associated with NSAID use. METHODS: A 38-center retrospective cohort study was performed that leveraged the harmonized, high-granularity electronic health record data of the National COVID Cohort Collaborative. A propensity-matched cohort of COVID-19 inpatients was constructed by matching cases (treated with NSAIDs) and controls (not treated) from 857,061 patients with COVID-19. The primary outcome of interest was COVID-19 severity in hospitalized patients, which was classified as: moderate, severe, or mortality/hospice. Secondary outcomes were acute kidney injury (AKI), extracorporeal membrane oxygenation (ECMO), invasive ventilation, and all-cause mortality at any time following COVID-19 diagnosis. RESULTS: Logistic regression showed that NSAID use was not associated with increased COVID-19 severity (OR: 0.57 95% CI: 0.53-0.61). Analysis of secondary outcomes using logistic regression showed that NSAID use was not associated with increased risk of all-cause mortality (OR 0.51 95% CI: 0.47-0.56), invasive ventilation (OR: 0.59 95% CI: 0.55-0.64), AKI (OR: 0.67 95% CI: 0.63-0.72), or ECMO (OR: 0.51 95% CI: 0.36-0.7). In contrast, the odds ratios indicate reduced risk of these outcomes, but our quantitative bias analysis showed E-values of between 1.9 and 3.3 for these associations, indicating that comparatively weak or moderate confounder associations could explain away the observed associations. CONCLUSIONS: Study interpretation is limited by the observational design. Recording of NSAID use may have been incomplete. Our study demonstrates that NSAID use is not associated with increased COVID-19 severity, all-cause mortality, invasive ventilation, AKI, or ECMO in COVID-19 inpatients. A conservative interpretation in light of the quantitative bias analysis is that there is no evidence that NSAID use is associated with risk of increased severity or the other measured outcomes. Our findings are the largest EHR-based analysis of the effect of NSAIDs on outcome in COVID-19 patients to date. Our results confirm and extend analogous findings in previous observational studies using a large cohort of patients drawn from 38 centers in a nationally representative multicenter database.

13.
Patterns (N Y) ; 2(1): 100155, 2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33196056

ABSTRACT

Integrated, up-to-date data about SARS-CoV-2 and COVID-19 is crucial for the ongoing response to the COVID-19 pandemic by the biomedical research community. While rich biological knowledge exists for SARS-CoV-2 and related viruses (SARS-CoV, MERS-CoV), integrating this knowledge is difficult and time-consuming, since much of it is in siloed databases or in textual format. Furthermore, the data required by the research community vary drastically for different tasks; the optimal data for a machine learning task, for example, is much different from the data used to populate a browsable user interface for clinicians. To address these challenges, we created KG-COVID-19, a flexible framework that ingests and integrates heterogeneous biomedical data to produce knowledge graphs (KGs), and applied it to create a KG for COVID-19 response. This KG framework also can be applied to other problems in which siloed biomedical data must be quickly integrated for different research applications, including future pandemics.

14.
bioRxiv ; 2020 Aug 18.
Article in English | MEDLINE | ID: mdl-32839776

ABSTRACT

Integrated, up-to-date data about SARS-CoV-2 and coronavirus disease 2019 (COVID-19) is crucial for the ongoing response to the COVID-19 pandemic by the biomedical research community. While rich biological knowledge exists for SARS-CoV-2 and related viruses (SARS-CoV, MERS-CoV), integrating this knowledge is difficult and time consuming, since much of it is in siloed databases or in textual format. Furthermore, the data required by the research community varies drastically for different tasks - the optimal data for a machine learning task, for example, is much different from the data used to populate a browsable user interface for clinicians. To address these challenges, we created KG-COVID-19, a flexible framework that ingests and integrates biomedical data to produce knowledge graphs (KGs) for COVID-19 response. This KG framework can also be applied to other problems in which siloed biomedical data must be quickly integrated for different research applications, including future pandemics. BIGGER PICTURE: An effective response to the COVID-19 pandemic relies on integration of many different types of data available about SARS-CoV-2 and related viruses. KG-COVID-19 is a framework for producing knowledge graphs that can be customized for downstream applications including machine learning tasks, hypothesis-based querying, and browsable user interface to enable researchers to explore COVID-19 data and discover relationships.

15.
Gigascience ; 9(5)2020 05 01.
Article in English | MEDLINE | ID: mdl-32444882

ABSTRACT

BACKGROUND: Several prediction problems in computational biology and genomic medicine are characterized by both big data as well as a high imbalance between examples to be learned, whereby positive examples can represent a tiny minority with respect to negative examples. For instance, deleterious or pathogenic variants are overwhelmed by the sea of neutral variants in the non-coding regions of the genome: thus, the prediction of deleterious variants is a challenging, highly imbalanced classification problem, and classical prediction tools fail to detect the rare pathogenic examples among the huge amount of neutral variants or undergo severe restrictions in managing big genomic data. RESULTS: To overcome these limitations we propose parSMURF, a method that adopts a hyper-ensemble approach and oversampling and undersampling techniques to deal with imbalanced data, and parallel computational techniques to both manage big genomic data and substantially speed up the computation. The synergy between Bayesian optimization techniques and the parallel nature of parSMURF enables efficient and user-friendly automatic tuning of the hyper-parameters of the algorithm, and allows specific learning problems in genomic medicine to be easily fit. Moreover, by using MPI parallel and machine learning ensemble techniques, parSMURF can manage big data by partitioning them across the nodes of a high-performance computing cluster. Results with synthetic data and with single-nucleotide variants associated with Mendelian diseases and with genome-wide association study hits in the non-coding regions of the human genome, involhing millions of examples, show that parSMURF achieves state-of-the-art results and an 80-fold speed-up with respect to the sequential version. CONCLUSIONS: parSMURF is a parallel machine learning tool that can be trained to learn different genomic problems, and its multiple levels of parallelization and high scalability allow us to efficiently fit problems characterized by big and imbalanced genomic data. The C++ OpenMP multi-core version tailored to a single workstation and the C++ MPI/OpenMP hybrid multi-core and multi-node parSMURF version tailored to a High Performance Computing cluster are both available at https://github.com/AnacletoLAB/parSMURF.


Subject(s)
Computational Biology/methods , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study/methods , Software , Algorithms , Databases, Genetic , Genomics/methods , Humans , Machine Learning , Reproducibility of Results
16.
IEEE Access ; 8: 196299-196325, 2020.
Article in English | MEDLINE | ID: mdl-34812365

ABSTRACT

Between January and October of 2020, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has infected more than 34 million persons in a worldwide pandemic leading to over one million deaths worldwide (data from the Johns Hopkins University). Since the virus begun to spread, emergency departments were busy with COVID-19 patients for whom a quick decision regarding in- or outpatient care was required. The virus can cause characteristic abnormalities in chest radiographs (CXR), but, due to the low sensitivity of CXR, additional variables and criteria are needed to accurately predict risk. Here, we describe a computerized system primarily aimed at extracting the most relevant radiological, clinical, and laboratory variables for improving patient risk prediction, and secondarily at presenting an explainable machine learning system, which may provide simple decision criteria to be used by clinicians as a support for assessing patient risk. To achieve robust and reliable variable selection, Boruta and Random Forest (RF) are combined in a 10-fold cross-validation scheme to produce a variable importance estimate not biased by the presence of surrogates. The most important variables are then selected to train a RF classifier, whose rules may be extracted, simplified, and pruned to finally build an associative tree, particularly appealing for its simplicity. Results show that the radiological score automatically computed through a neural network is highly correlated with the score computed by radiologists, and that laboratory variables, together with the number of comorbidities, aid risk prediction. The prediction performance of our approach was compared to that that of generalized linear models and shown to be effective and robust. The proposed machine learning-based computational system can be easily deployed and used in emergency departments for rapid and accurate risk prediction in COVID-19 patients.

17.
Ultrason Sonochem ; 27: 30-36, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26186817

ABSTRACT

An ultrasound-assisted three-component, one-pot domino reaction with ferrocenecarboxaldehyde is herein reported. The sequence of reactions entails the allylindation and dehydrative alkylation of stabilized C-nucleophiles (e.g. electron-rich-(hetero)aromatics and stabilized enols) and N-nucleophiles (e.g. azoles). Sonochemical reactions have been performed in three different high-intensity reactors: a bath (20.3 kHz, 60 W), as well as two cup horns working at 19.9 kHz (75 W) and 300.5 kHz (70 W) giving a library of 18 new ferrocenyl derivatives.

18.
Bioorg Med Chem ; 20(6): 2152-7, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22342267

ABSTRACT

Evidences of oseltamivir resistant influenza patients raised the need of novel neuraminidase inhibitors. In this study, five oseltamivir analogs PMC-31-PMC-36, synthesised according to the outcomes of a rational design analysis aimed to investigate the effects of substitution at the 5-amino and 4-amido groups of oseltamivir on its antiviral activity, were screened for their inhibition against neuraminidase N1 and N3. The enzymes used as models were from the avian influenza A H7N1 and H7N3 viruses. The neuraminidase inhibition assay was carried out by using recombinant species obtained from a baculovirus expression system and the fluorogenic substrate MUNANA. The assay was validated by using oseltamivir carboxylate as a reference inhibitor. Among the tested compounds, PMC-36 showed the highest inhibition on N1 with an IC(50) of 14.6±3.0nM (oseltamivir 25±4nM), while PMC-35 showed a significant inhibitory effect on N3 with an IC(50) of 0.1±0.03nM (oseltamivir 0.2±0.02nM). The analysis of the inhibitory properties of this panel of compounds allowed a preliminary assessment of a structure-activity relationship for the modification of the 4-amido and 5-amino groups of oseltamivir carboxylate. The substitution of the acetamido group in the oseltamivir structure with a 2-butenylamido moiety reduced the observed activity, while the introduction of a propenylamido group was well tolerated. Substitution of the free 5-amino group of oseltamivir carboxylate with an azide, decreased the activity against both N1 and N3. When these structural changes were both introduced, a dramatic reduction of activity was observed for both N1 and N3. The alkylation of the free 5-amino group in oseltamivir carboxylate introducing an isopropyl group seemed to increase the inhibitory effect for both N1 and N3 neuraminidases, displaying a more pronounced effect against N1.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Influenza A virus/enzymology , Influenza in Birds/drug therapy , Neuraminidase/antagonists & inhibitors , Oseltamivir/analogs & derivatives , Oseltamivir/pharmacology , Animals , Antiviral Agents/chemical synthesis , Binding Sites , Birds/virology , Influenza A Virus, H7N1 Subtype/chemistry , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/enzymology , Influenza A Virus, H7N3 Subtype/chemistry , Influenza A Virus, H7N3 Subtype/drug effects , Influenza A Virus, H7N3 Subtype/enzymology , Influenza A virus/chemistry , Influenza A virus/drug effects , Influenza in Birds/enzymology , Models, Molecular , Neuraminidase/chemistry , Neuraminidase/metabolism , Oseltamivir/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...