Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Brief Bioinform ; 24(6)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37833838

RESUMO

The available protein structure data are rapidly increasing. Within these structures, numerous local structural sites depict the details characterizing structure and function. However, searching and analyzing these sites extensively and at scale poses a challenge. We present a new method to search local sites in protein structure databases using residue-defined local 3D micro-environments. We implemented the method in a new tool called MicroMiner and demonstrate the capabilities of residue micro-environment search on the example of structural mutation analysis. Usually, experimental structures for both the wild-type and the mutant are unavailable for comparison. With MicroMiner, we extracted $>255 \times 10^{6}$ amino acid pairs in protein structures from the PDB, exemplifying single mutations' local structural changes for single chains and $>45 \times 10^{6}$ pairs for protein-protein interfaces. We further annotate existing data sets of experimentally measured mutation effects, like $\Delta \Delta G$ measurements, with the extracted structure pairs to combine the mutation effect measurement with the structural change upon mutation. In addition, we show how MicroMiner can bridge the gap between mutation analysis and structure-based drug design tools. MicroMiner is available as a command line tool and interactively on the https://proteins.plus/ webserver.


Assuntos
Aminoácidos , Proteínas , Bases de Dados de Proteínas , Proteínas/genética , Proteínas/química , Aminoácidos/química
2.
J Chem Inf Model ; 64(6): 2008-2020, 2024 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-38466793

RESUMO

Chemical fragment spaces exceed traditional virtual compound libraries by orders of magnitude, making them ideal search spaces for drug design projects. However, due to their immense size, they are not compatible with traditional analysis and search algorithms that rely on the enumeration of molecules. In this paper, we present SpaceProp2, an evolution of the SpaceProp algorithm, which enables the calculation of exact property distributions for chemical fragment spaces without enumerating them. We extend the original algorithm by the capabilities to compute distributions for the TPSA, the number of rotatable bonds, and the occurrence of user-defined molecular structures in the form of SMARTS patterns. Furthermore, SpaceProp2 produces example molecules for every property bin, enabling a detailed interpretation of the distributions. We demonstrate SpaceProp2 on six established make-on-demand chemical fragment spaces as well as BICLAIM, the in-house fragment space of Boehringer Ingelheim. The possibility to search multiple SMARTS patterns simultaneously as well as the produced example molecules offers previously impossible insights into the composition of these vast combinatorial molecule collections, making it an ideal tool for the analysis and design of chemical fragment spaces.


Assuntos
Algoritmos , Desenho de Fármacos , Estrutura Molecular
3.
J Chem Inf Model ; 64(8): 3332-3349, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38470439

RESUMO

Analyzing the similarity of protein interfaces in protein-protein interactions gives new insights into protein function and assists in discovering new drugs. Usually, tools that assess the similarity focus on the interactions between two protein interfaces, while sometimes we only have one predicted interface. Herein, we present PiMine, a database-driven protein interface similarity search. It compares interface residues of one or two interacting chains by calculating and searching tetrahedral geometric patterns of α-carbon atoms and calculating physicochemical and shape-based similarity. On a dedicated, tailor-made dataset, we show that PiMine outperforms commonly used comparison tools in terms of early enrichment when considering interfaces of sequentially and structurally unrelated proteins. In an application example, we demonstrate its usability for protein interaction partner prediction by comparing predicted interfaces to known protein-protein interfaces.


Assuntos
Bases de Dados de Proteínas , Proteínas , Proteínas/química , Proteínas/metabolismo , Conformação Proteica , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Modelos Moleculares
4.
J Chem Inf Model ; 64(1): 219-237, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-38108627

RESUMO

Molecular docking is a standard technique in structure-based drug design (SBDD). It aims to predict the 3D structure of a small molecule in the binding site of a receptor (often a protein). Despite being a common technique, it often necessitates multiple tools and involves manual steps. Here, we present the JAMDA preprocessing and docking workflow that is easy to use and allows fully automated docking. We evaluate the JAMDA docking workflow on binding sites extracted from the complete PDB and derive key factors determining JAMDA's docking performance. With that, we try to remove most of the bias due to manual intervention and provide a realistic estimate of the redocking performance of our JAMDA preprocessing and docking workflow for any PDB structure. On this large PDBScan22 data set, our JAMDA workflow finds a pose with an RMSD of at most 2 Å to the crystal ligand on the top rank for 30.1% of the structures. When applying objective structure quality filters to the PDBScan22 data set, the success rate increases to 61.8%. Given the prepared structures from the JAMDA preprocessing pipeline, both JAMDA and the widely used AutoDock Vina perform comparably on this filtered data set (the PDBScan22-HQ data set).


Assuntos
Desenho de Fármacos , Simulação de Acoplamento Molecular , Sítios de Ligação , Ligantes , Ligação Proteica
5.
J Comput Aided Mol Des ; 38(1): 23, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38814371

RESUMO

In this work, we present the frontend of GeoMine and showcase its application, focusing on the new features of its latest version. GeoMine is a search engine for ligand-bound and predicted empty binding sites in the Protein Data Bank. In addition to its basic text-based search functionalities, GeoMine offers a geometric query type for searching binding sites with a specific relative spatial arrangement of chemical features such as heavy atoms and intermolecular interactions. In contrast to a text search that requires simple and easy-to-formulate user input, a 3D input is more complex, and its specification can be challenging for users. GeoMine's new version aims to address this issue from the graphical user interface perspective by introducing an additional visualization concept and a new query template type. In its latest version, GeoMine extends its query-building capabilities primarily through input formulation in 2D. The 2D editor is fully synchronized with GeoMine's 3D editor and provides the same functionality. It enables template-free query generation and template-based query selection directly in 2D pose diagrams. In addition, the query generation with the 3D editor now supports predicted empty binding sites for AlphaFold structures as query templates. GeoMine is freely accessible on the ProteinsPlus web server ( https://proteins.plus ).


Assuntos
Bases de Dados de Proteínas , Ligação Proteica , Proteínas , Interface Usuário-Computador , Ligantes , Sítios de Ligação , Proteínas/química , Proteínas/metabolismo , Software , Ferramenta de Busca , Conformação Proteica , Modelos Moleculares
6.
J Comput Aided Mol Des ; 38(1): 13, 2024 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-38493240

RESUMO

The growing size of make-on-demand chemical libraries is posing new challenges to cheminformatics. These ultra-large chemical libraries became too large for exhaustive enumeration. Using a combinatorial approach instead, the resource requirement scales approximately with the number of synthons instead of the number of molecules. This gives access to billions or trillions of compounds as so-called chemical spaces with moderate hardware and in a reasonable time frame. While extremely performant ligand-based 2D methods exist in this context, 3D methods still largely rely on exhaustive enumeration and therefore fail to apply. Here, we present SpaceGrow: a novel shape-based 3D approach for ligand-based virtual screening of billions of compounds within hours on a single CPU. Compared to a conventional superposition tool, SpaceGrow shows comparable pose reproduction capacity based on RMSD and superior ranking performance while being orders of magnitude faster. Result assessment of two differently sized subsets of the eXplore space reveals a higher probability of finding superior results in larger spaces highlighting the potential of searching in ultra-large spaces. Furthermore, the application of SpaceGrow in a drug discovery workflow was investigated in four examples involving G protein-coupled receptors (GPCRs) with the aim to identify compounds with similar binding capabilities and molecular novelty.


Assuntos
Descoberta de Drogas , Bibliotecas de Moléculas Pequenas , Ligantes , Bibliotecas de Moléculas Pequenas/química , Descoberta de Drogas/métodos
7.
Nucleic Acids Res ; 50(W1): W611-W615, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35489057

RESUMO

Upon the ever-increasing number of publicly available experimentally determined and predicted protein and nucleic acid structures, the demand for easy-to-use tools to investigate these structural models is higher than ever before. The ProteinsPlus web server (https://proteins.plus) comprises a growing collection of molecular modeling tools focusing on protein-ligand interactions. It enables quick access to structural investigations ranging from structure analytics and search methods to molecular docking. It is by now well-established in the community and constantly extended. The server gives easy access not only to experts but also to students and occasional users from the field of life sciences. Here, we describe its recently added new features and tools, beyond them a novel method for on-the-fly molecular docking and a search method for single-residue substitutions in local regions of a protein structure throughout the whole Protein Data Bank. Finally, we provide a glimpse into new avenues for the annotation of AlphaFold structures which are directly accessible via a RESTful service on the ProteinsPlus web server.


Assuntos
Proteínas , Software , Simulação de Acoplamento Molecular , Proteínas/química , Modelos Moleculares , Internet
8.
Arch Pharm (Weinheim) ; 357(5): e2300661, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38335311

RESUMO

Drug discovery and design challenges, such as drug repurposing, analyzing protein-ligand and protein-protein complexes, ligand promiscuity studies, or function prediction, can be addressed by protein binding site similarity analysis. Although numerous tools exist, they all have individual strengths and drawbacks with regard to run time, provision of structure superpositions, and applicability to diverse application domains. Here, we introduce SiteMine, an all-in-one database-driven, alignment-providing binding site similarity search tool to tackle the most pressing challenges of binding site comparison. The performance of SiteMine is evaluated on the ProSPECCTs benchmark, showing a promising performance on most of the data sets. The method performs convincingly regarding all quality criteria for reliable binding site comparison, offering a novel state-of-the-art approach for structure-based molecular design based on binding site comparisons. In a SiteMine showcase, we discuss the high structural similarity between cathepsin L and calpain 1 binding sites and give an outlook on the impact of this finding on structure-based drug design. SiteMine is available at https://uhh.de/naomi.


Assuntos
Bases de Dados de Proteínas , Sítios de Ligação , Ligantes , Desenho de Fármacos , Descoberta de Drogas , Proteínas/química , Proteínas/metabolismo , Ligação Proteica , Conformação Proteica , Humanos , Catepsina L/metabolismo , Catepsina L/química , Catepsina L/antagonistas & inibidores
9.
J Chem Inf Model ; 63(10): 3128-3137, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37130052

RESUMO

Binding site prediction on protein structures is a crucial step in early phase drug discovery whenever experimental or predicted structure models are involved. DoGSite belongs to the widely used tools for this task. It is a grid-based method that uses a Difference-of-Gaussian filter to detect cavities on the protein surface. We recently reimplemented the first version of this method, released in 2010, focusing on improved binding site detection in the presence of ligands and optimized parameters for more robust, reliable, and fast predictions and binding site descriptor calculations. Here, we introduce the new version, DoGSite3, compare it to its predecessor, and re-evaluate DoGSite on published data sets for a large-scale comparative performance evaluation.


Assuntos
Descoberta de Drogas , Proteínas , Sítios de Ligação , Proteínas/química , Domínios Proteicos , Ligantes , Ligação Proteica
10.
J Chem Inf Model ; 63(8): 2573-2585, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37018549

RESUMO

In many molecular modeling applications, the standard procedure is still to handle proteins as single, rigid structures. While the importance of conformational flexibility is widely known, handling it remains challenging. Even the crystal structure of a protein usually contains variability exemplified in alternate side chain orientations or backbone segments. This conformational variability is encoded in PDB structure files by so-called alternate locations (AltLocs). Most modeling approaches either ignore AltLocs or resolve them with simple heuristics early on during structure import. We analyzed the occurrence and usage of AltLocs in the PDB and developed an algorithm to automatically handle AltLocs in PDB files enabling all structure-based methods using rigid structures to take the alternative protein conformations described by AltLocs into consideration. A respective software tool named AltLocEnumerator can be used as a structure preprocessor to easily exploit AltLocs. While the amount of data makes it difficult to show impact on a statistical level, handling AltLocs has a substantial impact on a case-by-case basis. We believe that the inspection and consideration of AltLocs is a very valuable approach in many modeling scenarios.


Assuntos
Proteínas , Software , Raios X , Proteínas/química , Conformação Proteica , Algoritmos
11.
J Chem Inf Model ; 63(21): 6587-6597, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37910814

RESUMO

Synthesizability is essential for compounds designed in silico. Regardless, synthetic accessibility is often considered only as an afterthought in the design and optimization process. In addition, the trend with modern computer-aided drug design methods is going toward full automation and away from the possibility of incorporating user knowledge. With this work, we present the second major release of our software tool, Synthesia, for synthesis-aware lead structure modification, where the user's expertise is now fully utilized. A provided retrosynthetic route is used as a pathway to guide structural modifications that introduce desired structural changes in the target compound. Moreover, the approach allows the user to define the exact position or component in the retrosynthetic route, which should be modified, further integrating the user's expert knowledge. This paper describes the functionality of Synthesia, its basic concepts, and several application scenarios ranging from simple examples to a comparison of the effects of the different exchange functions to an analysis of a set of bioisosteric linker structures, highlighting potential synthetically feasible replacements.


Assuntos
Desenho de Fármacos , Software , Automação
12.
J Comput Aided Mol Des ; 37(10): 491-503, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37515714

RESUMO

In this article, we present PoseEdit, a new, interactive frontend of the popular pose visualization tool PoseView. PoseEdit automatically produces high-quality 2D diagrams of intermolecular interactions in 3D binding sites calculated from ligands in complex with protein, DNA, and RNA. The PoseView diagrams have been improved in several aspects, most notably in their interactivity. Thanks to the easy-to-use 2D editor of PoseEdit, the diagrams are extensively editable and extendible by the user, can be merged with other diagrams, and even be created from scratch. A large variety of graphical objects in the diagram can be moved, rotated, selected and highlighted, mirrored, removed, or even newly added. Furthermore, PoseEdit enables a synchronized 2D-3D view of macromolecule-ligand complexes simplifying the analysis of structural features and interactions. The representation of individual diagram objects regarding their visualized chemical properties, like stereochemistry, and general graphical styles, like the color of interactions, can additionally be edited. The primary objective of PoseEdit is to support scientists with an enhanced way to communicate ligand binding mode information through graphical 2D representations optimized with the scientist's input in accordance with objective criteria and individual needs. PoseEdit is freely available on the ProteinsPlus web server ( https://proteins.plus ).


Assuntos
Proteínas , Software , Ligantes , Proteínas/química , Sítios de Ligação , Comunicação
13.
J Comput Aided Mol Des ; 37(1): 1-16, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36418668

RESUMO

Fragment spaces are an efficient way to model large chemical spaces using a handful of small fragments and a few connection rules. The development of Enamine's REAL Space has shown that large spaces of readily available compounds may be created this way. These are several orders of magnitude larger than previous libraries. So far, searching and navigating these spaces is mostly limited to topological approaches. A way to overcome this limitation is optimization via metaheuristics which can be combined with arbitrary scoring functions. Here we present Galileo, a novel Genetic Algorithm to sample fragment spaces. We showcase Galileo in combination with a novel pharmacophore mapping approach, called Phariety, enabling 3D searches in fragment spaces. We estimate the effectiveness of the approach with a small fragment space. Furthermore, we apply Galileo to two pharmacophore searches in the REAL Space, detecting hundreds of compounds fulfilling a HSP90 and a FXIa pharmacophore.


Assuntos
Desenho de Fármacos , Farmacóforo , Técnicas de Química Combinatória
14.
Proteins ; 90(8): 1521-1537, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35313380

RESUMO

Protein adaptations to extreme environmental conditions are drivers in biotechnological process optimization and essential to unravel the molecular limits of life. Most proteins with such desirable adaptations are found in extremophilic organisms inhabiting extreme environments. The deep sea is such an environment and a promising resource that poses multiple extremes on its inhabitants. Conditions like high hydrostatic pressure and high or low temperature are prevalent and many deep-sea organisms tolerate multiple of these extremes. While molecular adaptations to high temperature are comparatively good described, adaptations to other extremes like high pressure are not well-understood yet. To fully unravel the molecular mechanisms of individual adaptations it is probably necessary to disentangle multifactorial adaptations. In this study, we evaluate differences of protein structures from deep-sea organisms and their respective related proteins from nondeep-sea organisms. We created a data collection of 1281 experimental protein structures from 25 deep-sea organisms and paired them with orthologous proteins. We exhaustively evaluate differences between the protein pairs with machine learning and Shapley values to determine characteristic differences in sequence and structure. The results show a reasonable discrimination of deep-sea and nondeep-sea proteins from which we distinguish correlations previously attributed to thermal stability from other signals potentially describing adaptions to high pressure. While some distinct correlations can be observed the overall picture appears intricate.


Assuntos
Adaptação Fisiológica , Proteínas , Temperatura Baixa , Temperatura Alta , Pressão Hidrostática , Proteínas/metabolismo
15.
Bioinformatics ; 37(3): 424-425, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32735322

RESUMO

SUMMARY: The searching of user-defined 3D queries in molecular interfaces is a computationally challenging problem that is not satisfactorily solved so far. Most of the few existing tools focused on that purpose are desktop based and not openly available. Besides that, they show a lack of query versatility, search efficiency and user-friendliness. We address this issue with GeoMine, a publicly available web application that provides textual, numerical and geometrical search functionality for protein-ligand binding sites derived from structural data contained in the Protein Data Bank (PDB). The query generation is supported by a 3D representation of a start structure that provides interactively selectable elements like atoms, bonds and interactions. GeoMine gives full control over geometric variability in the query while performing a deterministic, precise search. Reasonably selective queries are processed on the entire set of protein-ligand complexes in the PDB within a few minutes. GeoMine offers an interactive and iterative search process of successive result analyses and query adaptations. From the numerous potential applications, we picked two from the field of side-effect analyze showcasing the usefulness of GeoMine. AVAILABILITY AND IMPLEMENTATION: GeoMine is part of the ProteinsPlus web application suite and freely available at https://proteins.plus. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Proteínas , Software , Sítios de Ligação , Bases de Dados de Proteínas , Ligantes , Ligação Proteica , Proteínas/metabolismo
16.
J Chem Inf Model ; 62(9): 2133-2150, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-34478299

RESUMO

Commercial make-on-demand compound spaces have become increasingly popular within the past few years. Since these libraries are too large for enumeration, they are usually accessed using combinatorial fragment space technologies like FTrees-FS and SpaceLight. Although both search types are of high practical impact, they lack the ability to search for precise structural features on the atomic level. To address this important use case, we developed SpaceMACS enabling efficient and precise maximum common induced substructure (MCIS) similarity and substructure searches within chemical fragment spaces. SpaceMACS enumerates a user-defined number of compounds in a multistep procedure. First, substructures of the query are extracted and matched to all fragments of the space. Then partial results are combined to actual compounds of the space. In this way, SpaceMACS identifies common substructures even if they cross fragment borders. We applied SpaceMACS on three commercial fragment spaces searching for the 150 000 most similar analogs to a glucosyltransferase binder from literature. We were able to find almost all building blocks used for the synthesis of the 90 listed analogs and a plethora of additional results. SpaceMACS is the missing link to enable rational drug discovery on make-on-demand combinatorial catalogs. No matter whether initial compound suggestions come from a de novo design, an AI-based compound generation, or a medicinal chemist's drawing board, the method gives access to the structurally closest chemically available analogs in seconds to at most minutes.


Assuntos
Descoberta de Drogas , Descoberta de Drogas/métodos
17.
J Chem Inf Model ; 62(11): 2800-2810, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35653228

RESUMO

The distributions of physicochemical property values, like the octanol-water partition coefficient, are routinely calculated to describe and compare virtual chemical libraries. Traditionally, these distributions are derived by processing each member of a library individually and summarizing all values in a distribution. This process becomes impractical when operating on chemical spaces which surpass billions of compounds in size. In this work, we present a novel algorithmic method called SpaceProp for the property distribution calculation of large nonenumerable combinatorial fragment spaces. The novel method follows a combinatorial approach and is able to calculate physicochemical property distributions of prominent spaces like Enamine's REAL Space, WuXi's GalaXi Space, and OTAVA's CHEMriya Space for the first time. Furthermore, we present a first approach of optimizing property distributions directly in combinatorial fragment spaces.


Assuntos
Técnicas de Química Combinatória , Bibliotecas de Moléculas Pequenas
18.
J Chem Inf Model ; 62(15): 3565-3576, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35867908

RESUMO

In modern drug design, one of the main issues is the optimization of an initial lead structure toward a drug candidate by modifying specific properties in the desired direction. The synthetic feasibility of the target structure is often neglected during this process, resulting in structures with low or suboptimal synthetic accessibility. In this work, we present a novel approach for synthesis-aware lead optimization called Synthesia. In contrast to the traditional approaches, Synthesia integrates the preservation of the synthesizability of the target structure into the lead structure modification process. Synthesia is able to create structural diversity for a lead structure that matches user-defined molecular properties without losing the applicability of a particular synthetic pathway. The methodology is validated by demonstrating that Synthesia is capable of providing structural analogues of DrugBank compounds that meet generic modification goals and maintain their synthetic pathways. In addition, Synthesia is used to cluster compounds from two different patent structure series (CDK7, Daurismo) according to their compatibility with the same synthetic pathways, maximizing the synthetic efficiency and providing an initial estimation of the effort of synthesizing the entire series. Altogether, we demonstrate Synthesia's ability to modify compound properties while maintaining in silico synthesizability.


Assuntos
Desenho de Fármacos
19.
J Chem Inf Model ; 62(19): 4680-4689, 2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36169383

RESUMO

Reaction schemes for organic molecules play a crucial role in modern in silico drug design processes. In contrast to the classical drawn reaction diagrams, computational chemists prefer SMARTS based line notations due to a substantially increased expressiveness and precision. They are used to search databases, calculate synthesizability, generate new molecules, or simulate novel reactions. Working with computer-readable representations of reaction schemes can be challenging due to the complexity of the features to be represented. Line representations of reaction schemes can often be cryptic, even to experienced users. To simplify the work with Reaction SMARTS for synthetic, computational, and medicinal chemists, we introduce a visualization technique for reaction schemes and provide a respective tool, called ReactionViewer. ReactionViewer is able to convert reaction schemes encoded as Reaction SMILES, Reaction SMARTS, or SMIRKS into a visual representation. The visualization technique is based on the concept of structure diagrams and follows IUPAC's "Compendium of Chemical Terminology" definition of chemical reaction equations for the reaction symbols. We demonstrate the applicability of the method using two data sets of organic synthesis reaction schemes taken from recent publications. We discuss various properties of the visualization and highlight its readability and interpretability.


Assuntos
Computadores , Técnicas de Química Sintética , Bases de Dados Factuais
20.
J Chem Inf Model ; 62(3): 553-566, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35050621

RESUMO

The set of chemical compounds shared by two or more chemical libraries is assessed routinely as means of comparing these libraries for various applications. Traditionally this is achieved by comparing the members of the chemical libraries individually for identity. This approach becomes impractical when operating on chemical libraries exceeding billions or even trillions of compounds in size. As a result, no such analysis exists for ultralarge chemical spaces like the Enamine REAL Space containing over 20 billion compounds. In this work, we present a novel tool called SpaceCompare for the overlap calculation of large, nonenumerable combinatorial fragment spaces. In contrast to existing methods, SpaceCompare utilizes topological fingerprints and the combinatorial character of these chemical spaces. The tool is able to determine the exact overlap of prominent spaces like Enamine's REAL Space, WuXi's GalaXi Space, and Otava's CHEMriya for the first time.


Assuntos
Técnicas de Química Combinatória , Bibliotecas de Moléculas Pequenas , Bibliotecas de Moléculas Pequenas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA