Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Int J Mol Sci ; 21(6)2020 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-32188029

RESUMO

The ability to quantitatively probe diverse panels of proteins and their post-translational modifications (PTMs) across multiple samples would aid a broad spectrum of biological, biochemical and pharmacological studies. We report a novel, microarray analytical technology that combines immuno-affinity capture with Matrix Assisted Laser Desorption Ionization Mass Spectrometry (MALDI MS), which is capable of supporting highly multiplexed, targeted proteomic assays. Termed "Affinity-Bead Assisted Mass Spectrometry" (Affi-BAMS), this LC-free technology enables development of highly specific and customizable assay panels for simultaneous profiling of multiple proteins and PTMs. While affinity beads have been used previously in combination with MS, the Affi-BAMS workflow uses enrichment on a single bead that contains one type of antibody, generally capturing a single analyte (protein or PTM) while having enough binding capacity to enable quantification within approximately 3 orders of magnitude. The multiplexing capability is achieved by combining Affi-BAMS beads with different protein specificities. To enable screening of bead-captured analytes by MS, we further developed a novel method of performing spatially localized elution of targets from individual beads arrayed on a microscope slide. The resulting arrays of micro spots contain highly concentrated analytes localized within 0.5 mm diameter spots that can be directly measured using MALDI MS. While both intact proteins and protein fragments can be monitored by Affi-BAMS, we initially focused on applying this technology for bottom-up proteomics to enable screening of hundreds of samples per day by combining the robust magnetic bead-based workflow with the high throughput nature of MALDI MS acquisition. To demonstrate the variety of applications and robustness of Affi-BAMS, several studies are presented that focus on the response of 4EBP1, RPS6, ERK1/ERK2, mTOR, Histone H3 and C-MET to stimuli including rapamycin, H2O2, EPO, SU11274, Staurosporine and Vorinostat.


Assuntos
Análise em Microsséries/métodos , Proteômica/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Cromatina , Humanos , Peróxido de Hidrogênio , Isótopos , Peptídeo Hidrolases/química , Mutação Puntual , Processamento de Proteína Pós-Traducional , Proteínas/química , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Transdução de Sinais
2.
ACS Chem Biol ; 18(2): 296-303, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36602435

RESUMO

Lactic acid transport is a key process maintaining glycolytic flux in tumors. Inhibition of this process will result in glycolytic shutdown, impacting on cell growth and survival and thus has been pursued as a therapeutic approach for cancers. Using a cell-based screen in a MCT4-dependent cell line, we identified and optimized compounds for their ability to inhibit the efflux of intracellular lactic acid with good physical and pharmacokinetic properties. To deconvolute the mechanism of lactic acid efflux inhibition, we have developed three assays to measure cellular target engagement. Specifically, we synthesized a biologically active photoaffinity probe (IC50 < 10 nM), and using this probe, we demonstrated selective engagement of MCT4 of our parent molecule through a combination of confocal microscopy and in-cell chemoproteomics. As an orthogonal assay, the cellular thermal shift assay (CETSA) confirmed binding to MCT4 in the cellular system. Comparisons of lactic acid efflux potencies in cells with differential expression of MCT family members further confirmed that the optimized compounds inhibit the efflux of lactic acid through the inhibition of MCT4. Taken together, these data demonstrate the power of orthogonal chemical biology methods to determine cellular target engagement, particularly for proteins not readily amenable to traditional biophysical methods.


Assuntos
Biologia , Ácido Láctico , Ácido Láctico/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Proliferação de Células
3.
Mol Cancer Ther ; 22(5): 630-645, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-36912782

RESUMO

Antitumor immunity can be hampered by immunosuppressive mechanisms in the tumor microenvironment, including recruitment of arginase (ARG) expressing myeloid cells that deplete l-arginine essential for optimal T-cell and natural killer cell function. Hence, ARG inhibition can reverse immunosuppression enhancing antitumor immunity. We describe AZD0011, a novel peptidic boronic acid prodrug to deliver an orally available, highly potent, ARG inhibitor payload (AZD0011-PL). We demonstrate that AZD0011-PL is unable to permeate cells, suggesting that this compound will only inhibit extracellular ARG. In vivo, AZD0011 monotherapy leads to arginine increases, immune cell activation, and tumor growth inhibition in various syngeneic models. Antitumor responses increase when AZD0011 is combined with anti-PD-L1 treatment, correlating with increases in multiple tumor immune cell populations. We demonstrate a novel triple combination of AZD0011, anti-PD-L1, and anti-NKG2A, and combination benefits with type I IFN inducers, including polyI:C and radiotherapy. Our preclinical data demonstrate AZD0011's ability to reverse tumor immunosuppression and enhance immune stimulation and antitumor responses with diverse combination partners providing potential strategies to increase immuno-oncology therapies clinically.


Assuntos
Arginase , Linfócitos T , Humanos , Linhagem Celular Tumoral , Terapia de Imunossupressão , Tolerância Imunológica , Microambiente Tumoral
4.
Nat Commun ; 13(1): 4146, 2022 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-35842429

RESUMO

Enhancing the removal of aggregate-prone toxic proteins is a rational therapeutic strategy for a number of neurodegenerative diseases, especially Huntington's disease and various spinocerebellar ataxias. Ideally, such approaches should preferentially clear the mutant/misfolded species, while having minimal impact on the stability of wild-type/normally-folded proteins. Furthermore, activation of both ubiquitin-proteasome and autophagy-lysosome routes may be advantageous, as this would allow effective clearance of both monomeric and oligomeric species, the latter which are inaccessible to the proteasome. Here we find that compounds that activate the D1 ATPase activity of VCP/p97 fulfill these requirements. Such effects are seen with small molecule VCP activators like SMER28, which activate autophagosome biogenesis by enhancing interactions of PI3K complex components to increase PI(3)P production, and also accelerate VCP-dependent proteasomal clearance of such substrates. Thus, this mode of VCP activation may be a very attractive target for many neurodegenerative diseases.


Assuntos
Adenosina Trifosfatases , Doenças Neurodegenerativas , Proteína com Valosina , Adenosina Trifosfatases/metabolismo , Autofagia , Proteínas de Ciclo Celular/metabolismo , Humanos , Doenças Neurodegenerativas/genética , Fosfatos de Fosfatidilinositol , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína com Valosina/genética , Proteína com Valosina/metabolismo
5.
ACS Chem Biol ; 17(1): 54-67, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-34955012

RESUMO

Cyclin-dependent-kinases (CDKs) are members of the serine/threonine kinase family and are highly regulated by cyclins, a family of regulatory subunits that bind to CDKs. CDK9 represents one of the most studied examples of these transcriptional CDKs. CDK9 forms a heterodimeric complex with its regulatory subunit cyclins T1, T2 and K to form the positive transcription elongation factor b (P-TEFb). This complex regulates transcription via the phosphorylation of RNA polymerase II (RNAPolII) on Ser-2, facilitating promoter clearance and transcription elongation and thus remains an attractive therapeutic target. Herein, we have utilized classical affinity purification chemical proteomics, kinobeads assay, compressed CEllular Thermal Shift Assay (CETSA)-MS and Limited Proteolysis (LiP) to study the selectivity, target engagement and downstream mechanistic insights of a CDK9 tool compound. The above experiments highlight the value of quantitative mass spectrometry approaches to drug discovery, specifically proteome wide target identification and selectivity profiling. The approaches utilized in this study unanimously indicated that the CDK family of kinases are the main target of the compound of interest, with CDK9, showing the highest target affinity with remarkable consistency across approaches. We aim to provide guidance to the scientific community on the available chemical biology/proteomic tools to study advanced lead molecules and to highlight pros and cons of each technology while describing our findings in the context of the CDKs biology.


Assuntos
Quinase 9 Dependente de Ciclina/antagonistas & inibidores , Proteômica , Linhagem Celular Tumoral , Fracionamento Químico , Quinase 9 Dependente de Ciclina/genética , Quinase 9 Dependente de Ciclina/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Espectrometria de Massas
6.
Nat Chem Biol ; 5(11): 823-5, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19767736

RESUMO

The internal ribosome entry site (IRES), a highly conserved structured element of the hepatitis C virus (HCV) genomic RNA, is an attractive target for antiviral drugs. Here we show that benzimidazole inhibitors of the HCV replicon act by conformational induction of a widened interhelical angle in the IRES subdomain IIa, which facilitates the undocking of subdomain IIb from the ribosome and ultimately leads to inhibition of IRES-driven translation in HCV-infected cells.


Assuntos
Hepacivirus/metabolismo , Ribossomos/metabolismo , Antivirais/farmacologia , Antivirais/uso terapêutico , Sequência de Bases , Benzimidazóis/farmacologia , Citosina/química , Genoma Viral/efeitos dos fármacos , Genótipo , Hepacivirus/química , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Magnésio/farmacologia , Modelos Moleculares , Conformação de Ácido Nucleico , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Replicon/efeitos dos fármacos , Replicon/genética , Ribossomos/genética , Ribossomos/virologia , Transdução de Sinais
7.
Br J Pharmacol ; 177(8): 1709-1718, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32022252

RESUMO

Proteolysis-targeting chimeras are a new drug modality that exploits the endogenous ubiquitin proteasome system to degrade a protein of interest for therapeutic benefit. As the first-generation of proteolysis-targeting chimeras have now entered clinical trials for oncology indications, it is timely to consider the theoretical safety risks inherent with this modality which include off-target degradation, intracellular accumulation of natural substrates for the E3 ligases used in the ubiquitin proteasome system, proteasome saturation by ubiquitinated proteins, and liabilities associated with the "hook effect" of proteolysis-targeting chimeras This review describes in vitro and non-clinical in vivo data that provide mechanistic insight of these safety risks and approaches being used to mitigate these risks in the next generation of proteolysis-targeting chimera molecules to extend therapeutic applications beyond life-threatening diseases.


Assuntos
Quimera , Preparações Farmacêuticas , Quimera/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteólise , Ubiquitina-Proteína Ligases/metabolismo
8.
Cell Chem Biol ; 27(1): 41-46.e17, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31786184

RESUMO

Deregulation of the PRC2 complex, comprised of the core subunits EZH2, SUZ12, and EED, drives aberrant hypermethylation of H3K27 and tumorigenicity of many cancers. Although inhibitors of EZH2 have shown promising clinical activity, preclinical data suggest that resistance can be acquired through secondary mutations in EZH2 that abrogate drug target engagement. To address these limitations, we have designed several hetero-bifunctional PROTACs (proteolysis-targeting chimera) to efficiently target EED for elimination. Our PROTACs bind to EED (pKD ∼ 9.0) and promote ternary complex formation with the E3 ubiquitin ligase. The PROTACs potently inhibit PRC2 enzyme activity (pIC50 ∼ 8.1) and induce rapid degradation of not only EED but also EZH2 and SUZ12 within the PRC2 complex. Furthermore, the PROTACs selectively inhibit proliferation of PRC2-dependent cancer cells (half maximal growth inhibition [GI50] = 49-58 nM). In summary, our data demonstrate a therapeutic modality to target PRC2-dependent cancer through a PROTAC-mediated degradation mechanism.


Assuntos
Complexo Repressor Polycomb 2/metabolismo , Proteólise/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Estrutura Molecular , Complexo Repressor Polycomb 2/antagonistas & inibidores , Relação Estrutura-Atividade
9.
ACS Chem Biol ; 14(9): 1913-1920, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31329413

RESUMO

Demonstration of target binding is a key requirement for understanding the mode of action of new therapeutics. The cellular thermal shift assay (CETSA) has been introduced as a powerful label-free method to assess target engagement in physiological environments. Here, we present the application of live-cell CETSA to different classes of integral multipass transmembrane proteins using three case studies, the first showing a large and robust stabilization of the outer mitochondrial five-pass transmembrane protein TSPO, the second being a modest stabilization of SERCA2, and the last describing an atypical compound-driven stabilization of the GPCR PAR2. Our data demonstrated that using modified protocols with detergent extraction after the heating step, CETSA can reliably be applied to several membrane proteins of different complexity. By showing examples with distinct CETSA behaviors, we aim to provide the scientific community with an overview of different scenarios to expect during CETSA experiments, especially for challenging, membrane bound targets.


Assuntos
Receptor PAR-2/metabolismo , Receptores de GABA/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Aminoquinolinas/farmacologia , Benzamidas/farmacologia , Benzimidazóis/farmacologia , Benzodiazepinonas/farmacologia , Benzodioxóis/farmacologia , Álcoois Benzílicos/farmacologia , Bioensaio , Linhagem Celular Tumoral , Antagonistas GABAérgicos/farmacologia , Células HEK293 , Temperatura Alta , Humanos , Imidazóis/farmacologia , Transição de Fase/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Piridinas/farmacologia , Receptor PAR-2/antagonistas & inibidores , Receptor PAR-2/química , Receptores de GABA/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , Tapsigargina/farmacologia
10.
ACS Chem Biol ; 13(10): 2849-2854, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30216042

RESUMO

Targeting the protein-protein interaction between p53 and MDM2/MDMX (MDM4) represents an attractive anticancer strategy for the treatment of p53-competent tumors. Several selective and potent MDM2 inhibitors have been developed and entered the clinic; however, the repertoire of MDMX antagonists is still limited. The arylmethylidenepyrazolinone SJ-172550 has been reported as a selective MDMX antagonist; yet, uncertainties about its mechanism of action have raised doubts about its use as a chemical probe. Here, we show that, in addition to its unclear mode of action, SJ-172550 is unstable in aqueous buffers, giving rise to side products of unknown biological activity. Using an SJ-172550-derived affinity probe, we observed promiscuous binding to cellular proteins whereas cellular thermal shift assays did not reveal a stabilizing effect on MDMX. Overall, our results raise further questions about the interpretation of data using SJ-172550 and related compounds to investigate cellular phenotypes.


Assuntos
Acetatos/metabolismo , Inibidores Enzimáticos/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Pirazóis/metabolismo , Acetatos/química , Marcadores de Afinidade/química , Alcinos/química , Sítios de Ligação , Carbocianinas/química , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Química Click , Estabilidade de Medicamentos , Inibidores Enzimáticos/química , Humanos , Proteínas Nucleares/química , Ligação Proteica , Estabilidade Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/química , Pirazóis/química
11.
ACS Synth Biol ; 7(4): 1152-1162, 2018 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-29609459

RESUMO

Monoclonal antibody therapeutics have revolutionized the treatment of diseases such as cancer and autoimmune disorders, and also serve as research reagents for diverse and unparalleled applications. To extend their utility in both contexts, we have begun development of tunable antibodies, whose activity can be controlled by addition of a small molecule. Conceptually, we envision that incorporating cavity-forming mutations into an antibody can disrupt its structure, thereby reducing its affinity for antigen; addition of a small molecule may then restore the active structure, and thus rescue antigen binding. As a first proof of concept toward implementing this strategy, we have incorporated individual tryptophan to glycine mutations into FITC-E2, an anti-fluorescein single-chain variable fragment (scFv). We find that these can disrupt the protein structure and diminish antigen binding, and further that both structure and function can be rescued by addition of indole to complement the deleted side chain. While the magnitude of the affinity difference triggered by indole is modest in this first model system, it nonetheless provides a framework for future mutation/ligand pairs that may induce more dramatic responses. Disrupting and subsequently rescuing antibody activity, as exemplified by this first example, may represent a new approach to "design in" fine-tuned control of antibody activity for a variety of future applications.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Engenharia de Proteínas/métodos , Substituição de Aminoácidos , Anticorpos Monoclonais/genética , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Fluorescência , Glicina/genética , Indóis/química , Modelos Moleculares , Mutagênese Sítio-Dirigida/métodos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Relação Estrutura-Atividade , Triptofano/genética
12.
ACS Chem Biol ; 13(11): 3131-3141, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30335946

RESUMO

B-cell lymphoma 6 (BCL6) inhibition is a promising mechanism for treating hematological cancers but high quality chemical probes are necessary to evaluate its therapeutic potential. Here we report potent BCL6 inhibitors that demonstrate cellular target engagement and exhibit exquisite selectivity for BCL6 based on mass spectrometry analyses following chemical proteomic pull down. Importantly, a proteolysis-targeting chimera (PROTAC) was also developed and shown to significantly degrade BCL6 in a number of diffuse large B-cell lymphoma (DLBCL) cell lines, but neither BCL6 inhibition nor degradation selectively induced marked phenotypic response. To investigate, we monitored PROTAC directed BCL6 degradation in DLBCL OCI-Ly1 cells by immunofluorescence and discovered a residual BCL6 population. Analysis of subcellular fractions also showed incomplete BCL6 degradation in all fractions despite having measurable PROTAC concentrations, together providing a rationale for the weak antiproliferative response seen with both BCL6 inhibitor and degrader. In summary, we have developed potent and selective BCL6 inhibitors and a BCL6 PROTAC that effectively degraded BCL6, but both modalities failed to induce a significant phenotypic response in DLBCL despite achieving cellular concentrations.


Assuntos
Antineoplásicos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-6/antagonistas & inibidores , Quinolonas/farmacologia , Talidomida/análogos & derivados , Talidomida/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Ligantes , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Proteólise , Proteínas Proto-Oncogênicas c-bcl-6/química , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Quinolonas/síntese química , Quinolonas/metabolismo , Talidomida/síntese química , Talidomida/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
13.
Cell Chem Biol ; 25(6): 705-717.e11, 2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29628435

RESUMO

Activating KRAS mutations are major oncogenic drivers in multiple tumor types. Synthetic lethal screens have previously been used to identify targets critical for the survival of KRAS mutant cells, but their application to drug discovery has proven challenging, possibly due in part to a failure of monolayer cultures to model tumor biology. Here, we report the results of a high-throughput synthetic lethal screen for small molecules that selectively inhibit the growth of KRAS mutant cell lines in soft agar. Chemoproteomic profiling identifies the target of the most KRAS-selective chemical series as dihydroorotate dehydrogenase (DHODH). DHODH inhibition is shown to perturb multiple metabolic pathways. In vivo preclinical studies demonstrate strong antitumor activity upon DHODH inhibition in a pancreatic tumor xenograft model.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Pirimidinas/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Di-Hidro-Orotato Desidrogenase , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Camundongos , Camundongos SCID , Mutação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirimidinas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Células Tumorais Cultivadas
14.
ChemMedChem ; 12(12): 917-924, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28371485

RESUMO

Wnt signaling is critical for development, cell proliferation and differentiation, and mutations in this pathway resulting in constitutive signaling have been implicated in various cancers. A pathway screen using a Wnt-dependent reporter identified a chemical series based on a 1,2,3-thiadiazole-5-carboxamide (TDZ) core with sub-micromolar potency. Herein we report a comprehensive mechanism-of-action deconvolution study toward identifying the efficacy target(s) and biological implication of this chemical series involving bottom-up quantitative chemoproteomics, cell biology, and biochemical methods. Through observing the effects of our probes on metabolism and performing confirmatory cellular and biochemical assays, we found that this chemical series inhibits ATP synthesis by uncoupling the mitochondrial potential. Affinity chemoproteomics experiments identified sarco(endo)plasmic reticulum Ca2+ -dependent ATPase (SERCA2) as a binding partner of the TDZ series, and subsequent validation studies suggest that the TDZ series can act as ionophores through SERCA2 toward Wnt pathway inhibition.


Assuntos
Fosforilação Oxidativa/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Tiadiazóis/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Tiadiazóis/síntese química , Tiadiazóis/química
15.
Chem Commun (Camb) ; (29): 3045-62, 2006 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-16855685

RESUMO

Throughout history symmetry and chirality have inspired artists and scientists alike. Given that rotational axes are the only elements of symmetry compatible with chirality, it is not surprising that C2- and C3-symmetrical molecules have attracted considerable attention. In recent years, the aesthetic appeal of C2-symmetrical molecules has been translated into many widely-used applications some of which are of commercial importance by its exploitation in the area of asymmetric catalysis. In contrast, exploitation of the arguably greater aesthetic appeal of C3-symmetric molecules is still in its infancy. This review, which surveys the applications of chiral C3-symmetrical molecules in the areas of asymmetric catalysis, molecular recognition and nanoarchitecture, has been designed with a view to identifying some of the most promising areas of application of these very beautiful molecules.

19.
Org Lett ; 11(15): 3362-5, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19719184

RESUMO

The synthesis of spirocyclic oxindole pyran and oxepene frameworks using highly stereoselective Prins cyclizations of homoallylic and bis-homoallylic alcohols and isatin ketals is described.


Assuntos
Alcenos/síntese química , Indóis/síntese química , Oxepinas/síntese química , Piranos/síntese química , Silanos/síntese química , Alcenos/química , Ciclização , Indóis/química , Isatina/química , Oxepinas/química , Oxindóis , Propanóis/química , Piranos/química , Silanos/química , Compostos de Espiro/química , Estereoisomerismo
20.
Chemistry ; 12(1): 138-48, 2005 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-16278918

RESUMO

Chiral base chemistry has been used to create three chiral centres in one pot on a C3-symmetric substrate. The potential of this new approach to C3-symmetric molecules is exemplified by the creation of an enantiopure C3v-symmetric triol, triphosphane and tripyridine. A ruthenium complex of the last compound has been studied by X-ray crystallography.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA