Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 10(1): 40-55, 2022 01.
Article in English | MEDLINE | ID: mdl-34795032

ABSTRACT

Macrophages often abound within tumors, express colony-stimulating factor 1 receptor (CSF1R), and are linked to adverse patient survival. Drugs blocking CSF1R signaling have been used to suppress tumor-promoting macrophage responses; however, their mechanisms of action remain incompletely understood. Here, we assessed the lung tumor immune microenvironment in mice treated with BLZ945, a prototypical small-molecule CSF1R inhibitor, using single-cell RNA sequencing and mechanistic validation approaches. We showed that tumor control was not caused by CSF1R+ cell depletion; instead, CSF1R targeting reshaped the CSF1R+ cell landscape, which unlocked cross-talk between antitumoral CSF1R- cells. These cells included IFNγ-producing natural killer and T cells, and an IL12-producing dendritic cell subset, denoted as DC3, which were all necessary for CSF1R inhibitor-mediated lung tumor control. These data indicate that CSF1R targeting can activate a cardinal cross-talk between cells that are not macrophages and that are essential to mediate the effects of T cell-targeted immunotherapies and promote antitumor immunity.See related Spotlight by Burrello and de Visser, p. 4.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lung Neoplasms/therapy , Animals , Benzothiazoles/pharmacology , Cell Line, Tumor , Female , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Picolinic Acids/pharmacology , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays
2.
Sensors (Basel) ; 21(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069517

ABSTRACT

Microplastics (MPs) have been found in aqueous environments ranging from rural ponds and lakes to the deep ocean. Despite the ubiquity of MPs, our ability to characterize MPs in the environment is limited by the lack of technologies for rapidly and accurately identifying and quantifying MPs. Although standards exist for MP sample collection and preparation, methods of MP analysis vary considerably and produce data with a broad range of data content and quality. The need for extensive analysis-specific sample preparation in current technology approaches has hindered the emergence of a single technique which can operate on aqueous samples in the field, rather than on dried laboratory preparations. In this perspective, we consider MP measurement technologies with a focus on both their eventual field-deployability and their respective data products (e.g., MP particle count, size, and/or polymer type). We present preliminary demonstrations of several prospective MP measurement techniques, with an eye towards developing a solution or solutions that can transition from the laboratory to the field. Specifically, experimental results are presented from multiple prototype systems that measure various physical properties of MPs: pyrolysis-differential mobility spectroscopy, short-wave infrared imaging, aqueous Nile Red labeling and counting, acoustophoresis, ultrasound, impedance spectroscopy, and dielectrophoresis.

3.
ACS Med Chem Lett ; 11(11): 2312-2317, 2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33214846

ABSTRACT

Fibrosis is a common feature of several diseases, involves different organs, and results in significant morbidity and mortality. There are currently no effective therapies to halt the progression of fibrosis or reverse it. We have identified the highly water-soluble MMS-350, a novel bis-oxetanyl sulfoxide, as an antifibrotic agent. MMS-350 reduced the profibrotic phenotype induced in vitro in primary human fibroblasts and ameliorated bleomycin-induced pulmonary fibrosis in vivo. Furthermore, MMS-350 reversed fibrosis in human skin in organ culture. MMS-350 reduced levels of extracellular matrix proteins, the activation of fibroblasts, and the induction of pro-fibrotic factors. Similar effects at lower concentrations were observed with KRL507-031 and CL-613-091, two more lipophilic MMS-350 analogues. The fact that MMS-350 was effective at reducing pulmonary fibrosis induced by different triggers, the differential biological effects of its close structural analogues and its oral availability make it an attractive therapeutic candidate for organ fibrosis.

4.
Bioconjug Chem ; 31(9): 2191-2200, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32786373

ABSTRACT

A hybrid approach to covalently detachable molecules for nanoparticle capture and release from several custom-functionalized surfaces is described. This new surface chemistry capability provides a means for reversible binding of functionalized nanoparticles without relying on costly nucleic acid-based complexation. A new surface linker motif was devised wherein custom molecules were synthesized with components for surface anchoring, cleavage, and target capture through biotin-streptavidin binding. All capture-and-release chemistry is performed using physiological conditions (aqueous, pH 7). Covalent cleavage of linker molecules was achieved through incorporation of a tunable orthogonal reversible covalent (TORC) hydrazone functional group which underwent exchange with a competitive hydrazide aided by an aniline catalyst. The influence of the linker architecture on hydrazone exchange and nanoparticle release was probed by altering the distance between hydrazone and biotin groups using different length PEG spacers. Cleavable linkers were used to functionalize microwells, magnetic separation beads, and gold-coated glass surfaces. Upon functionalization, all surface types bound streptavidin and conjugated nanoparticles regardless of the linker structure. Conversely, the extent of hydrazone exchange as well as release of nanoparticles were influenced both by the hydrazone surface density and the linker molecular structure.


Subject(s)
Biotin/chemistry , Hydrazones/chemistry , Nanoparticles/chemistry , Streptavidin/chemistry , Gold/chemistry , Polyethylene Glycols/chemistry , Surface Properties
5.
Science ; 352(6282): 242-6, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26989197

ABSTRACT

Tumor-derived extracellular vesicles (tEVs) are important signals in tumor-host cell communication, yet it remains unclear how endogenously produced tEVs affect the host in different areas of the body. We combined imaging and genetic analysis to track melanoma-derived vesicles at organismal, cellular, and molecular scales to show that endogenous tEVs efficiently disseminate via lymphatics and preferentially bind subcapsular sinus (SCS) CD169(+) macrophages in tumor-draining lymph nodes (tdLNs) in mice and humans. The CD169(+) macrophage layer physically blocks tEV dissemination but is undermined during tumor progression and by therapeutic agents. A disrupted SCS macrophage barrier enables tEVs to enter the lymph node cortex, interact with B cells, and foster tumor-promoting humoral immunity. Thus, CD169(+) macrophages may act as tumor suppressors by containing tEV spread and ensuing cancer-enhancing immunity.


Subject(s)
B-Lymphocytes/immunology , Extracellular Vesicles/immunology , Immune Tolerance , Macrophages/immunology , Melanoma/immunology , Skin Neoplasms/immunology , Animals , B-Lymphocytes/ultrastructure , Cell Communication , Humans , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Macrophages/chemistry , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Sialic Acid Binding Ig-like Lectin 1/analysis , Sialic Acid Binding Ig-like Lectin 1/immunology , Skin Neoplasms/pathology
6.
Sci Transl Med ; 7(314): 314ra183, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26582898

ABSTRACT

Therapeutic nanoparticles (TNPs) have shown heterogeneous responses in human clinical trials, raising questions of whether imaging should be used to identify patients with a higher likelihood of NP accumulation and thus therapeutic response. Despite extensive debate about the enhanced permeability and retention (EPR) effect in tumors, it is increasingly clear that EPR is extremely variable; yet, little experimental data exist to predict the clinical utility of EPR and its influence on TNP efficacy. We hypothesized that a 30-nm magnetic NP (MNP) in clinical use could predict colocalization of TNPs by magnetic resonance imaging (MRI). To this end, we performed single-cell resolution imaging of fluorescently labeled MNPs and TNPs and studied their intratumoral distribution in mice. MNPs circulated in the tumor microvasculature and demonstrated sustained uptake into cells of the tumor microenvironment within minutes. MNPs could predictably demonstrate areas of colocalization for a model TNP, poly(d,l-lactic-co-glycolic acid)-b-polyethylene glycol (PLGA-PEG), within the tumor microenvironment with >85% accuracy and circulating within the microvasculature with >95% accuracy, despite their markedly different sizes and compositions. Computational analysis of NP transport enabled predictive modeling of TNP distribution based on imaging data and identified key parameters governing intratumoral NP accumulation and macrophage uptake. Finally, MRI accurately predicted initial treatment response and drug accumulation in a preclinical efficacy study using a paclitaxel-encapsulated NP in tumor-bearing mice. These approaches yield valuable insight into the in vivo kinetics of NP distribution and suggest that clinically relevant imaging modalities and agents can be used to select patients with high EPR for treatment with TNPs.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Ferrosoferric Oxide/metabolism , Fibrosarcoma/drug therapy , Magnetic Resonance Imaging/methods , Magnetics/methods , Nanomedicine/methods , Nanoparticles , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , Polyethylene Glycols/metabolism , Polyglactin 910/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Cell Line, Tumor , Chemistry, Pharmaceutical , DNA Damage , Disease Progression , Female , Ferrosoferric Oxide/chemistry , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/chemistry , Paclitaxel/metabolism , Particle Size , Polyethylene Glycols/chemistry , Polyglactin 910/chemistry , Predictive Value of Tests , Time Factors , Tissue Distribution , Tumor Microenvironment , Xenograft Model Antitumor Assays
7.
Sci Transl Med ; 6(261): 261ra152, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25378644

ABSTRACT

Eribulin mesylate was developed as a potent microtubule-targeting cytotoxic agent to treat taxane-resistant cancers, but recent clinical trials have shown that it eventually fails in many patient subpopulations for unclear reasons. To investigate its resistance mechanisms, we developed a fluorescent analog of eribulin with pharmacokinetic (PK) properties and cytotoxic activity across a human cell line panel that are sufficiently similar to the parent drug to study its cellular PK and tissue distribution. Using intravital imaging and automated tracking of cellular dynamics, we found that resistance to eribulin and the fluorescent analog depended directly on the multidrug resistance protein 1 (MDR1). Intravital imaging allowed for real-time analysis of in vivo PK in tumors that were engineered to be spatially heterogeneous for taxane resistance, whereby an MDR1-mApple fusion protein distinguished resistant cells fluorescently. In vivo, MDR1-mediated drug efflux and the three-dimensional tumor vascular architecture were discovered to be critical determinants of drug accumulation in tumor cells. We furthermore show that standard intravenous administration of a third-generation MDR1 inhibitor, HM30181, failed to rescue drug accumulation; however, the same MDR1 inhibitor encapsulated within a nanoparticle delivery system reversed the multidrug-resistant phenotype and potentiated the eribulin effect in vitro and in vivo in mice. Our work demonstrates that in vivo assessment of cellular PK of an anticancer drug is a powerful strategy for elucidating mechanisms of drug resistance in heterogeneous tumors and evaluating strategies to overcome this resistance.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzopyrans/administration & dosage , Drug Resistance, Neoplasm/drug effects , Furans/pharmacokinetics , Isoquinolines/administration & dosage , Ketones/pharmacokinetics , Neoplasms/drug therapy , Tetrazoles/administration & dosage , Tubulin Modulators/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Benzopyrans/chemistry , Cell Line , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Carriers , Female , Humans , Isoquinolines/chemistry , Mice, Nude , Microscopy, Confocal , Microscopy, Fluorescence , Nanoparticles , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/metabolism , Tetrazoles/chemistry , Transfection , Xenograft Model Antitumor Assays
8.
Bioconjug Chem ; 25(11): 2081-5, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25333750

ABSTRACT

Overexpression of anti-apoptotic proteins such as Bcl-2 is a cellular mechanism to evade apoptosis; consequently, Bcl-2 inhibitors are being developed as anticancer agents. In this work, we have synthesized a fluorescent version of ABT-199 in an effort to visualize a drug surrogate by high resolution imaging. We show that this fluorescent conjugate has comparable Bcl-2 binding efficacy and cell line potency to the parent compound and can be used as an imaging agent in several cancer cell types. We anticipate that this agent will be a valuable tool for studying the single-cell distribution and pharmacokinetics of ABT-199 as well the broader group of BH3-mimetics.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/metabolism , Fluorescent Dyes/metabolism , Molecular Imaging/methods , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/metabolism , Apoptosis , Biological Transport , Boron Compounds/chemistry , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Humans , Intracellular Space/metabolism , Models, Molecular , Protein Conformation , Proto-Oncogene Proteins c-bcl-2/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology
9.
In Vivo ; 28(4): 457-65, 2014.
Article in English | MEDLINE | ID: mdl-24982210

ABSTRACT

The ionizing irradiation mitigator MMS350 prolongs survival of mice treated with total-body irradiation and prevents radiation-induced pulmonary fibrosis when added to drinking water at day 100 after thoracic irradiation. The effects of MMS350 on hematopoiesis in long-term bone marrow culture and on the radiobiology of derived bone marrow stromal cell lines were tested. Long-term bone marrow cultures were established from C57BL/6NTac mice and maintained in a high-humidity incubator, with 7% CO2 and the addition of 100 µM MMS350 at the weekly media change. Over 10 weeks in culture, MMS350 had no significant effect on maintenance of hematopoietic stem cell production, or on nonadherent cells or colony-forming units of hematopoietic progenitor cells. Stromal cell lines derived from non MMS350-treated long-term cultures or control stromal cells treated with MMS350 were radioresistant in the clonogenic survival curve assay. MMS350 is a non-toxic, highly water-soluble radiation mitigator that exhibits radioprotective effects on bone marrow stromal cells.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Ethers, Cyclic/pharmacology , Hematopoiesis/drug effects , Radiation Tolerance/drug effects , Radiation-Protective Agents/pharmacology , Sulfoxides/pharmacology , Animals , Cell Line , Cell Survival , Colony-Forming Units Assay , Gene Expression Profiling , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Mice , Radiation Tolerance/genetics , Radiation, Ionizing , Stromal Cells , Transcription, Genetic
10.
Bioconjug Chem ; 25(6): 1137-42, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24806886

ABSTRACT

Cellular up-regulation of multidrug resistance protein 1 (MDR1) is a common cause for resistance to chemotherapy; development of third generation MDR1 inhibitors-several of which contain a common 6,7-dimethoxy-2-phenethyl-1,2,3,4-tetrahydroisoquinoline substructure-is underway. Efficacy of these agents has been difficult to ascertain, partly due to a lack of pharmacokinetic reporters for quantifying inhibitor localization and transport dynamics. Some of the recent third generation inhibitors have a pendant heterocycle, for example, a chromone moiety, which we hypothesized could be converted to a fluorophore. Following synthesis and teasing of a small set of analogues, we identified one lead compound that can be used as a cellular imaging agent that exhibits structural similarity and behavior akin to the latest generation of MDR1 inhibitors.


Subject(s)
Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Fluorescent Dyes/pharmacology , Tetrahydroisoquinolines/pharmacology , ortho-Aminobenzoates/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Fluorescent Dyes/chemistry , Humans , Models, Molecular , Molecular Structure , Paclitaxel/pharmacology , Structure-Activity Relationship , Tetrahydroisoquinolines/chemistry , ortho-Aminobenzoates/chemistry
11.
Radiat Res ; 180(5): 474-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24125487

ABSTRACT

A water-soluble ionizing radiation mitigator would have considerable advantages for the management of acute and chronic effects of ionizing radiation. We report that a novel oxetanyl sulfoxide (MMS350) is effective both as a protector and a mitigator of clonal mouse bone marrow stromal cell lines in vitro, and is an effective in vivo mitigator when administered 24 h after 9.5 Gy (LD100/30) total-body irradiation of C57BL/6NHsd mice, significantly improving survival (P = 0.0097). Furthermore, MMS350 (400 µM) added weekly to drinking water after 20 Gy thoracic irradiation significantly decreased: expression of pulmonary inflammatory and profibrotic gene transcripts and proteins; migration into the lungs of bone marrow origin luciferase+/GFP+ (luc+/GFP+) fibroblast progenitors (in both luc+ marrow chimeric and luc+ stromal cell line injected mouse models) and decreased radiation-induced pulmonary fibrosis (P < 0.0001). This nontoxic and orally administered small molecule may be an effective therapeutic in clinical radiotherapy and as a counter measure against the acute and chronic effects of ionizing radiation.


Subject(s)
Ethers, Cyclic/pharmacology , Lung/drug effects , Radiation Pneumonitis/drug therapy , Radiation-Protective Agents/administration & dosage , Safrole/analogs & derivatives , Sulfoxides/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Cell Line , Humans , Lung/radiation effects , Mice , Radiation Pneumonitis/pathology , Radiation, Ionizing , Safrole/administration & dosage , Water/chemistry , Whole-Body Irradiation
12.
Exp Hematol ; 41(11): 957-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23933481

ABSTRACT

We evaluated the use of colony formation (colony-forming unit-granulocyte macrophage [CFU-GM], burst-forming unit erythroid [BFU-E], and colony-forming unit-granulocyte-erythroid-megakaryocyte-monocytes [CFU-GEMM]) by human umbilical cord blood (CB) hematopoietic progenitor cells for testing novel small molecule ionizing irradiation protectors and mitigators. The following compounds were added before (protection) or after (mitigation) ionizing irradiation: GS-nitroxides (JP4-039 and XJB-5-131), the bifunctional sulfoxide MMS-350, the phosphoinositol-3-kinase inhibitor LY29400, triphenylphosphonium-imidazole fatty acid, the nitric oxide synthase inhibitor (MCF-201-89), the p53/mdm2/mdm4 inhibitor (BEB55), methoxamine, isoproterenol, propranolol, and the adenosine triphosphate-sensitive potassium channel blocker (glyburide). The drugs XJB-5-131, JP4-039, and MMS-350 were radiation protectors for CFU-GM. JP4-039 was also a radiation protector for CFU-GEMM. The drugs XJB-5-131, JP4-039, and MMS-350 were radiation mitigators for BFU-E, MMS-350 and JP4-039 were mitigators for CFU-GM, and MMS350 was a mitigator for CFU-GEMM. In contrast, other drugs were effective in murine assays; TTP-IOA, LY294002, MCF201-89, BEB55, propranolol, isoproterenol, methoxamine, and glyburide but showed no significant protection or mitigation in human CB assays. These data support the testing of new candidate clinical radiation protectors and mitigators using human CB clonogenic assays early in the drug discovery process, thus reducing the need for animal experiments.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Radiation, Ionizing , Radiation-Protective Agents/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , Colony-Forming Units Assay , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Radiation , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/radiation effects , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/radiation effects , Hematopoietic Stem Cells/cytology , Humans , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/radiation effects , Nitrogen Oxides/pharmacology , Safrole/analogs & derivatives , Safrole/pharmacology
13.
In Vivo ; 27(3): 291-7, 2013.
Article in English | MEDLINE | ID: mdl-23606683

ABSTRACT

BACKGROUND/AIM: Thoracic irradiation results in an acute inflammatory response, latent period, and late fibrosis. Little is known about the mechanisms involved in triggering late radiation fibrosis. MATERIALS AND METHODS: Thoracic irradiated fibrosis prone C57BL/6NTac mice were followed for detectable mRNA transcripts in isolated lung cells and micro-RNA in whole-tissues, and the effect of administration of water-soluble oxetanyl sulfoxide MMS350 was studied. Marrow stromal cell motility in medium from fibrotic-phase explanted pulmonary endothelial and alveolar type-II cells was measured. RESULTS: RNA and micro-RNA expression in lung correlated with fibrosis. MMS350 reduced pro-fibrotic gene expression in both endothelial and alveolar type-II cells in irradiated mice. Conditioned medium from irradiated cells did not alter cell motility in vitro. CONCLUSION: These studies should facilitate identification of potential new drug targets for ameliorating irradiation-induced pulmonary fibrosis.


Subject(s)
Alveolar Epithelial Cells/radiation effects , Ethers, Cyclic/metabolism , Pulmonary Alveoli/radiation effects , Pulmonary Fibrosis/etiology , Radiation Pneumonitis , Radiotherapy/adverse effects , Sulfoxides/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Cytokines/biosynthesis , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Radiation Pneumonitis/genetics , Radiation Pneumonitis/pathology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/radiation effects , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects
14.
Assay Drug Dev Technol ; 10(3): 269-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22192308

ABSTRACT

An oxetane-substituted sulfoxide has demonstrated potential as a dimethylsulfoxide substitute for enhancing the dissolution of organic compounds with poor aqueous solubilities. This sulfoxide may find utility in applications of library storage and biological assays. For the model compounds studied, significant solubility enhancements were observed using the sulfoxide as a cosolvent in aqueous media. Brine shrimp, breast cancer (MDA-MB-231), and liver cell line (HepG2) toxicity data for the new additive are also presented, in addition to comparative IC(50) values for a series of PKD1 inhibitors.


Subject(s)
Dimethyl Sulfoxide/chemistry , Organic Chemicals/chemistry , Water/chemistry , Animals , Cell Line , Dimethyl Sulfoxide/toxicity , Humans , Liver/drug effects , Organic Chemicals/toxicity , Solubility , Solvents/chemistry , Solvents/toxicity
15.
Tetrahedron ; 65(34): 6834-6839, 2009 Aug 22.
Article in English | MEDLINE | ID: mdl-20161364

ABSTRACT

The rapid synthesis of cis-2,6-disubstituted dihydropyrans is achieved in a three-component, one-pot cascade reaction. BiBr(3)-ediated addition of ketene silyl acetals or silyl enol ethers to beta,gamma-unsaturated cis-4-trimethylsilyl-3-butenal provides a Mukaiyama aldol adduct containing a vinylsilane moiety tethered to a silyl ether. Addition of a second aldehyde initiates a domino sequence involving intermolecular addition followed by an intramolecular silyl-modified Sakurai (ISMS) reaction. Isolated yields of this one-pot reaction vary from 44 to 80% and all compounds were isolated as the cis-diastereomers (10 examples).

SELECTION OF CITATIONS
SEARCH DETAIL
...