Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Anal Chem ; 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38319243

ABSTRACT

Large immune complexes formed by the cross-linking of antibodies with polyvalent antigens play critical roles in modulating cell-mediated immunity. While both the size and the shape of immune complexes are important determinants in Fc receptor-mediated signaling responsible for phagocytosis, degranulation, and, in some instances, autoimmune pathologies, their characterization remains extremely challenging due to their large size and structural heterogeneity. We use native mass spectrometry (MS) supplemented with limited charge reduction in the gas phase to determine the stoichiometry of immune complexes formed by a bivalent (homodimeric) antigen, a 163 kDa aminopeptidase P2 (APP2), and a monoclonal antibody (mAb) to APP2. The observed (APP2·mAb)n complexes populate a wide range of stoichiometries (n = 1-4) with the largest detected species exceeding 1 MDa, although the gas-phase dissociation products are also evident in the mass spectra. While frequently considering a nuisance that complicates interpretation of native MS data, limited dissociation provides an additional dimension for characterization of the immune complex quaternary structure. APP2/mAb associations with identical composition but slightly different elution times in size exclusion chromatography exhibit notable differences in their spontaneous fragmentation profiles. The latter indicates the presence of both extended linear and cyclized (APP2·mAb)n configurations. The unique ability of MS to distinguish between such isomeric structures will be invaluable for a variety of applications where the biological effects of immune complexes are determined by their ability to assemble Fc receptor clusters of certain density on cell surfaces, such as platelet activation by clustering the low-affinity receptors FcγRIIa on their surface.

2.
Chem Sci ; 14(11): 2901-2909, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36937588

ABSTRACT

Although metallacycle-based photosensitizers have attracted increasing attention in biomedicine, their clinical application has been hindered by their inherent dark toxicity and unsatisfactory phototherapeutic efficiency. Herein, we employ a π-expansion strategy for ruthenium acceptors to develop a series of Ru(ii) metallacycles (Ru1-Ru4), while simultaneously reducing dark toxicity and enhancing phototoxicity, thus obtaining a high phototoxicity index (PI). These metallacycles enable deep-tissue (∼7 mm) fluorescence imaging and reactive oxygen species (ROS) production and exhibit remarkable anti-tumor activity even under hypoxic conditions. Notably, Ru4 has the lowest dark toxicity, highest ROS generation ability and an optimal PI (∼146). Theoretical calculations verify that Ru4 exhibits the largest steric bulk and the lowest singlet-triplet energy gap (ΔE ST, 0.62 eV). In vivo studies confirm that Ru4 allows for effective and safe phototherapy against A549 tumors. This work thus is expected to open a new avenue for the design of high-performance metal-based photosensitizers for potential clinical applications.

3.
PLoS One ; 17(11): e0276462, 2022.
Article in English | MEDLINE | ID: mdl-36413536

ABSTRACT

The long-sought-after "magic bullet" in systemic therapy remains unrealized for disease targets existing inside most tissues, theoretically because vascular endothelium impedes passive tissue entry and full target engagement. We engineered the first "dual precision" bispecific antibody with one arm pair to precisely bind to lung endothelium and drive active delivery and the other to precisely block TGF-ß effector function inside lung tissue. Targeting caveolae for transendothelial pumping proved essential for delivering most of the injected intravenous dose precisely into lungs within one hour and for enhancing therapeutic potency by >1000-fold in a rat pneumonitis model. Ultra-low doses (µg/kg) inhibited inflammatory cell infiltration, edema, lung tissue damage, disease biomarker expression and TGF-ß signaling. The prodigious benefit of active vs passive transvascular delivery of a precision therapeutic unveils a new promising drug design, delivery and therapy paradigm ripe for expansion and clinical testing.


Subject(s)
Antibodies, Bispecific , Caveolae , Rats , Animals , Caveolae/metabolism , Transforming Growth Factor beta/metabolism , Antibodies, Bispecific/metabolism , Lung/metabolism , Endothelium, Vascular/metabolism
4.
Med Chem Res ; 29(4): 607-616, 2020 Apr.
Article in English | MEDLINE | ID: mdl-34552311

ABSTRACT

The orphan nuclear receptors estrogen-related receptors (ERRs) bind to the estrogen-related receptor response element (ERRE) to regulate transcriptional programs in cellular metabolism and cancer cell growth. In this study, we evaluated the potential for a pyrrole-imidazole polyamide to block ERRα binding to ERREs to inhibit gene expression. We demonstrated that the ERRE-targeted polyamide 1 blocked the binding of ERRα to the consensus ERRE and reduced the transcriptional activity of ERRα in cell culture. We further showed that inhibiting ERRα transcriptional activity with polyamide 1 led to reduced mitochondrial oxygen consumption, a primary biological effect regulated by ERRα. Finally, our data demonstrated that polyamide 1 is an inhibitor for cancer cell growth.

5.
J Pharm Anal ; 8(3): 153-159, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29922483

ABSTRACT

A high-performance liquid chromatography-electrospray ionization tandem mass spectrometric (HPLC-ESI-MS/MS) method was developed for the quantification of MHI148-clorgyline amide (NMI-amide), a novel tumor-targeting monoamine oxidase A inhibitor, in mouse plasma. The method was validated in terms of sensitivity, precision, accuracy, recovery and stability and then applied to a pharmacokinetic study of NMI-amide in mice following intravenous administration. NMI-amide together with the internal standard (IS), MHI-148, was extracted by protein precipitation using acetonitrile. Multiple reaction monitoring was used for quantification of NMI-amide by detecting m/z transition of 491.2-361.9, and 685.3-258.2 for NMI-amide and the IS, respectively. The lower limit of quantification (LLOQ) of the HPLC-MS/MS method for NMI-amide was 0.005 µg/mL and the linear calibration curve was acquired with R2 > 0.99 in the concentration range of 0.005-2 µg/mL. The intra- and inter-day precisions of the assay were assessed by percentage of the coefficient of variations, which was within 9.8% at LLOQ and 14.0% for other quality control samples, whereas the mean accuracy ranged from 86.8% to 113.2%. The samples were stable under storage and experimental conditions. This method was successfully applied to a pharmacokinetic study in mice following intravenous administration of 5 mg/kg NMI-amide.

6.
Oncotarget ; 7(12): 13842-53, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26871599

ABSTRACT

Glioblastoma (GBM) is an aggressive brain tumor which is currently treated with temozolomide (TMZ). Tumors usually become resistant to TMZ and recur; no effective therapy is then available. Monoamine Oxidase A (MAO A) oxidizes monoamine neurotransmitters resulting in reactive oxygen species which cause cancer. This study shows that MAO A expression is increased in human glioma tissues and cell lines. MAO A inhibitors, clorgyline or the near-infrared-dye MHI-148 conjugated to clorgyline (NMI), were cytotoxic for glioma and decreased invasion in vitro. Using the intracranial TMZ-resistant glioma model, clorgyline or NMI alone or in combination with low-dose TMZ reduced tumor growth and increased animal survival. NMI was localized specifically to the tumor. Immunocytochemistry studies showed that the MAO A inhibitor reduced proliferation, microvessel density and invasion, and increased macrophage infiltration. In conclusion, we have identified MAO A inhibitors as potential novel stand-alone drugs or as combination therapy with low dose TMZ for drug-resistant gliomas. NMI can also be used as a non-invasive imaging tool. Thus has a dual function for both therapy and diagnosis.


Subject(s)
Brain Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Glioma/pathology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/chemistry , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Disease Progression , Glioma/drug therapy , Glioma/metabolism , Humans , Male , Mice , Mice, Nude , Temozolomide , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 112(35): 10950-5, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26269569

ABSTRACT

Age-related macular degeneration (AMD) is associated with dysfunction and death of retinal pigment epithelial (RPE) cells. Cell-based approaches using RPE-like cells derived from human pluripotent stem cells (hPSCs) are being developed for AMD treatment. However, most efficient RPE differentiation protocols rely on complex, stepwise treatments and addition of growth factors, whereas small-molecule-only approaches developed to date display reduced yields. To identify new compounds that promote RPE differentiation, we developed and performed a high-throughput quantitative PCR screen complemented by a novel orthogonal human induced pluripotent stem cell (hiPSC)-based RPE reporter assay. Chetomin, an inhibitor of hypoxia-inducible factors, was found to strongly increase RPE differentiation; combination with nicotinamide resulted in conversion of over one-half of the differentiating cells into RPE. Single passage of the whole culture yielded a highly pure hPSC-RPE cell population that displayed many of the morphological, molecular, and functional characteristics of native RPE.


Subject(s)
Cell Differentiation/drug effects , Pluripotent Stem Cells/drug effects , Retinal Pigment Epithelium/cytology , High-Throughput Screening Assays , Humans , Pluripotent Stem Cells/cytology , Polymerase Chain Reaction
8.
J Am Chem Soc ; 137(6): 2366-74, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25585152

ABSTRACT

Development of anti-cancer agents with high tumor-targeting specificity and efficacy is critical for modern multidisciplinary cancer research. Monoamine oxidase A (MAOA), a mitochondria-bound enzyme, degrades monoamine neurotransmitters and dietary monoamines. Recent evidence suggests a correlation between increased MAOA expression and prostate cancer (PCa) progression with poor outcomes for patients. MAOA induces epithelial-mesenchymal transition (EMT) and augments hypoxic effects by producing excess reactive oxygen species. Thus, development of MAOA inhibitors which selectively target tumors becomes an important goal in cancer pharmacology. Here we describe the design, synthesis, and in vitro and in vivo evaluation of NMI, a conjugate that combines a near-infrared dye for tumor targeting with the moiety derived from the MAOA inhibitor clorgyline. NMI inhibits MAOA with low micromolar IC50, suppresses PCa cell proliferation and colony formation, and reduces migration and invasion. In mouse PCa xenografts, NMI targets tumors with no detectable accumulation in normal tissues, providing effective reduction of the tumor burden. Analysis of tumor specimens shows reduction in Ki-67(+) and CD31(+) cells, suggesting a decrease of cell proliferation and angiogenesis and an increase in M30(+) cells, indicating increased apoptosis. Gene expression profiles of tumors treated with NMI demonstrate reduced expression of oncogenes FOS, JUN, NFKB, and MYC and cell cycle regulators CCND1, CCNE1, and CDK4/6, along with increases in the levels of tumor suppressor gene TP53, cell cycle inhibitors CDKN1A and CDKN2A, and MAOA-downstream genes that promote EMT, tumor hypoxia, cancer cell migration, and invasion. These data suggest that NMI exerts its effect through tumor-targeted delivery of a MAOA-inactivating group, making NMI a valuable anti-tumor agent.


Subject(s)
Cell Proliferation/drug effects , Coloring Agents/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/drug effects , Prostatic Neoplasms/pathology , Animals , Male , Mice , Mice, Nude , Spectroscopy, Near-Infrared
9.
Bioconjug Chem ; 26(1): 78-89, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25350602

ABSTRACT

We report in vitro and in vivo evaluation of a newly designed trifunctional theranostic agent for targeting solid tumors. This agent combines a dendritic wedge with high boron content for boron neutron capture therapy or boron MRI, a monomethine cyanine dye for visible-light fluorescent imaging, and an integrin ligand for efficient tumor targeting. We report photophysical properties of the new agent, its cellular uptake and in vitro targeting properties. Using live animal imaging and intravital microscopy (IVM) techniques, we observed a rapid accumulation of the agent and its retention for a prolonged period of time (up to 7 days) in fully established animal models of human melanoma and murine mammary adenocarcinoma. This macromolecular theranostic agent can be used for targeted delivery of high boron load into solid tumors for future applications in boron neutron capture therapy.


Subject(s)
Dendrimers/therapeutic use , Neoplasms/diagnosis , Neoplasms/therapy , Animals , Biological Transport , Cell Line, Tumor , Cell Survival , Dendrimers/metabolism , Dendrimers/pharmacokinetics , Humans , Integrins/metabolism , Ligands , Mice , Neoplasms/pathology , Tissue Distribution
10.
Proc Natl Acad Sci U S A ; 111(52): 18448-53, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25516985

ABSTRACT

The development of novel antitumor agents that have high efficacy in suppressing tumor growth, have low toxicity to nontumor tissues, and exhibit rapid localization in the targeted tumor sites is an ongoing avenue of research at the interface of chemistry, cancer biology, and pharmacology. Supramolecular metal-based coordination complexes (SCCs) have well-defined shapes and geometries, and upon their internalization, SCCs could affect multiple oncogenic signaling pathways in cells and tissues. We investigated the uptake, intracellular localization, and antitumor activity of two rhomboidal Pt(II)-based SCCs. Laser-scanning confocal microscopy in A549 and HeLa cells was used to determine the uptake and localization of the assemblies within cells and their effect on tumor growth was investigated in mouse s.c. tumor xenograft models. The SCCs are soluble in cell culture media within the entire range of studied concentrations (1 nM-5 µM), are nontoxic, and showed efficacy in reducing the rate of tumor growth in s.c. mouse tumor xenografts. These properties reveal the potential of Pt(II)-based SCCs for future biomedical applications as therapeutic agents.


Subject(s)
Antineoplastic Agents , Neoplasms, Experimental/drug therapy , Organometallic Compounds , Platinum , Signal Transduction/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , HeLa Cells , Humans , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Platinum/chemistry , Platinum/pharmacology , Xenograft Model Antitumor Assays
11.
Proc Natl Acad Sci U S A ; 111(21): 7531-6, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821806

ABSTRACT

Development of small-molecule inhibitors of protein-protein interactions is a fundamental challenge at the interface of chemistry and cancer biology. Successful methods for design of protein-protein interaction inhibitors include computational and experimental high-throughput and fragment-based screening strategies to locate small-molecule fragments that bind protein surfaces. An alternative rational design approach seeks to mimic the orientation and disposition of critical binding residues at protein interfaces. We describe the design, synthesis, biochemical, and in vivo evaluation of a small-molecule scaffold that captures the topography of α-helices. We designed mimics of a key α-helical domain at the interface of hypoxia-inducible factor 1α and p300 to develop inhibitors of hypoxia-inducible signaling. The hypoxia-inducible factor/p300 interaction regulates the transcription of key genes, whose expression contributes to angiogenesis, metastasis, and altered energy metabolism in cancer. The designed compounds target the desired protein with high affinity and in a predetermined manner, with the optimal ligand providing effective reduction of tumor burden in experimental animal models.


Subject(s)
Biomimetics/methods , Drug Discovery/methods , Protein Interaction Domains and Motifs/drug effects , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Anaerobiosis , Animals , Blotting, Western , Cloning, Molecular , Gene Expression Profiling , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases , Mice , Molecular Sequence Data , Molecular Structure , Mutagenesis , Piperazine , Piperazines/chemistry , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
12.
J Med Chem ; 56(22): 9170-9, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24147900

ABSTRACT

Nitrogen mustards, widely used as chemotherapeutics, have limited safety and efficacy. Mitochondria lack a functional nucleotide excision repair mechanism to repair DNA adducts and are sensitive to alkylating agents. Importantly, cancer cells have higher intrinsic mitochondrial membrane potential (Δψmt) than normal cells. Therefore, selectively targeting nitrogen mustards to cancer cell mitochondria based on Δψmt could overcome those limitations. Herein, we describe the design, synthesis, and evaluation of Mito-Chlor, a triphenylphosphonium derivative of the nitrogen mustard chlorambucil. We show that Mito-Chlor localizes to cancer cell mitochondria where it acts on mtDNA to arrest cell cycle and induce cell death, resulting in a 80-fold enhancement of cell kill in a panel of breast and pancreatic cancer cell lines that are insensitive to the parent drug. Significantly, Mito-Chlor delayed tumor progression in a mouse xenograft model of human pancreatic cancer. This is a first example of repurposing chlorambucil, a drug not used in breast and pancreatic cancer treatment, as a novel drug candidate for these diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Chlorambucil/pharmacology , Mitochondria/drug effects , Pancreatic Neoplasms/pathology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Biological Transport , Cell Proliferation/drug effects , Chlorambucil/chemical synthesis , Chlorambucil/chemistry , Chlorambucil/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA, Mitochondrial/genetics , Drug Resistance, Neoplasm/drug effects , Histones/metabolism , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 110(39): 15602-7, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24019500

ABSTRACT

Selective blockade of gene expression by designed small molecules is a fundamental challenge at the interface of chemistry, biology, and medicine. Transcription factors have been among the most elusive targets in genetics and drug discovery, but the fields of chemical biology and genetics have evolved to a point where this task can be addressed. Herein we report the design, synthesis, and in vivo efficacy evaluation of a protein domain mimetic targeting the interaction of the p300/CBP coactivator with the transcription factor hypoxia-inducible factor-1α. Our results indicate that disrupting this interaction results in a rapid down-regulation of hypoxia-inducible genes critical for cancer progression. The observed effects were compound-specific and dose-dependent. Gene expression profiling with oligonucleotide microarrays revealed effective inhibition of hypoxia-inducible genes with relatively minimal perturbation of nontargeted signaling pathways. We observed remarkable efficacy of the compound HBS 1 in suppressing tumor growth in the fully established murine xenograft models of renal cell carcinoma of the clear cell type. Our results suggest that rationally designed synthetic mimics of protein subdomains that target the transcription factor-coactivator interfaces represent a unique approach for in vivo modulation of oncogenic signaling and arresting tumor growth.


Subject(s)
Hypoxia-Inducible Factor 1/chemistry , Hypoxia-Inducible Factor 1/metabolism , Peptides/pharmacology , Signal Transduction/drug effects , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Cell Hypoxia , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Profiling , HeLa Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Ligands , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemistry , Protein Binding/drug effects , Protein Multimerization , Protein Stability/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Response Elements/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/metabolism
14.
J Am Chem Soc ; 135(11): 4537-49, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23448368

ABSTRACT

Hypoxia is a hallmark of solid tumors, is associated with local invasion, metastatic spread, resistance to chemo- and radiotherapy, and is an independent, negative prognostic factor for a diverse range of malignant neoplasms. The cellular response to hypoxia is primarily mediated by a family of transcription factors, among which hypoxia-inducible factor 1 (HIF1) plays a major role. Under normoxia, the oxygen-sensitive α subunit of HIF1 is rapidly and constitutively degraded but is stabilized and accumulates under hypoxia. Upon nuclear translocation, HIF1 controls the expression of over 100 genes involved in angiogenesis, altered energy metabolism, antiapoptotic, and pro-proliferative mechanisms that promote tumor growth. A designed transcriptional antagonist, dimeric epidithiodiketopiperazine (ETP 2), selectively disrupts the interaction of HIF1α with p300/CBP coactivators and downregulates the expression of hypoxia-inducible genes. ETP 2 was synthesized via a novel homo-oxidative coupling of the aliphatic primary carbons of the dithioacetal precursor. It effectively inhibits HIF1-induced activation of VEGFA, LOX, Glut1, and c-Met genes in a panel of cell lines representing breast and lung cancers. We observed an outstanding antitumor efficacy of both (±)-ETP 2 and meso-ETP 2 in a fully established breast carcinoma model by intravital microscopy. Treatment with either form of ETP 2 (1 mg/kg) resulted in a rapid regression of tumor growth that lasted for up to 14 days. These results suggest that inhibition of HIF1 transcriptional activity by designed dimeric ETPs could offer an innovative approach to cancer therapy with the potential to overcome hypoxia-induced tumor growth and resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/drug therapy , Lung/drug effects , Piperazines/therapeutic use , p300-CBP Transcription Factors/metabolism , Animals , Antineoplastic Agents/chemistry , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Dimerization , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Models, Molecular , Piperazines/chemistry , Protein Interaction Maps/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Adv Drug Deliv Rev ; 65(1): 121-38, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23026636

ABSTRACT

Targeting of drugs and their carrier systems by using receptor-mediated endocytotic pathways was in its nascent stages 25 years ago. In the intervening years, an explosion of knowledge focused on design and synthesis of nanoparticulate delivery systems as well as elucidation of the cellular complexity of what was previously-termed receptor-mediated endocytosis has now created a situation when it has become possible to design and test the feasibility of delivery of highly specific nanoparticle drug carriers to specific cells and tissue. This review outlines the mechanisms governing the major modes of receptor-mediated endocytosis used in drug delivery and highlights recent approaches using these as targets for in vivo drug delivery of nanoparticles. The review also discusses some of the inherent complexity associated with the simple shift from a ligand-drug conjugate versus a ligand-nanoparticle conjugate, in terms of ligand valency and its relationship to the mode of receptor-mediated internalization.


Subject(s)
Drug Delivery Systems/methods , Endocytosis/drug effects , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Signal Transduction/drug effects , Animals , Humans , Ligands , Molecular Targeted Therapy
16.
Biopolymers ; 95(1): 8-16, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20882602

ABSTRACT

Designed small molecule inhibitors of hypoxia-inducible gene expression have potential to become new research tools for molecular biology, genetics and serve as leads to new therapeutics. We report design, synthesis evaluation of biological activity, and a preliminary mechanistic study of epipolythiodiketopiperazine (ETP) transcriptional antagonist that targets the interaction between the C-terminal transactivation domain (C-TAD) of hypoxia-inducible factor 1α (HIF-1α) and cysteine-histidine rich region (CH1) of transcriptional coactivator p300/CBP. Our results indicate that in cultured cells synthetic ETP 3 disrupts the structure and function of this complex in a dose-dependent manner, resulting in rapid downregulation of hypoxia-inducible gene expression.


Subject(s)
Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Piperazines/pharmacology , Amino Acid Sequence , Cell Line, Tumor , Cell Survival/drug effects , Humans , Molecular Sequence Data , Piperazines/chemical synthesis , Piperazines/chemistry
17.
Tetrahedron Lett ; 51(4): 609-612, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20161551

ABSTRACT

The indole-diketopiperazine bridge is an important structural feature of many bispyrrolidinoindoline and epipolythiodiketopiperazine fungal metabolites. Organocatalytic conjugate addition of diketopiperazines to indoles was achieved in good to excellent yields through electrophilic indolenine intermediates generated under mild conditions. Screening of catalysts and solvents at different temperatures was performed in order to achieve high product yields.

18.
J Am Chem Soc ; 132(3): 941-3, 2010 01 27.
Article in English | MEDLINE | ID: mdl-20041650

ABSTRACT

Designed ligands that inhibit hypoxia-inducible gene expression could offer new tools for genomic research and, potentially, drug discovery efforts for the treatment of neovascularization in cancers. We report a stabilized alpha-helix designed to target the binding interface between the C-terminal transactivation domain (C-TAD) of hypoxia-inducible factor 1alpha (HIF-1alpha) and cysteine-histidine rich region (CH1) of transcriptional coactivator CBP/p300. The synthetic helix disrupts the structure and function of this complex, resulting in a rapid downregulation of two hypoxia-inducible genes (VEGF and GLUT1) in cell culture.


Subject(s)
Disulfides/pharmacology , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Indole Alkaloids/pharmacology , Trans-Activators/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , HeLa Cells , Humans , Hydrogen Bonding , Hypoxia-Inducible Factor 1/chemistry , Hypoxia-Inducible Factor 1/metabolism , Protein Structure, Secondary , Trans-Activators/chemistry , Trans-Activators/metabolism
19.
J Am Chem Soc ; 131(50): 18078-88, 2009 Dec 23.
Article in English | MEDLINE | ID: mdl-20000859

ABSTRACT

Selective blockade of hypoxia-inducible gene expression by designed small molecules would prove valuable in suppressing tumor angiogenesis, metastasis and altered energy metabolism. We report the design, synthesis, and biological evaluation of a dimeric epidithiodiketopiperazine (ETP) small molecule transcriptional antagonist targeting the interaction of the p300/CBP coactivator with the transcription factor HIF-1alpha. Our results indicate that disrupting this interaction results in rapid downregulation of hypoxia-inducible genes critical for cancer progression. The observed effects are compound-specific and dose-dependent. Controlling gene expression with designed small molecules targeting the transcription factor-coactivator interface may represent a new approach for arresting tumor growth.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Diketopiperazines/pharmacology , Disulfides/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , p300-CBP Transcription Factors/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/toxicity , Binding, Competitive , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Diketopiperazines/chemical synthesis , Diketopiperazines/chemistry , Diketopiperazines/toxicity , Disulfides/chemical synthesis , Disulfides/chemistry , Disulfides/toxicity , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Expression Profiling , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases/genetics , Models, Molecular , Molecular Structure , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Oligonucleotide Array Sequence Analysis , Protein Binding , Vascular Endothelial Growth Factor A/biosynthesis , p300-CBP Transcription Factors/metabolism
20.
Acta Crystallogr C ; 65(Pt 8): o381-4, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19652319

ABSTRACT

A crystallographic investigation of the title compound, C22H28Cl2N4O4, using crystals obtained under different crystallization conditions, revealed the presence of two distinct polymorphic forms. The molecular conformation in the two polymorphs is very different: one adopts a 'C' shape, whereas the other adopts an 'S' shape. In the latter, the molecule lies across a crystallographic twofold axis. The 'S'-shaped polymorph undergoes a reversible orthorhombic-to-monoclinic phase transition on cooling, whereas the structure of the 'C'-shaped polymorph is temperature insensitive.


Subject(s)
Diketopiperazines/chemistry , Crystallization , Crystallography, X-Ray , Models, Molecular , Phase Transition , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...