Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-34056626

ABSTRACT

The integration of salivary biomeasures in biobehavioral, psychophysiological, and clinical research has greatly expanded our ability to study the biopsychosocial processes underlying health. Much of this research, however, has failed to adequately assess and adjust for the impact of oral immune activity on salivary biomeasure concentrations and associations with serum levels. Aiming to improve the validity and reliability of salivary biomeasure data, we examine salivary total Immunoglobulin G (IgG) as a potential surrogate marker of oral inflammation and immune activity. During a single study visit in Baltimore, Maryland, healthy young adult participants provided matched blood and saliva samples (N=99; age 18-37 years, 42% female) and completed an oral health questionnaire. Biospecimens were assayed for total IgG and immune markers related to inflammation (cytokines), blood in saliva (transferrin), and tissue remodeling (matrix metalloproteinase-8). Total IgG (µg/mL) concentrations were higher in serum than saliva. Salivary total IgG was associated with some self-reported oral health measures, and strongly positively associated with all salivary immune markers. Controlling for salivary total IgG may be a feasible, affordable approach to adjusting salivary biomeasure findings for the influence of the oral immune environment when it is not possible or practical to obtain clinical oral health data.

2.
Biomark Med ; 12(6): 583-596, 2018 06.
Article in English | MEDLINE | ID: mdl-29873515

ABSTRACT

AIM: Serum uric acid (UA) is associated with many health conditions, including kidney, cardiovascular, and metabolic disorders. We examined the validity and stability of salivary UA as a noninvasive measure of serum UA. MATERIALS & METHODS: Using serum and salivary UA data from healthy adults (n = 99), we examined the UA serum-saliva correlation, and UA associations with adiponectin and C-reactive protein. Using longitudinal data from young adults (n = 182), we examined salivary UA stability. RESULTS: We found robust positive serum-saliva correlations for UA. UA and adiponectin were inversely related in serum and saliva. Salivary UA was relatively stable; 62-66% of variance could be attributed to a latent trait-like component. CONCLUSION: Salivary UA may be an important biomarker indexing health and disease risk.


Subject(s)
Saliva/chemistry , Uric Acid/analysis , Adolescent , Adult , Biomarkers/analysis , Biomarkers/blood , Body Mass Index , Female , Healthy Volunteers , Humans , Male , Reproducibility of Results , Uric Acid/blood , Young Adult
3.
Peptides ; 91: 58-64, 2017 05.
Article in English | MEDLINE | ID: mdl-28363793

ABSTRACT

This study addresses gaps in our understanding about the validity and utility of using salivary adiponectin to index serum adiponectin levels. Matched blood and saliva samples were collected on a single occasion from healthy adults (n=99; age 18-36 years, 53% male). Serum and saliva was assayed for adiponectin and inflammatory cytokines (IL-1ß, IL-6, IL-8, TNFα), and saliva was also assayed for markers of blood contamination (transferrin), total protein (salivary flow rate) and matrix metalloproteinase-8 (MMP-8). We examined the extent to which salivary adiponectin was associated with serum adiponectin, and the influence of potential confounders on the serum-saliva correlation, including age, sex, body mass index, and markers of inflammation, oral health, salivary blood contamination, and flow rate. Findings revealed a modest serum-saliva association for adiponectin, and strong positive associations between salivary adiponectin and salivary levels of inflammatory cytokines, MMP-8, transferrin, and total protein. By contrast, salivary adiponectin was not related to serum levels of inflammatory activity. The magnitude of the serum-saliva association was strengthened when controlling for total protein in saliva, blood leakage into oral fluid, salivary inflammatory cytokines, and MMP-8. The pattern of findings extends our understanding of salivary adiponectin and its potential use as an index of circulating adiponectin levels.


Subject(s)
Adiponectin/blood , Inflammation/immunology , Saliva/chemistry , Salivary Proteins and Peptides/analysis , Serum/chemistry , Adolescent , Adult , Age Factors , Biomarkers/analysis , Biomarkers/blood , Body Mass Index , Cytokines/blood , Female , Humans , Immunologic Factors/analysis , Male , Matrix Metalloproteinase 8/analysis , Oral Health , Sex Factors , Transferrin/analysis
4.
Oncotarget ; 7(13): 17103-10, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26959746

ABSTRACT

First line treatment for pancreatic cancer consists of surgical resection, if possible, and a subsequent course of chemotherapy using the nucleoside analogue gemcitabine. In some patients, an active transport mechanism allows gemcitabine to enter efficiently into the tumor cells, resulting in a significant clinical benefit. However, in most patients, low expression of gemcitabine transporters limits the efficacy of the drug to marginal levels, and patients need frequent administration of the drug at high doses, significantly increasing systemic drug toxicity. In this article we focus on a novel targeted delivery approach for gemcitabine consisting of conjugating the drug with an EphA2 targeting agent. We show that the EphA2 receptor is highly expressed in pancreatic cancers, and accordingly, the drug-conjugate is more effective than gemcitabine alone in targeting pancreatic tumors. Our preliminary observations suggest that this approach may provide a general benefit to pancreatic cancer patients and offers a comprehensive strategy for enhancing delivery of diverse therapeutic agents to a wide range of cancers overexpressing EphA2, thereby potentially reducing toxicity while enhancing therapeutic efficacy.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Molecular Targeted Therapy/methods , Pancreatic Neoplasms/drug therapy , Receptor, EphA2/metabolism , Animals , Cell Line, Tumor , Deoxycytidine/administration & dosage , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays , Gemcitabine
5.
Chem Biol ; 22(7): 876-887, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26165155

ABSTRACT

The development of novel, targeted delivery agents for anti-cancer therapies requires the design and optimization of potent and selective tumor-targeting agents that are stable and amenable to conjugation with chemotherapeutic drugs. While short peptides represent potentially an excellent platform for these purposes, they often get degraded and are eliminated too rapidly in vivo. In this study, we used a combination of nuclear magnetic resonance-guided structure-activity relationships along with biochemical and cellular studies to derive a novel tumor-homing agent, named 123B9, targeting the EphA2 tyrosine kinase receptor ligand-binding domain. Conjugating 123B9 to the chemotherapeutic drug paclitaxel (PTX) via a stable linker results in an agent that is significantly more effective than the unconjugated drug in both a pancreatic cancer xenograft model and a melanoma lung colonization and metastases model. Hence, 123B9 could represent a promising strategy for the development of novel targeted therapies for cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Paclitaxel/analogs & derivatives , Receptor, EphA2/agonists , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems/methods , Female , Humans , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Models, Animal , Molecular Targeted Therapy , Paclitaxel/chemistry , Paclitaxel/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Rats , Receptor, EphA2/chemistry , Xenograft Model Antitumor Assays
6.
ChemMedChem ; 9(7): 1403-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24677792

ABSTRACT

Because of its overexpression in a range of solid tumors, the EphA2 receptor is a validated target for cancer therapeutics. We recently described a new targeted delivery system based on specific EphA2-targeting peptides conjugated with the chemotherapeutic agent paclitaxel. Here, we investigate the chemical determinants responsible for the stability and degradation of these agents in plasma. Introducing modifications in both the peptide and the linker between the peptide and paclitaxel resulted in drug conjugates that are both long-lived in rat plasma and that markedly decrease tumor size in a prostate cancer xenograft model compared with paclitaxel alone treatment. These studies identify critical rate-limiting degradation sites on the peptide-drug conjugates, enabling the design of agents with increased stability and efficacy. These results provide support for our central hypothesis that peptide-drug conjugates targeting EphA2 represent an innovative and potentially effective strategy to selectively deliver cytotoxic drugs to cancer cells.


Subject(s)
Drug Carriers/chemical synthesis , Drug Design , Receptor, EphA2/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Female , Humans , Male , Mice , Mice, Inbred BALB C , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Peptides/chemical synthesis , Peptides/chemistry , Prostatic Neoplasms/drug therapy , Receptor, EphA2/chemistry , Transplantation, Heterologous
7.
Chem Biol ; 20(8): 973-82, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23891150

ABSTRACT

The E3 ubiquitin ligase Siah regulates key cellular events that are central to cancer development and progression. A promising route to Siah inhibition is disrupting its interactions with adaptor proteins. However, typical of protein-protein interactions, traditional unbiased approaches to ligand discovery did not produce viable hits against this target, despite considerable effort and a multitude of approaches. Ultimately, a rational structure-based design strategy was successful for the identification of Siah inhibitors in which peptide binding drives specific covalent bond formation with the target. X-ray crystallography, mass spectrometry, and functional data demonstrate that these peptide mimetics are efficient covalent inhibitors of Siah and antagonize Siah-dependent regulation of Erk and Hif signaling in the cell. The proposed strategy may result useful as a general approach to the design of peptide-based inhibitors of other protein-protein interactions.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Nuclear Proteins/antagonists & inhibitors , Peptides/chemistry , Peptidomimetics/chemistry , Ubiquitin-Protein Ligases/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptides/pharmacology , Peptidomimetics/pharmacology , Protein Interaction Maps/drug effects , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
8.
Clin Cancer Res ; 19(1): 128-37, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23155185

ABSTRACT

PURPOSE: YSA is an EphA2-targeting peptide that effectively delivers anticancer agents to prostate cancer tumors. Here, we report on how we increased the drug-like properties of this delivery system. EXPERIMENTAL DESIGN: By introducing non-natural amino acids, we have designed two new EphA2 targeting peptides: YNH, where norleucine and homoserine replace the two methionine residues of YSA, and dYNH, where a D-tyrosine replaces the L-tyrosine at the first position of the YNH peptide. We describe the details of the synthesis of YNH and dYNH paclitaxel conjugates (YNH-PTX and dYNH-PTX) and their characterization in cells and in vivo. RESULTS: dYNH-PTX showed improved stability in mouse serum and significantly reduced tumor size in a prostate cancer xenograft model and also reduced tumor vasculature in a syngeneic orthotopic allograft mouse model of renal cancer compared with vehicle or paclitaxel treatments. CONCLUSION: This study reveals that targeting EphA2 with dYNH drug conjugates could represent an effective way to deliver anticancer agents to a variety of tumor types.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems , Neoplasms/genetics , Paclitaxel/administration & dosage , Peptides , Receptor, EphA2/genetics , Animals , Antineoplastic Agents, Phytogenic/chemistry , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Paclitaxel/chemistry , Peptides/chemistry , Receptor, EphA2/metabolism , Transplantation, Homologous , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
ChemMedChem ; 7(12): 2227-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23139022

ABSTRACT

Successful replication of the influenza A virus requires both viral proteins and host cellular factors. In this study we used a cellular assay to screen for small molecules capable of interfering with any of such necessary viral or cellular components. We used an established reporter assay to assess influenza viral replication by monitoring the activity of co-expressed luciferase. We screened a diverse chemical compound library, resulting in the identification of compound 7, which inhibits a novel yet elusive target. Quantitative real-time PCR studies confirmed the dose-dependent inhibitory activity of compound 7 in a viral replication assay. Furthermore, we showed that compound 7 is effective in rescuing high-dose influenza infection in an in vivo mouse model. As oseltamivir-resistant influenza strains emerge, compound 7 could be further investigated as a new and potentially suitable scaffold for the development of anti-influenza agents that act on novel targets.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Virus Replication/drug effects , Animals , Antiviral Agents/pharmacology , Female , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/drug therapy , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/virology , Real-Time Polymerase Chain Reaction , Small Molecule Libraries/pharmacology , Tetrazoles/chemistry , Tetrazoles/pharmacology , Tetrazoles/therapeutic use
10.
Anticancer Agents Med Chem ; 12(9): 1143-55, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22931411

ABSTRACT

Structure-based modeling combined with rational drug design, and high throughput screening approaches offer significant potential for identifying and developing lead compounds with therapeutic potential. The present review focuses on these two approaches using explicit examples based on specific derivatives of Gossypol generated through rational design and applications of a cancer-specificpromoter derived from Progression Elevated Gene-3. The Gossypol derivative Sabutoclax (BI-97C1) displays potent anti-tumor activity against a diverse spectrum of human tumors. The model of the docked structure of Gossypol bound to Bcl-XL provided a virtual structure-activity-relationship where appropriate modifications were predicted on a rational basis. These structure-based studies led to the isolation of Sabutoclax, an optically pure isomer of Apogossypol displaying superior efficacy and reduced toxicity. These studies illustrate the power of combining structure-based modeling with rational design to predict appropriate derivatives of lead compounds to be empirically tested and evaluated for bioactivity. Another approach to cancer drug discovery utilizes a cancer-specific promoter as readouts of the transformed state. The promoter region of Progression Elevated Gene-3 is such a promoter with cancer-specific activity. The specificity of this promoter has been exploited as a means of constructing cancer terminator viruses that selectively kill cancer cells and as a systemic imaging modality that specifically visualizes in vivo cancer growth with no background from normal tissues. Screening of small molecule inhibitors that suppress the Progression Elevated Gene-3-promoter may provide relevant lead compounds for cancer therapy that can be combined with further structure-based approaches leading to the development of novel compounds for cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Drug Screening Assays, Antitumor/methods , Gossypol/analogs & derivatives , Gossypol/pharmacology , Neoplasms/drug therapy , Animals , Drug Screening Assays, Antitumor/economics , High-Throughput Screening Assays , Humans , Neoplasms/genetics , Promoter Regions, Genetic/drug effects
11.
Cancer Res ; 72(9): 2339-49, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22406620

ABSTRACT

Membrane type-1 matrix metalloproteinase (MT1-MMP) is a promising drug target in malignancy. The structure of MT1-MMP includes the hemopexin domain (PEX) that is distinct from and additional to the catalytic domain. Current MMP inhibitors target the conserved active site in the catalytic domain and, as a result, repress the proteolytic activity of multiple MMPs instead of MT1-MMP alone. In our search for noncatalytic inhibitors of MT1-MMP, we compared the protumorigenic activity of wild-type MT1-MMP with an MT1-MMP mutant lacking PEX (ΔPEX). In contrast to MT1-MMP, ΔPEX did not support tumor growth in vivo, and its expression resulted in small fibrotic tumors that contained increased levels of collagen. Because these findings suggested an important role for PEX in tumor growth, we carried out an inhibitor screen to identify small molecules targeting the PEX domain of MT1-MMP. Using the Developmental Therapeutics Program (National Cancer Institute/NIH), virtual ligand screening compound library as a source and the X-ray crystal structure of PEX as a target, we identified and validated a novel PEX inhibitor. Low dosage, intratumoral injections of PEX inhibitor repressed tumor growth and caused a fibrotic, ΔPEX-like tumor phenotype in vivo. Together, our findings provide a preclinical proof of principle rationale for the development of novel and selective MT1-MMP inhibitors that specifically target the PEX domain.


Subject(s)
Hemopexin/chemistry , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Female , Humans , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Protein Structure, Tertiary , Xenograft Model Antitumor Assays
12.
J Med Chem ; 55(5): 2427-36, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22329578

ABSTRACT

The efficacy of anticancer drugs is often limited by their systemic toxicities and adverse side effects. We report that the EphA2 receptor is overexpressed preferentially in several human cancer cell lines compared to normal tissues and that an EphA2 targeting peptide (YSAYPDSVPMMS) can be effective in delivering anticancer agents to such tumors. Hence, we report on the synthesis and characterizations of a novel EphA2-targeting agent conjugated with the chemotherapeutic drug paclitaxel. We found that the peptide-drug conjugate is dramatically more effective than paclitaxel alone at inhibiting tumor growth in a prostate cancer xenograft model, delivering significantly higher levels of drug to the tumor site. We believe these studies open the way to the development of a new class of therapeutic compounds that exploit the EphA2 receptor for drug delivery to cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Oligopeptides/chemical synthesis , Paclitaxel/administration & dosage , Receptor, EphA2/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems , Drug Screening Assays, Antitumor , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Oligopeptides/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , RNA, Messenger/metabolism , Receptor, EphA2/genetics , Transplantation, Heterologous
13.
J Med Chem ; 54(18): 6206-14, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21815634

ABSTRACT

c-Jun N-terminal kinases (JNKs) represent valuable targets in the development of new therapies. Present on the surface of JNK is a binding pocket for substrates and the scaffolding protein JIP1 in close proximity to the ATP binding pocket. We propose that bidentate compounds linking the binding energies of weakly interacting ATP and substrate mimetics could result in potent and selective JNK inhibitors. We describe here a bidentate molecule, 19, designed against JNK. 19 inhibits JNK kinase activity (IC(50) = 18 nM; K(i) = 1.5 nM) and JNK/substrate association in a displacement assay (IC(50) = 46 nM; K(i) = 2 nM). Our data demonstrate that 19 targets for the ATP and substrate-binding sites on JNK concurrently. Finally, compound 19 successfully inhibits JNK in a variety of cell-based experiments, as well as in vivo where it is shown to protect against Jo-2 induced liver damage and improve glucose tolerance in diabetic mice.


Subject(s)
Adenosine Triphosphate/chemistry , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Peptides/chemistry , Amino Acid Motifs , Animals , Binding, Competitive , Cell Line , Chemical and Drug Induced Liver Injury/prevention & control , Consensus Sequence , Cytokines/metabolism , Diabetes Mellitus, Type 2/drug therapy , Drug Design , Female , Glucose Intolerance/drug therapy , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Mimicry , Peptides/chemical synthesis , Peptides/pharmacology , Phosphorylation , Protein Binding , Structure-Activity Relationship , Substrate Specificity
14.
Expert Opin Investig Drugs ; 20(10): 1397-411, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21851287

ABSTRACT

INTRODUCTION: Human cancers are genetically and epigenetically heterogeneous and have the capacity to commandeer a variety of cellular processes to aid in their survival, growth and resistance to therapy. One strategy is to overexpress proteins that suppress apoptosis, such as the Bcl-2 family protein Mcl-1. The Mcl-1 protein plays a pivotal role in protecting cells from apoptosis and is overexpressed in a variety of human cancers. AREAS COVERED: Targeting Mcl-1 for extinction in these cancers, using genetic and pharmacological approaches, represents a potentially effectual means of developing new efficacious cancer therapeutics. Here we review the multiple strategies that have been employed in targeting this fundamental protein, as well as the significant potential these targeting agents provide in not only suppressing cancer growth, but also in reversing resistance to conventional cancer treatments. EXPERT OPINION: We discuss the potential issues that arise in targeting Mcl-1 and other Bcl-2 anti-apoptotic proteins, as well problems with acquired resistance. The application of combinatorial approaches that involve inhibiting Mcl-1 and manipulation of additional signaling pathways to enhance therapeutic outcomes is also highlighted. The ability to specifically inhibit key genetic/epigenetic elements and biochemical pathways that maintain the tumor state represent a viable approach for developing rationally based, effective cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Drug Design , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasms/metabolism , Neoplasms/physiopathology , Proto-Oncogene Proteins c-bcl-2/metabolism
15.
J Cell Physiol ; 226(10): 2484-93, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792905

ABSTRACT

Glutamate is an essential excitatory neurotransmitter regulating brain functions. Excitatory amino acid transporter (EAAT)-2 is one of the major glutamate transporters expressed predominantly in astroglial cells and is responsible for 90% of total glutamate uptake. Glutamate transporters tightly regulate glutamate concentration in the synaptic cleft. Dysfunction of EAAT2 and accumulation of excessive extracellular glutamate has been implicated in the development of several neurodegenerative diseases including Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis. Analysis of the 2.5 kb human EAAT2 promoter showed that NF-κB is an important regulator of EAAT2 expression in astrocytes. Screening of approximately 1,040 FDA-approved compounds and nutritionals led to the discovery that many ß-lactam antibiotics are transcriptional activators of EAAT2 resulting in increased EAAT2 protein levels. Treatment of animals with ceftriaxone (CEF), a ß-lactam antibiotic, led to an increase of EAAT2 expression and glutamate transport activity in the brain. CEF has neuroprotective effects in both in vitro and in vivo models based on its ability to inhibit neuronal cell death by preventing glutamate excitotoxicity. CEF increases EAAT2 transcription in primary human fetal astrocytes through the NF-κB signaling pathway. The NF-κB binding site at -272 position was critical in CEF-mediated EAAT2 protein induction. These studies emphasize the importance of transcriptional regulation in controlling glutamate levels in the brain. They also emphasize the potential utility of the EAAT2 promoter for developing both low and high throughput screening assays to identify novel small molecule regulators of glutamate transport with potential to ameliorate pathological changes occurring during and causing neurodegeneration.


Subject(s)
Excitatory Amino Acid Transporter 2/physiology , Glutamic Acid/physiology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Humans
16.
Proc Natl Acad Sci U S A ; 108(21): 8785-90, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555592

ABSTRACT

Limited options are available for treating patients with advanced prostate cancer (PC). Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), an IL-10 family cytokine, exhibits pleiotropic anticancer activities without adversely affecting normal cells. We previously demonstrated that suppression of the prosurvival Bcl-2 family member, myeloid cell leukemia-1 (Mcl-1), is required for mda-7/IL-24-mediated apoptosis of prostate carcinomas. Here we demonstrate that pharmacological inhibition of Mcl-1 expression with the unique Apogossypol derivative BI-97C1, also called Sabutoclax, is sufficient to sensitize prostate tumors to mda-7/IL-24-induced apoptosis, whereas ABT-737, which lacks efficacy in inhibiting Mcl-1, does not sensitize mda-7/IL-24-mediated cytotoxicity. A combination regimen of tropism-modified adenovirus delivered mda-7/IL-24 (Ad.5/3-mda-7) and BI-97C1 enhances cytotoxicity in human PC cells, including those resistant to mda-7/IL-24 or BI-97C1 alone. The combination regimen causes autophagy that facilitates NOXA- and Bim-induced and Bak/Bax-mediated mitochondrial apoptosis. Treatment with Ad.5/3-mda-7 and BI-97C1 significantly inhibits the growth of human PC xenografts in nude mice and spontaneously induced PC in Hi-myc transgenic mice. Tumor growth inhibition correlated with increased TUNEL staining and decreased Ki-67 expression in both PC xenografts and prostates of Hi-myc mice. These findings demonstrate that pharmacological inhibition of Mcl-1 with the Apogossypol derivative, BI-97C1, sensitizes human PCs to mda-7/IL-24-mediated cytotoxicity, thus potentially augmenting the therapeutic benefit of this combinatorial approach toward PC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Genetic Therapy/methods , Gossypol/analogs & derivatives , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Gossypol/pharmacology , Gossypol/therapeutic use , Humans , Interleukins/administration & dosage , Male , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Transfection , Xenograft Model Antitumor Assays
17.
Pigment Cell Melanoma Res ; 24(4): 703-13, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21592316

ABSTRACT

In melanoma, the activation of pro-survival signaling pathways, such as the AKT and NF-κB pathways, is critical for tumor growth. We have recently reported that the AKT inhibitor BI-69A11 causes efficient inhibition of melanoma growth. Here, we show that in addition to its AKT inhibitory activity, BI-69A11 also targets the NF-κB pathway. In melanoma cell lines, BI-69A11 inhibited TNF-α-stimulated IKKα/ß and IκB phosphorylation as well as NF-κB reporter gene expression. Furthermore, the effective inhibition of melanoma growth by BI-69A11 was attenuated upon NF-κB activation. Mechanistically, reduced NF-κB signaling by BI-69-A11 is mediated by the inhibition of sphingosine kinase 1, identified in a screen of 315 kinases. Significantly, we demonstrate that BI-69A11 is well tolerated and orally active against UACC 903 and SW1 melanoma xenografts. Our results demonstrate that BI-69A11 inhibits both the AKT and the NF-κB pathways and that the dual targeting of these pathways may be efficacious as a therapeutic strategy in melanoma.


Subject(s)
Benzimidazoles/therapeutic use , Melanoma/drug therapy , Molecular Targeted Therapy , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinolones/therapeutic use , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Administration, Oral , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Melanoma/enzymology , Mice , Mice, Nude , NF-kappa B/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinolones/administration & dosage , Quinolones/pharmacology , Skin Neoplasms/enzymology , Xenograft Model Antitumor Assays
18.
Bioorg Med Chem ; 19(8): 2582-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21458276

ABSTRACT

We report comprehensive structure-activity relationship studies on a novel series of c-Jun N-terminal kinase (JNK) inhibitors. Intriguingly, the compounds have a dual inhibitory activity by functioning as both ATP and JIP mimetics, possibly by binding to both the ATP binding site and to the docking site of the kinase. Several of such novel compounds display potent JNK inhibitory profiles both in vitro and in cell.


Subject(s)
JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Thiophenes/pharmacology , Adenosine Triphosphate , Binding Sites , Cell Line , Drug Design , Humans , Molecular Mimicry , Protein Binding , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , Thiophenes/chemistry
19.
Front Oncol ; 1: 28, 2011.
Article in English | MEDLINE | ID: mdl-22655238

ABSTRACT

Our focus in the past several years has been on the identification of novel and effective pan-Bcl-2 antagonists. We have recently reported a series of Apogossypolone (ApoG2) derivatives, resulting in the chiral compound (±) BI97D6. We report here the synthesis and evaluation on its optically pure (-) and (+) atropisomers. Compound (-) BI97D6 potently inhibits the binding of BH3 peptides to Bcl-X(L), Bcl-2, Mcl-1, and Bfl-1 with IC(50) values of 76 ± 5, 31 ± 2, 25 ± 8, and 122 ± 28 nM, respectively. In a cellular assay, compound (-) BI97D6 effectively inhibits cell growth in the PC-3 human prostate cancer and H23 human lung cancer cell lines with EC(50) values of 0.22 ± 0.08 and 0.14 ± 0.02 µM, respectively. Similarly, compound (-) BI97D6 effectively induces apoptosis in the BP3 human lymphoma cell line in a dose-dependent manner. The compound also shows little cytotoxicity against bax(-/-)/bak(-/-) cells, suggesting that it kills cancers cells predominantly via a Bcl-2 pathway. Moreover, compound (-) BI97D6 displays in vivo efficacy in both a Bcl-2-transgenic mouse model and in a prostate cancer xenograft model in mice. Therefore, compound (-) BI97D6 represents a promising drug lead for the development of novel apoptosis-based therapies for cancer.

20.
J Med Chem ; 53(22): 8000-11, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-21033669

ABSTRACT

Overexpression of antiapoptotic Bcl-2 family proteins is commonly related with tumor maintenance, progression, and chemoresistance. Inhibition of these antiapoptotic proteins is an attractive approach for cancer therapy. Guided by nuclear magnetic resonance (NMR) binding assays, a series of 5,5' substituted compound 6a (Apogossypolone) derivatives was synthesized and identified pan-active antagonists of antiapoptotic Bcl-2 family proteins, with binding potency in the low micromolar to nanomolar range. Compound 6f inhibits the binding of BH3 peptides to Bcl-X(L), Bcl-2, and Mcl-1 with IC(50) values of 3.10, 3.12, and 2.05 µM, respectively. In a cellular assay, 6f potently inhibits cell growth in several human cancer cell lines in a dose-dependent manner. Compound 6f further displays in vivo efficacy in transgenic mice and demonstrated superior single-agent antitumor efficacy in a PPC-1 mouse xenograft model. Together with its negligible toxicity, compound 6f represents a promising drug lead for the development of novel apoptosis-based therapies for cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Gossypol/analogs & derivatives , Naphthoquinones/chemical synthesis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Stability , Female , Gossypol/chemical synthesis , Gossypol/chemistry , Gossypol/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Nude , Mice, Transgenic , Microsomes/metabolism , Models, Molecular , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Neoplasm Transplantation , Peptide Fragments/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Structure-Activity Relationship , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...