Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 20(1): 114-126, 2022 01.
Article in English | MEDLINE | ID: mdl-34635507

ABSTRACT

In medulloblastoma, p53 expression has been associated with chemoresistance and radiation resistance and with poor long-term outcomes in the p53-mutated sonic hedgehog, MYC-p53, and p53-positive medulloblastoma subgroups. We previously established a direct role for p53 in supporting drug resistance in medulloblastoma cells with high basal protein expression levels (D556 and DAOY). We now show that p53 genetic suppression in medulloblastoma cells with low basal p53 protein expression levels (D283 and UW228) significantly reduced drug responsiveness, suggesting opposing roles for low p53 protein expression levels. Mechanistically, the enhanced cell death by p53 knockdown in high-p53 cells was associated with an induction of mTOR/PI3K signaling. Both mTOR inhibition and p110α/PIK3CA induction confirmed these findings, which abrogated or accentuated the enhanced chemosensitivity response in D556 cells respectively while converse was seen in D283 cells. Co-treatment with G-actin-sequestering peptide, thymosin ß4 (Tß4), induced p-AKTS473 in both p53-high and p53-low cells, enhancing chemosensitivity in D556 cells while enhancing chemoresistance in D283 and UW228 cells. IMPLICATIONS: Collectively, we identified an unexpected role for the PI3K signaling in enhancing cell death in medulloblastoma cells with high basal p53 expression. These studies indicate that levels of p53 immunopositivity may serve as a diagnostic marker of chemotherapy resistance and for defining therapeutic targeting.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Phosphatidylinositol 3-Kinase/metabolism , Tumor Suppressor Protein p53/metabolism , Cerebellar Neoplasms/pathology , Humans , Medulloblastoma/pathology , Signal Transduction
2.
Sci Rep ; 10(1): 8096, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415084

ABSTRACT

Abnormal regulation of ß-catenin initiates an oncogenic program that serves as a main driver of many cancers. Albeit challenging, ß-catenin is an attractive drug target due to its role in maintenance of cancer stem cells and potential to eliminate cancer relapse. We have identified C2, a novel ß-catenin inhibitor, which is a small molecule that binds to a novel allosteric site on the surface of ß-catenin. C2 selectively inhibits ß-catenin, lowers its cellular load and significantly reduces viability of ß-catenin-driven cancer cells. Through direct binding to ß-catenin, C2 renders the target inactive that eventually activates proteasome system for its removal. Here we report a novel pharmacologic approach for selective inhibition of ß-catenin via targeting a cryptic allosteric modulation site. Our findings may provide a new perspective for therapeutic targeting of ß-catenin.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Allosteric Regulation , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
ACS Chem Neurosci ; 11(9): 1218-1230, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32286796

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine (ser)/threonine (Thr) kinase that has been demonstrated to be one of the most functionally diverse kinases within neurons. Cdk5 is regulated via binding with its neuron-specific regulatory subunits, p35 or p39. Cdk5-p35 activity is critical for a variety of developmental and cellular processes in the brain, including neuron migration, memory formation, microtubule regulation, and cell cycle suppression. Aberrant activation of Cdk5 via the truncated p35 byproduct, p25, is implicated in the pathogenesis of several neurodegenerative diseases. The present review highlights the importance of Cdk5 activity and function in the brain and demonstrates how deregulation of Cdk5 can contribute to the development of neurodegenerative conditions such as Alzheimer's and Parkinson's disease. Additionally, we cover past drug discovery attempts at inhibiting Cdk5-p25 activity and discuss which types of targeting strategies may prove to be the most successful moving forward.


Subject(s)
Cyclin-Dependent Kinase 5 , Neurodegenerative Diseases , Cyclin-Dependent Kinase 5/metabolism , Humans , Neurodegenerative Diseases/drug therapy , Neurogenesis , Neurons/metabolism , Phosphorylation
4.
Biotechniques ; 68(4): 180-184, 2020 04.
Article in English | MEDLINE | ID: mdl-32040335

ABSTRACT

CRISPR-Cas9 has proven to be a versatile tool for the discovery of essential genetic elements involved in various disease states. CRISPR-assisted dense mutagenesis focused on therapeutically challenging protein complexes allows us to systematically perturb protein-coding sequences in situ and correlate them with functional readouts. Such perturbations can mimic targeting by therapeutics and serve as a foundation for the discovery of highly specific modulators. However, translation of such genomics data has been challenging due to the missing link for proteomics under the physiological state of the cell. We present a method based on cellular thermal shift assays to easily interrogate proteomic shifts generated by CRISPR-assisted dense mutagenesis, as well as a case focused on NuRD epigenetic complex.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Mutagenesis, Insertional/genetics , Proteome/genetics , Proteomics/methods , Cell Line , Humans , Proteome/analysis
5.
Int J Nanomedicine ; 14: 1937-1952, 2019.
Article in English | MEDLINE | ID: mdl-30936695

ABSTRACT

Lipid-polymer hybrid nanoparticles (LPHNPs) are next-generation core-shell nanostructures, conceptually derived from both liposome and polymeric nanoparticles (NPs), where a polymer core remains enveloped by a lipid layer. Although they have garnered significant interest, they remain not yet widely exploited or ubiquitous. Recently, a fundamental transformation has occurred in the preparation of LPHNPs, characterized by a transition from a two-step to a one-step strategy, involving synchronous self-assembly of polymers and lipids. Owing to its two-in-one structure, this approach is of particular interest as a combinatorial drug delivery platform in oncology. In particular, the outer surface can be decorated in multifarious ways for active targeting of anticancer therapy, delivery of DNA or RNA materials, and use as a diagnostic imaging agent. This review will provide an update on recent key advancements in design, synthesis, and bioactivity evaluation as well as discussion of future clinical possibilities of LPHNPs.


Subject(s)
Drug Delivery Systems/methods , Lipids/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Animals , Humans , Lipids/administration & dosage , Liposomes/administration & dosage , Liposomes/chemistry , Nanoparticles/administration & dosage , Nanotechnology/methods , Polymers/administration & dosage
6.
Hum Antibodies ; 27(1): 53-62, 2019.
Article in English | MEDLINE | ID: mdl-30223393

ABSTRACT

Antibody drug conjugates (ADCs) represent a promising and an efficient strategy for targeted cancer therapy. Comprised of a monoclonal antibody, a cytotoxic drug, and a linker, ADCs offer tumor selectively, reduced toxicity, and improved stability in systemic circulation. Recent approvals of two ADCs have led to a resurgence in ADC research, with more than 60 ADCs under various stages of clinical development. The therapeutic success of future ADCs is dependent on adherence to key requirements of their design and careful selection of the target antigen on cancer cells. Here we review the main components in the design of antibody drug conjugates, improvements made, and lessons learned over two decades of research, as well as the future of third generation ADCs.


Subject(s)
Drug Therapy/trends , Immunoconjugates/therapeutic use , Molecular Targeted Therapy/trends , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents , Humans
7.
Eur J Pharmacol ; 845: 1-7, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30529197

ABSTRACT

Sazetidine-A [6-(5(((S)-azetidine-2-yl)methoxy)pyridine-3-yl)hex-5-yn-1-ol] is a selective α4ß2 nicotinic receptor desensitizing agent and partial agonist. Sazetidine-A has been shown in our previous studies to significantly reduce nicotine and alcohol self-administration in rats. The question arises whether sazetidine-A would reduce self-administration of other addictive drugs as well. Nicotinic receptors on the dopaminergic neurons in the ventral tegmental area play an important role in controlling the activity of these neurons and release of dopamine in the nucleus accumbens, which is critical mechanism for reinforcing value of drugs of abuse. Previously, we showed that the nonspecific nicotinic antagonist mecamylamine significantly reduces cocaine self-administration in rats. In this study, we acutely administered systemically sazetidine-A and two other selective α4ß2 nicotinic receptor-desensitizing agents, VMY-2-95 and YL-2-203, to young adult female Sprague-Dawley rats and determined their effects on IV self-administration of cocaine and methamphetamine. Cocaine self-administration was significantly reduced by 0.3 mg/kg of sazetidine-A. In another set of rats, sazetidine-A (3 mg/kg) significantly reduced methamphetamine self-administration. VMY-2-95 significantly reduced both cocaine and methamphetamine self-administration with threshold effective doses of 3 and 0.3 mg/kg, respectively. In contrast, YL-2-203 did not significantly reduce cocaine self-administration at the same dose range and actually significantly increased cocaine self-administration at the 1 mg/kg dose. YL-2-203 (3 mg/kg) did significantly decrease methamphetamine self-administration. Sazetidine-A and VMY-2-95 are promising candidates to develop as new treatments to help addicts successfully overcome a variety of addictions including tobacco, alcohol as well as the stimulant drugs cocaine and methamphetamine.


Subject(s)
Amphetamine-Related Disorders/drug therapy , Azetidines/pharmacology , Cocaine-Related Disorders/drug therapy , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Animals , Azetidines/administration & dosage , Cocaine/administration & dosage , Female , Methamphetamine/administration & dosage , Nicotinic Agonists/administration & dosage , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Self Administration
9.
Hum Antibodies ; 26(2): 95-101, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29036806

ABSTRACT

Immunotherapy is now at the forefront of cancer therapeutic development. Gliomas are a particularly aggressive form of brain cancer for which immunotherapy may hold promise. Pritumumab (also known in the literature as CLNH11, CLN-IgG, and ACA-11) was the first monoclonal antibody tested in cancer patients. Pritumumab is a natural human monoclonal antibody developed from a B lymphocyte isolated from a regional draining lymph node of a patient with cervical carcinoma. The antibody binds ecto-domain vimentin on the surface of cancer cells. Pritumumab was originally tested in clinical trials with brain cancer patients in Japan where it demonstrated therapeutic benefit. It was reported to be a safe and effective therapy for brain cancer patients at doses 5-10 fold less than currently approved antibodies. Phase I dose escalation clinical trials are now being planned with pritumumab for the near future. Here we review data on the development and characterization of pritumumab, and review clinical trails data assessing immunotherapeutic effects of pritumumab for glioma patients.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antineoplastic Agents, Immunological/isolation & purification , Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunoglobulin G/isolation & purification , Vimentin/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/metabolism , Antineoplastic Agents, Immunological/therapeutic use , B-Lymphocytes/chemistry , B-Lymphocytes/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Clinical Trials as Topic , Gene Expression , Glioma/genetics , Glioma/immunology , Glioma/mortality , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/therapeutic use , Immunotherapy/methods , Mice , Survival Analysis , Vimentin/antagonists & inhibitors , Vimentin/genetics , Xenograft Model Antitumor Assays
10.
J Transl Med ; 15(1): 210, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29047383

ABSTRACT

BACKGROUND: The mitochondrial protein p32 is a validated therapeutic target of cancer overexpressed in glioma. Therapeutic targeting of p32 with monoclonal antibody or p32-binding LyP-1 tumor-homing peptide can limit tumor growth. However, these agents do not specifically target mitochondrial-localized p32 and would not readily cross the blood-brain barrier to target p32-overexpressing gliomas. Identifying small molecule inhibitors of p32 overexpressed in cancer is a more rational therapeutic strategy. Thus, in this study we employed a pharmacophore modeling strategy to identify small molecules that could bind and inhibit mitochondrial p32. METHODS: A pharmacophore model of C1q and LyP-1 peptide association with p32 was used to screen a virtual compound library. A primary screening assay for inhibitors of p32 was developed to identify compounds that could rescue p32-dependent glutamine-addicted glioma cells from glutamine withdrawal. Inhibitors from this screen were analyzed for direct binding to p32 by fluorescence polarization assay and protein thermal shift. Affect of the p32 inhibitor on glioma cell proliferation was assessed by Alamar Blue assay, and affect on metabolism was examined by measuring lactate secretion. RESULTS: Identification of a hit compound (M36) validates the pharmacophore model. M36 binds directly to p32 and inhibits LyP-1 tumor homing peptide association with p32 in vitro. M36 effectively inhibits the growth of p32 overexpressing glioma cells, and sensitizes the cells to glucose depletion. CONCLUSIONS: This study demonstrates a novel screening strategy to identify potential inhibitors of mitochondrial p32 protein overexpressed in glioma. High throughput screening employing this strategy has potential to identify highly selective, potent, brain-penetrant small molecules amenable for further drug development.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Brain Neoplasms/pathology , Carrier Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Fluorescence Polarization , Glioma/pathology , Glucose/pharmacology , Humans , Lactic Acid/metabolism , Mitochondrial Proteins/chemistry , Models, Molecular , Recombinant Proteins/metabolism , Small Molecule Libraries/chemistry
11.
Immunotherapy ; 9(7): 589-606, 2017 06.
Article in English | MEDLINE | ID: mdl-28595516

ABSTRACT

The clinical success of checkpoint inhibitors has led to a renaissance of interest in cancer immunotherapies. In particular, the possibility of ex vivo expanding autologous lymphocytes that specifically recognize tumor cells has attracted much research and clinical trial interest. In this review, we discuss the historical background of tumor immunotherapy using cell-based approaches, and provide some rationale for overcoming current barriers to success of autologous immunotherapy. An overview of adoptive transfer of lymphocytes, tumor infiltrating lymphocytes and dendritic cell therapies is provided. We conclude with discussing the possibility of gene-manipulating immune cells in order to augment therapeutic activity, including silencing of the immune-suppressive zinc finger orphan nuclear receptor, NR2F6, as an attractive means of overcoming tumor-associated immune suppression.


Subject(s)
Dendritic Cells/transplantation , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/transplantation , Neoplasms/therapy , Receptors, Steroid/genetics , T-Lymphocytes/transplantation , Animals , Dendritic Cells/immunology , Genetic Therapy , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , RNA Interference , Repressor Proteins , T-Lymphocytes/immunology , Tumor Microenvironment
12.
Psychopharmacology (Berl) ; 234(17): 2517-2523, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28555315

ABSTRACT

RATIONALE AND OBJECTIVES: Desensitization of neuronal nicotinic acetylcholine receptors holds promise as an effective treatment of tobacco addiction. Previously, we found that sazetidine-A (Saz-A), which selectively desensitizes α4ß2 nicotinic receptors, significantly decreased intravenous (IV) nicotine self-administration (SA) in rats with an effective dose of 3 mg/kg in acute and repeated injection studies. We also found that chronic infusions of Saz-A at doses of 2 and 6 mg/kg/day significantly reduced nicotine SA in rats. In continuing studies, we have characterized other Saz-A analogs, YL-2-203 and VMY-2-95, to determine their efficacies in reducing nicotine SA in rats. METHODS: Young adult female Sprague-Dawley rats were fitted with IV catheters and were trained for nicotine SA (0.03 mg/kg/infusion) on a fixed ratio 1 schedule for ten sessions. The same rats were also implanted subcutaneously with osmotic minipumps to continually deliver 2 or 6 mg/kg body weight YL-2-203, VMY-2-95, or saline for four consecutive weeks. RESULTS: Chronic administration of VMY-2-95 at doses of 2 and 6 mg/kg/day caused significant (p < 0.01) decreases in nicotine SA over the 2 weeks of continued nicotine SA and for the 1-week period of resumed access after a week of enforced abstinence, whereas chronic administration of YL-2-203 at the same doses was not found to be effective. CONCLUSIONS: These studies, together with our previous studies of Saz-A, revealed a spectrum of efficacies for these α4ß2 nicotinic receptor desensitizing agents and provide a path forward for the most effective compounds to be further developed as possible aids to smoking cessation.


Subject(s)
Azetidines/administration & dosage , Behavior, Animal/drug effects , Nicotine/administration & dosage , Nicotinic Antagonists/administration & dosage , Pyridines/administration & dosage , Receptors, Nicotinic , Animals , Dose-Response Relationship, Drug , Female , Nicotinic Agonists/administration & dosage , Rats , Rats, Sprague-Dawley , Self Administration
13.
Aging (Albany NY) ; 7(10): 854-68, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26540407

ABSTRACT

Medulloblastoma (MB), a primitive neuroectodermal tumor, is the most common malignant childhood brain tumor and remains incurable in about a third of patients. Currently, survivors carry a significant burden of late treatment effects. The p53 tumor suppressor protein plays a crucial role in influencing cell survival in response to cellular stress and while the p53 pathway is considered a key determinant of anti-tumor responses in many tumors, its role in cell survival in MB is much less well defined. Herein, we report that the experimental drug VMY-1-103 acts through induction of a partial DNA damage-like response as well induction of non-survival autophagy. Surprisingly, the genetic or chemical silencing of p53 significantly enhanced the cytotoxic effects of both VMY and the DNA damaging drug, doxorubicin. The inhibition of p53 in the presence of VMY revealed increased late stage apoptosis, increased DNA fragmentation and increased expression of genes involved in apoptosis, including CAPN12 and TRPM8, p63, p73, BIK, EndoG, CIDEB, P27Kip1 and P21cip1. These data provide the groundwork for additional studies on VMY as a therapeutic drug and support further investigations into the intriguing possibility that targeting p53 function may be an effective means of enhancing clinical outcomes in MB.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Dansyl Compounds/pharmacology , Medulloblastoma/drug therapy , Tumor Suppressor Protein p53/antagonists & inhibitors , Adenine/pharmacology , Adenine/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Dansyl Compounds/therapeutic use , Drug Evaluation, Preclinical , Humans , Signal Transduction/drug effects
14.
Bioorg Med Chem ; 23(5): 1102-11, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25650313

ABSTRACT

Interest in the mechanisms of DNA repair pathways, including the base excision repair (BER) pathway specifically, has heightened since these pathways have been shown to modulate important aspects of human disease. Modulation of the expression or activity of a particular BER enzyme, N-methylpurine DNA glycosylase (MPG), has been demonstrated to play a role in carcinogenesis and resistance to chemotherapy as well as neurodegenerative diseases, which has intensified the focus on studying MPG-related mechanisms of repair. A specific small molecule inhibitor for MPG activity would be a valuable biochemical tool for understanding these repair mechanisms. By screening several small molecule chemical libraries, we identified a natural polyphenolic compound, morin hydrate, which inhibits MPG activity specifically (IC50=2.6µM). Detailed mechanism analysis showed that morin hydrate inhibited substrate DNA binding of MPG, and eventually the enzymatic activity of MPG. Computational docking studies with an x-ray derived MPG structure as well as comparison studies with other structurally-related flavonoids offer a rationale for the inhibitory activity of morin hydrate observed. The results of this study suggest that the morin hydrate could be an effective tool for studying MPG function and it is possible that morin hydrate and its derivatives could be utilized in future studies focused on the role of MPG in human disease.


Subject(s)
DNA Glycosylases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Cell Line, Tumor , DNA Repair , Drug Evaluation, Preclinical , Flavonoids/chemistry , Humans , Models, Molecular , Structure-Activity Relationship
16.
Mol Pharm ; 12(2): 393-402, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25533629

ABSTRACT

The discovery and development of small molecules that antagonize neuronal nicotinic acetylcholine receptors may provide new ligands for evaluation in models of depression or addiction. We discovered a small molecule, VMY-2-95, a nAChR ligand with picomolar affinity and high selectivity for α4ß2 receptors. In this study, we investigated its preclinical profile in regards to solubility, lipophilicity, metabolic stability, intestinal permeability, bioavailability, and drug delivery to the rat brain. Metabolic stability of VMY-2-95·2HCl was monitored on human liver microsomes, and specific activity of VMY-2-95·2HCl on substrate metabolism by CYP1A2, 2C9, 2C19, 2D6, and 3A4 was tested in a high-throughput manner. The intestinal transport of VMY-2-95·2HCl was studied through Caco-2 cell monolayer permeability. VMY-2-95·2HCl was soluble in water and chemically stable, and the apparent partition coefficient was 0.682. VMY-2-95·2HCl showed significant inhibition of CYP2C9 and 2C19, but weak or no effect on 1A2, 2D6, and 3A4. The Caco-2 cell model studies revealed that VMY-2-95·2HCl was highly permeable with efflux ratio of 1.11. VMY-2-95·2HCl achieved a maximum serum concentration of 0.56 mg/mL at 0.9 h and was orally available with a half-life of ∼9 h. Furthermore, VMY-2-95·2HCl was detected in the rat brain after 3 mg/kg oral administration and achieved a maximal brain tissue concentration of 2.3 µg/g within 60 min. Overall, the results demonstrate that VMY-2-95·2HCl has good drug like properties and can penetrate the blood-brain barrier with oral administration.


Subject(s)
Azetidines/metabolism , Microsomes, Liver/metabolism , Pyridines/metabolism , Receptors, Nicotinic/metabolism , Animals , Blood-Brain Barrier/metabolism , Caco-2 Cells , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/metabolism , Drug Stability , Humans , Hydrogen-Ion Concentration , Ligands , Male , Rats , Rats, Sprague-Dawley
17.
Oncotarget ; 5(21): 10678-91, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25296977

ABSTRACT

The p53 tumor suppressor protein plays a crucial role in influencing cell fate decisions in response to cellular stress. As p53 elicits cell cycle arrest, senescence or apoptosis, the integrity of the p53 pathway is considered a key determinant of anti-tumor responses. p53 can also promote autophagy, however the role of p53-dependent autophagy in chemosensitivity is poorly understood. VMY-1-103 (VMY), a dansylated analog of purvalanol B, displays rapid and potent anti-tumor activities, however the pathways by which VMY works are not fully defined. Using established prostate cancer cell lines and novel conditionally reprogrammed cells (CRCs) derived from prostate cancer patients; we have defined the mechanisms of VMY-induced prostate cancer cell death. Herein, we show that the cytotoxic effects of VMY required a p53-dependent induction of autophagy, and that inhibition of autophagy abrogated VMY-induced cell death. Cancer cell lines harboring p53 missense mutations evaded VMY toxicity and treatment with a small molecule compound that restores p53 activity re-established VMY-induced cell death. The elucidation of the molecular mechanisms governing VMY-dependent cell death in cell lines, and importantly in CRCs, provides the rationale for clinical studies of VMY, alone or in combination with p53 reactivating compounds, in human prostate cancer.


Subject(s)
Adenine/analogs & derivatives , Apoptosis/drug effects , Autophagy/drug effects , Dansyl Compounds/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Adenine/pharmacology , Blotting, Western , Cell Proliferation , Flow Cytometry , Humans , Male , Mutation/genetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
18.
J Med Chem ; 56(21): 8404-21, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24047231

ABSTRACT

Developing novel and selective compounds that desensitize α4ß2 nicotinic acetylcholine receptors (nAChRs) could provide new effective treatments for nicotine addiction, as well as other disorders. Here we report a new class of nAChR ligands that display high selectivity and picomolar binding affinity for α4ß2 nicotinic receptors. The novel compounds have Ki values in the range of 0.031-0.26 nM and properties that should make them good candidates as drugs acting in the CNS. The selected lead compound 1 (VMY-2-95) binds with high affinity and potently desensitizes α4ß2 nAChRs. At a dose of 3 mg/kg, compound 1 significantly reduced rat nicotine self-administration. The overall results support further characterizations of compound 1 and its analogues in preclinical models of nicotine addiction and perhaps other disorders involving nAChRs.


Subject(s)
Azetidines/pharmacology , Drug Discovery , Pyridines/pharmacology , Receptors, Nicotinic/metabolism , Azetidines/chemical synthesis , Azetidines/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Software , Structure-Activity Relationship
19.
Eur J Pharmacol ; 718(1-3): 167-72, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24036108

ABSTRACT

Nicotine elicits hypothermic responses in rodents. This effect appears to be related to nicotinic receptor desensitization because sazetidine-A, an α4ß2 nicotinic receptor desensitizing agent, produces marked hypothermia and potentiates nicotine-induced hypothermia in mice. To determine the specificity of sazetidine-A induced hypothermia to ß2 subunit-containing nicotinic receptors, we tested its efficacy in ß2 knockout (ß2(-/-)) mice. These effects were compared with wildtype (WT) and α7 knockout (α7(-/-)) mice. Confirming our earlier results, sazetidine-A elicited a pronounced and long-lasting hypothermia in WT mice. In comparison, sazetidine-A induced a much attenuated and shorter hypothermic response in ß2(-/-) mice. This indicates that the greater proportion of sazetidine-A induced hypothermia is mediated via actions on ß2-containing nicotinic receptors, while a smaller component of hypothermia induced by sazetidine-A is mediated by non-ß2 receptors. Similar to WT mice, α7(-/-) mice showed the full extent of the sazetidine-A effect, suggesting that the hypothermia produced by sazetidine-A did not depend on actions on α7 nicotinic receptor subtype. Three other novel nicotinic receptor desensitizing agents derived from sazetidine-A, triazetidine-O, VMY-2-95 and YL-1-127 also produced hypothermia in WT and α7(-/-) mice. Furthermore, unlike sazetidine-A, triazetidine-O and YL-1-127 did not show any hint of a hypothermic effect in ß2(-/-) mice. VMY-2-95 like sazetidine-A did show a residual hypothermic effect in the ß2(-/-) mice. These studies show that the hypothermic effects of sazetidine-A and the related compound VMY-2-95 are mainly mediated by nicotinic receptors containing ß2 subunit, but that a small component of the effect is apparently mediated by non-ß2 containing receptors.


Subject(s)
Azetidines/pharmacology , Body Temperature/drug effects , Pyridines/pharmacology , Receptors, Nicotinic/deficiency , alpha7 Nicotinic Acetylcholine Receptor/deficiency , Animals , Gene Knockout Techniques , Male , Mice , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
20.
J Med Chem ; 56(7): 3000-11, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23540678

ABSTRACT

Neuronal acetylcholine receptors mediate the addictive effects of nicotine and may also be involved in alcohol addiction. Varenicline, an approved smoking cessation medication, showed clear efficacy in reducing alcohol consumption in heavy-drinking smokers. More recently, sazetidine-A, which selectively desensitizes α4ß2 nicotinic receptors, was shown to significantly reduce alcohol intake in a rat model. To develop novel therapeutics for treating alcohol use disorder, we designed and synthesized novel sazetidine-A analogues containing a methyl group at the 2-position of the pyridine ring. In vitro pharmacological studies revealed that some of the novel compounds showed overall pharmacological property profiles similar to that of sazetidine-A but exhibited reduced agonist activity across all nicotinic receptor subtypes tested. In rat studies, compound (S)-9 significantly reduced alcohol uptake. More importantly, preliminary results from studies in a ferret model indicate that these novel nAChR ligands have an improved adverse side-effect profile in comparison with that of varenicline.


Subject(s)
Alcohol Drinking/prevention & control , Ethanol/administration & dosage , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Animals , Ferrets , Ligands , Magnetic Resonance Spectroscopy , Rats , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...