Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Opioid Manag ; 12(1): 47-55, 2016.
Article in English | MEDLINE | ID: mdl-26908303

ABSTRACT

D-Methadone is the d optical isomer of racemic mixture (DL-methadone) used clinically to treat pain and addiction in the United States. D-Methadone is practically devoid of opioid activity but maintains N-methyl-D-aspartate (NMDA) receptor antagonism. Evidence from extensive preclinical studies suggests that NMDA receptor antagonists attenuate neuronal plasticity, reverse opioid analgesic tolerance, and alleviate chronic pain states. The authors conducted a phase I open label study of D-methadone administered for the first time to patients with chronic pain to determine the safety and tolerability of D-methadone. In addition to their long-term regimen of opioids, the patients received 40 mg of D-methadone twice daily for 12 days. Analgesia and toxicity were recorded by the patients in a daily diary and assessed in clinic on days 1, 8, and 12. Eight patients of the 10 enrolled completed the study. Pain scores on Edmonton Symptom Assessment System (ESAS) did not change between days 1 and 12, but five of eight patients (62.5 percent) characterized D-methadone as moderately or very effective in relieving pain on the Global Assessment for pain. Five of the eight patients (62.5 percent) who completed the study requested to start treatment with commercially available methadone (DL-racemic methadone) after completing the study. D-Methadone at the dose of 40 mg PO Q 12 hours was well tolerated. Perspective: This is the first clinical study of D-methadone in patients suffering from chronic pain. Additional phase I and phase II studies are needed to confirm its safety and analgesic effects. If D-methadone is well tolerated, it is likely to become a useful adjuvant to the treatment of a wide spectrum of pain syndromes.


Subject(s)
Analgesics, Opioid/therapeutic use , Chronic Pain/drug therapy , Methadone/therapeutic use , Humans , Methadone/adverse effects , Prospective Studies
2.
Pain ; 148(2): 237-246, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20005044

ABSTRACT

Following peripheral inflammation, NMDA receptor (NMDAR) activation in spinal cord dorsal horn neurons facilitates the generation of pain in response to low threshold inputs (allodynia) and signals the phosphorylation of protein kinase C (pPKC) and extracellular signal-regulated kinase 2 (pERK2). Intraplantar complete Freund's adjuvant (CFA) induces inflammatory nociception (allodynic pain) at 24 hours (h) with a concurrent increase in neuronal pPKCgamma and pERK2 but not glial pERK2. These effects are attenuated in a spatial knockout of the NMDAR (NR1 KO) confined to SCDH neurons. Although glia and proinflammatory cytokines are implicated in the maintenance of inflammatory pain and neuronal activation, the role of NMDARs and neuronal-glial-cytokine interactions that initiate and maintain inflammatory pain are not well defined. In the maintenance phase of inflammatory pain at 96h after CFA the NR1 KO mice are no longer protected from allodynia and the SCDH expression of pPKCgamma and pERK2 are increased. At 96h the expression of the proinflammatory cytokine, IL-1beta, and pERK2 are increased in astrocytes. Intrathecal IL-1 receptor antagonist (IL-1ra), acting on neuronal IL-1 receptors, completely reverses the allodynia at 96h after CFA. Deletion of NMDAR-dependent signaling in neurons protects against early CFA-induced allodynia. Subsequent NMDAR-independent signaling that involves neuronal expression of pPKCgamma and the induction of pERK2 and IL-1beta in activated astrocytes contributes to the emergence of NMDAR-independent inflammatory pain behavior at 96h after CFA. Effective reduction of the initiation and maintenance of inflammatory pain requires targeting the neuron-astrocyte-cytokine interactions revealed in these studies.


Subject(s)
Inflammation/complications , Neurons/metabolism , Pain Threshold/physiology , Pain/etiology , Pain/pathology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Antirheumatic Agents/pharmacology , Astrocytes/metabolism , Disease Models, Animal , Female , Freund's Adjuvant , Functional Laterality , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation/chemically induced , Inflammation/drug therapy , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/metabolism , MAP Kinase Kinase Kinase 2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/drug therapy , Pain/genetics , Phosphopyruvate Hydratase/metabolism , Phosphorylation , Protein Kinase C/metabolism , Receptors, N-Methyl-D-Aspartate/deficiency , Spinal Cord/pathology , Time Factors
3.
J Opioid Manag ; 3(3): 167-70, 2007.
Article in English | MEDLINE | ID: mdl-18027543

ABSTRACT

This article describes a case of severe opioid-induced pruritus following systemic morphine administration. Symptoms did not resolve after administration of antihistamines or rotation to fentanyl or hydromorphone, but oral oxycodone and small-dose intravenous naloxone did alleviate the patient's itching. The pathogenesis of opioid-induced pruritus and the rationale for opioid rotation are briefly discussed. Current and possible future therapeutic options are mentioned.


Subject(s)
Analgesics, Opioid/adverse effects , Morphine/adverse effects , Pruritus/chemically induced , Adolescent , Analgesics, Opioid/administration & dosage , Humans , Male , Morphine/administration & dosage , Pain/drug therapy
4.
J Cell Physiol ; 201(3): 374-84, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15389545

ABSTRACT

Geldanamycin (GA) binds to heat shock protein 90 (Hsp90) and interferes with its function which is to protect various cellular proteins involved in signaling, growth control, and survival from ubiquitination and subsequent degradation by the proteasome. Recently, we demonstrated that GA inhibited migration of glioma cells in vitro associated with downregulation of hypoxia-inducible factor (HIF-1 alpha) and phosphorylation of focal adhesion kinase (FAK) (Zagzag et al., 2003, J Cell Physiol 196:394-402). Here, we have investigated the mechanisms through which GA treatment of the T98G glioma cell line induces apoptosis. We found that GA treatment induced cell death in a caspase-dependent manner through activation of caspase-3 and PARP cleavage together with release of cytochrome c and apoptosis inducing factor (AIF) from the mitochondria. Use of synchronized T98G cells showed that GA treatment of glioma cells during S-phase enhanced cytotoxicity followed by M-phase arrest, resulting in mitotic catastrophe. In addition, apoptosis was associated with the downregulation of the survival protein, phosphorylated Akt (pAkt), an important signaling protein in the PI3K pathway, that is overexpressed in many cancers including gliomas. Given that many glioma tumors show deregulation of the PI3K signaling pathway, either through loss of the tumor suppressor protein PTEN or overexpression of the growth factor EGFR, the ability to identify different subsets of patients using simple immunohistochemistry for the presence of absence of pAkt could enable selection of the appropriate kinase inhibitor, such as GA, for drug therapy. Based on our data presented here, GA or its analogs may have potential in the treatment of glioma.


Subject(s)
Apoptosis/drug effects , Glioma/pathology , Mitosis/drug effects , Quinones/pharmacology , Apoptosis Inducing Factor , Benzoquinones , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytochromes c/metabolism , Flavoproteins/metabolism , Humans , Lactams, Macrocyclic , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism
5.
Cell Cycle ; 3(2): 230-4, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14712094

ABSTRACT

The mechanism of action of many chemotherapeutic agents targets the cell cycle. Recently, we demonstrated cytotoxic and other anti-tumor effects of flavopiridol, the first synthetic cyclin dependent kinase (CDK) inhibitor to enter clinical trials, on the murine GL261 glioma cell line in vitro (Newcomb et al., Cell Cycle 2003; 2:243). Given that flavopiridol has demonstrated anti-tumor activity in several human xenograft models, we wanted to evaluate it for anti-glioma activity in vivo in our established subcutaneous and intracranial GL261 experimental tumor models. In particular, the intracranial animal model recapitulates many of the histopathological and biological features of human high-grade glioma including both necrosis with pseudopalisading and invasion of the brain adjacent to tumor. Here we tested the activity of flavopiridol against tumors formed by GL261 cells, first as subcutaneous implants, and then in the intracranial model. We demonstrate efficacy of flavopiridol as a single modality treatment in delaying tumor growth in both animal models. We hypothesize that flavopiridol treatment induced tumor growth delay by two possible mechanisms involving growth arrest combined with recruitment of tumor cells to S-phase. Based on our findings, flavopiridol should be considered as a treatment approach for patients with high-grade glioma.


Subject(s)
Brain Neoplasms/drug therapy , Cell Division/physiology , Flavonoids/pharmacology , Glioma/drug therapy , Piperidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Brain Neoplasms/metabolism , Cell Death , Female , Flavonoids/therapeutic use , Glioma/metabolism , Mice , Models, Animal , Piperidines/therapeutic use , Tumor Cells, Cultured
6.
Cell Cycle ; 2(3): 243-50, 2003.
Article in English | MEDLINE | ID: mdl-12734434

ABSTRACT

Glioblastoma (GBM) remains one of the most challenging solid cancers to treat due to its highly proliferative, angiogenic and invasive nature. The small molecule CDK inhibitor, flavopiridol, has demonstrated antitumor activity in human xenograft models and is currently in clinical trials showing efficacy in patients with advanced disease. We have developed an experimental animal model using the murine glioma GL261 cells as a novel in vivo system to screen potential therapeutic agents for GBM. Results of in vitro testing demonstrate that flavopiridol has several relevant clinical characteristics such as its ability to: 1. inhibit cell growth; 2. inhibit cell migration; 3. decrease expression of cyclin D1, CDK4 and p21; 4. induce apoptosis in cells with high levels of p27 expression; and 5. decrease the expression of the anti-apoptotic protein Bcl-2. The mechanism by which flavopiridol induces apoptosis is mitochondrial-mediated. We demonstrate by electron microscopy and immunohistochemistry that drug treatment induces mitochondrial damage that was accompanied by the release of cytochrome c into the cytosol together with the translocation of apoptosis inducing factor (AIF) into the nucleus. This finding in murine glioma cells differs from the mechanism of flavopiridolinduced cell death reported by us for human glioma cells (Alonso et al., Mol Cancer Ther 2003; 2:139) where drug treatment induced a caspase- and cytochrome c-independent pathway in the absence of detectable damage to mitochondria. In apoptotic human glioma cells only translocation of AIF into the nucleus occurred. Thus, the same drug kills different types of glioma cells by different mitochondrial-dependent pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Eukaryotic Cells/drug effects , Flavonoids/pharmacology , Glioblastoma/drug therapy , Mitochondria/drug effects , Piperidines/pharmacology , Animals , Apoptosis/physiology , Apoptosis Inducing Factor , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cytochrome c Group/metabolism , Cytotoxins/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Eukaryotic Cells/metabolism , Flavoproteins/metabolism , G2 Phase/drug effects , Glioblastoma/genetics , Glioblastoma/metabolism , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Tumor Cells, Cultured
7.
Mol Cancer Ther ; 2(2): 139-50, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589031

ABSTRACT

Flavopiridol is a synthetic flavone, which inhibits growth in vitro and in vivo of several solid malignancies such as renal, prostate, and colon cancers. It is a potent cyclin-dependent kinase inhibitor presently in clinical trials. In this study, we examined the effect of flavopiridol on a panel of glioma cell lines having different genetic profiles: five of six have codeletion of p16(INK4a) and p14(ARF); three of six have p53 mutations; and one of six shows overexpression of mouse double minute-2 (MDM2) protein. Independent of retinoblastoma and p53 tumor suppressor pathway alterations, flavopiridol induced apoptosis in all cell lines but through a caspase-independent mechanism. No cleavage products for caspase 3 or its substrate poly(ADP-ribose) polymerase or caspase 8 were detected. The pan-caspase inhibitor Z-VAD-fmk did not inhibit flavopiridol-induced apoptosis. Mitochondrial damage measured by cytochrome c release and transmission electron microscopy was not observed in drug-treated glioma cells. In contrast, flavopiridol treatment induced translocation of apoptosis-inducing factor from the mitochondria to the nucleus. The proteins cyclin D(1) and MDM2 involved in the regulation of retinoblastoma and p53 activity, respectively, were down-regulated early after flavopiridol treatment. Given that MDM2 protein can confer oncogenic properties under certain circumstances, loss of MDM2 expression in tumor cells could promote increased chemosensitivity. After drug treatment, a low Bcl-2/Bax ratio was observed, a condition that may favor apoptosis. Taken together, the data indicate that flavopiridol has activity against glioma cell lines in vitro and should be considered for clinical development in the treatment of glioblastoma multiforme.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Glioma/pathology , Nuclear Proteins , Piperidines/pharmacology , Retinoblastoma Protein/physiology , Tumor Suppressor Protein p53/physiology , Apoptosis Inducing Factor , Blotting, Northern , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cytochrome c Group/metabolism , Flavoproteins/metabolism , Flow Cytometry , Glioma/drug therapy , Glioma/metabolism , Humans , Immunoenzyme Techniques , Membrane Proteins/metabolism , Microscopy, Electron , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...