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.
Ann Oncol ; 29(5): 1304-1311, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29788155

ABSTRACT

BACKGROUND: Checkpoint kinase 1 (Chk1) inhibition following chemotherapy-elicited DNA damage overrides cell cycle arrest and induces mitotic catastrophe and cell death. GDC-0575 is a highly-selective oral small-molecule Chk1 inhibitor that results in tumor shrinkage and growth delay in xenograft models. We evaluated the safety, tolerability, and pharmacokinetic properties of GDC-0575 alone and in combination with gemcitabine. Antitumor activity and Chk1 pathway modulation were assessed. PATIENTS AND METHODS: In this phase I open-label study, in the dose escalation stage, patients were enrolled in a GDC-0575 monotherapy Arm (1) or GDC-0575 combination with gemcitabine Arm (2) to determine the maximum tolerated dose. Patients in arm 2 received either i.v. gemcitabine 1000 mg/m2 (arm 2a) or 500 mg/m2 (arm 2b), followed by GDC-0575 (45 or 80 mg, respectively, as RP2D). Stage II enrolled disease-specific cohorts. RESULTS: Of 102 patients treated, 70% were female, the median age was 59 years (range 27-85), and 47% were Eastern Cooperative Oncology Group PS 0. The most common tumor type was breast (37%). The most frequent adverse events (all grades) related to GDC-0575 and/or gemcitabine were neutropenia (68%), anemia (48%), nausea (43%), fatigue (42%), and thrombocytopenia (35%). Maximum concentrations of GDC-0575 were achieved within 2 hours of dosing, and half-life was ∼23 hours. No pharmacokinetic drug-drug interaction was observed between GDC-0575 and gemcitabine. Among patients treated with GDC-0575 and gemcitabine, there were four confirmed partial responses, three occurring in patients with tumors harboring TP53 mutation. Pharmacodynamic data were consistent with GDC-0575 inhibition of gemcitabine-induced expression of pCDK1/2. CONCLUSION: GDC-0575 can be safely administered as a monotherapy and in combination with gemcitabine; however, overall tolerability with gemcitabine was modest. Hematological toxicities were frequent but manageable. Preliminary antitumor activity was observed but limited to a small number of patients with a variety of refractory solid tumors treated with GDC-0575 and gemcitabine. CLINICAL TRIAL NUMBER: NCT01564251.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Pyrroles/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Fatigue , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea , Neutropenia/chemically induced , Neutropenia/epidemiology , Piperidines/adverse effects , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Thrombocytopenia , Treatment Outcome , Gemcitabine
2.
CPT Pharmacometrics Syst Pharmacol ; 5(4): 211-221, 2016 04.
Article in English | MEDLINE | ID: mdl-27104090

ABSTRACT

We previously investigated novel therapies for pediatric ependymoma and found 5-fluorouracil (5-FU) i.v. bolus increased survival in a representative mouse model. However, without a quantitative framework to derive clinical dosing recommendations, we devised a translational pharmacokinetic-pharmacodynamic (PK-PD) modeling and simulation approach. Results from our preclinical PK-PD model suggested tumor concentrations exceeded the 1-hour target exposure (in vitro IC90), leading to tumor growth delay and increased survival. Using an adult population PK model, we scaled our preclinical PK-PD model to children. To select a 5-FU dosage for our clinical trial in children with ependymoma, we simulated various 5-FU dosages for tumor exposures and tumor growth inhibition, as well as considering tolerability to bolus 5-FU administration. We developed a pediatric population PK model of bolus 5-FU and simulated tumor exposures for our patients. Simulations for tumor concentrations indicated that all patients would be above the 1-hour target exposure for antitumor effect.


Subject(s)
Ependymoma/drug therapy , Fluorouracil/administration & dosage , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Blood Proteins/metabolism , Child , Child, Preschool , Computer Simulation , Drug Dosage Calculations , Ependymoma/blood , Ependymoma/metabolism , Fluorouracil/blood , Fluorouracil/pharmacokinetics , Humans , Mice , Models, Biological , Nonlinear Dynamics , Protein Binding , Random Allocation
3.
Br J Pharmacol ; 155(7): 1076-84, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18806808

ABSTRACT

BACKGROUND AND PURPOSE: Allergic inflammation and autoimmune diseases, such as atopic dermatitis, psoriasis and multiple sclerosis (MS), involve both mast cell and T-cell activation. However, possible interactions between the two and the mechanism of such activations are largely unknown. EXPERIMENTAL APPROACH: Human umbilical cord blood-derived cultured mast cells (hCBMCs) and Jurkat T cells were incubated separately or together, following activation with myelin basic protein (MBP), as well as with or without pretreatment with the flavonoid luteolin for 15 min. The supernatant fluid was assayed for inflammatory mediators released from mast cells and interleukin (IL)-2 release from Jurkat cells. KEY RESULTS: MBP (10 microM) stimulates hCBMCs to release IL-6, IL-8, transforming growth factor (TGF)-beta1, tumour necrosis factor-alpha (TNF-alpha), vascular endothelial growth factor (VEGF), histamine and tryptase (n=6, P<0.05). Addition of mast cells to Jurkat cells activated by anti-CD3/anti-CD28 increases IL-2 release by 30-fold (n=3, P<0.05). MBP-stimulated mast cells and their supernatant fluid further increase Jurkat cell IL-2 release (n=3, P<0.05). Separation of mast cells and activated Jurkat cells by a Transwell permeable membrane inhibits Jurkat cell stimulation by 60%. Pretreatment of Jurkat cells with a TNF-neutralizing antibody reduces IL-2 release by another 40%. Luteolin pretreatment inhibits mast cell activation (n=3-6, P<0.05), Jurkat cell activation and mast cell-dependent Jurkat cell stimulation (n=3, P<0.05). CONCLUSIONS AND IMPLICATIONS: Mast cells can stimulate activated Jurkat cells. This interaction is inhibited by luteolin, suggesting that this flavonoid may be useful in the treatment of autoimmune diseases.


Subject(s)
Jurkat Cells/drug effects , Luteolin/pharmacology , Mast Cells/drug effects , Myelin Basic Protein/antagonists & inhibitors , Cells, Cultured , Humans , Inflammation Mediators/metabolism , Interleukin-2/metabolism , Jurkat Cells/metabolism , Mast Cells/immunology , Myelin Basic Protein/metabolism , Tryptases/drug effects , Tryptases/metabolism
4.
Int J Immunopathol Pharmacol ; 20(3): 447-53, 2007.
Article in English | MEDLINE | ID: mdl-17880758

ABSTRACT

Chemokines are inflammatory proteins acting via G-protein coupled chemokine receptors that trigger different signaling pathways. Monocyte chemoattractant protein-1 (CCL2/MCP-1) and regulated on activation, normal T expressed and secreted (CCL5/RANTES) are the two major members of the CC chemokine beta subfamily. The roles of RANTES and MCP-1 are emerging in regulating the recruitment of inflammatory cells into tissue during inflammation. The inhibition of MCP-1 and RANTES with corresponding antibodies or other inhibitors may provide benefits in different clinical scenarios including cancer, inflammation, CNS disorders, parasitic disease, autoimmune and heart diseases. RANTES and MCP-1 may represent targets for diagnostic procedures and therapeutic intervention, and may be useful as a prognostic factor in the above diseases.


Subject(s)
Chemokines/antagonists & inhibitors , Inflammation/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/immunology , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/immunology , Chemokines/immunology , Humans , Inflammation/immunology
5.
Pharmacol Res ; 55(1): 57-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17110126

ABSTRACT

Allergy is the result of a complex immune cascade leading to the disregulated production of Th2 cytokines, the generation of allergen-specific IgE-producing B cells and the subsequent activation and degranulation of mast cells upon allergen challenge. Mast cell effector function significantly influences the quantity, duration and magnitude of most allergic reactions. Here, using isolated human umbilical cord blood mast cells (HUCBMC) from CD34+ cells, activated with anti-IgE (10 microg/ml) in culture, we found an augmented release of IL-6, tryptase and histamine (p < 0.01 compared with control). In addition, in these cells anti-IgE (10 microg/ml) activated the expression of histidine decarboxylase (HDC) and IL-6. In these studies we describe a new biological activity of anti-IgE in inducing histidine decarboxylase and IL-6, suggesting that this cytokine may have an important effect on allergic and inflammatory diseases mediated by mast cells. Moreover, with these data we confirm the immunoregulatory and inflammatory function of mast cells.


Subject(s)
Fetal Blood/cytology , Histidine Decarboxylase/biosynthesis , Interleukin-6/biosynthesis , Mast Cells/immunology , RNA, Messenger/biosynthesis , Tryptases/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Histamine Release/physiology , Histidine Decarboxylase/genetics , Humans , Immunoglobulin E/immunology , Mast Cells/enzymology , Microscopy, Electron, Transmission , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Stem Cell Rev ; 2(2): 143-54, 2006.
Article in English | MEDLINE | ID: mdl-17237553

ABSTRACT

Findings obtained using animal models have often failed to reflect the processes involved in human disease. Moreover, human cultured cells do not necessarily function as their actual tissue counterparts. Therefore, there is great demand for sources of human progenitor cells that may be directed to acquire specific tissue characteristics and be available in sufficient quantities to carry out functional and pharmacological studies. Acase in point is the mast cell, well known for its involvement in allergic reactions, but also implicated in inflammatory diseases. Mast cells can be activated by allergens, anaphylatoxins, immunoglobulin-free light chains, superantigens, neuropeptides, and cytokines, leading to selective release of mediators. These could be involved in many inflammatory diseases, such as asthma and atopic dermatitis, which worsen by stress, through activation by local release of corticotropin-releasing hormone or related peptides. Umbilical cord blood and cord matrix-derived mast cell progenitors can be separated magnetically and grown in the presence of stem cell factor, interleukin-6, interleukin-4, and other cytokines to yield distinct mast cell populations. The recent use of live cell array, with its ability to study such interactions rapidly at the single-cell level, provides unique new opportunities for fast output screening of mast cell triggers and inhibitors.


Subject(s)
Fetal Blood/cytology , Mast Cells/cytology , Models, Biological , Neurosecretory Systems/immunology , Humans , Inflammation/diagnosis , Neurosecretory Systems/cytology , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...