Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
AAPS J ; 23(2): 31, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619657

ABSTRACT

Physiologically based pharmacokinetic (PBPK) absorption modeling and simulation is increasingly used as a tool in drug product development, not only in support of clinical pharmacology applications (e.g., drug-drug interaction, dose selection) but also from quality perspective, enhancing drug product understanding. This report provides a summary of the status and the application of PBPK absorption modeling and simulation in new drug application (NDA) submissions to the U.S. Food and Drug Administration to support drug product quality (e.g., clinically relevant dissolution specifications, active pharmaceutical ingredient (API) particle size distribution specifications). During the 10 years from 2008 to 2018, a total of 24 NDA submissions included the use of PBPK absorption modeling and simulations for biopharmaceutics-related assessment. In these submissions, PBPK absorption modeling and simulation served as an impactful tool in establishing the relationship of critical quality attributes (CQAs) including formulation variables, specifically in vitro dissolution, to the in vivo performance. This article also summarizes common practices in PBPK approaches and proposes future directions for the use of PBPK absorption modeling and simulation in drug product quality assessment.Graphical abstract.


Subject(s)
Drug Approval , Drug Development/methods , Gastrointestinal Absorption/physiology , Models, Biological , United States Food and Drug Administration/standards , Chemistry, Pharmaceutical/standards , Computer Simulation/standards , Drug Development/standards , Drug Liberation/physiology , Humans , Metabolic Clearance Rate/physiology , Tissue Distribution/physiology , United States
2.
AAPS J ; 21(2): 29, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30746576

ABSTRACT

This publication summarizes the proceedings of day 2 of a 3-day workshop on "Dissolution and Translational Modeling Strategies Enabling Patient-Centric Product Development." Patient-centric drug product development from a drug product quality perspective necessitates the establishment of clinically relevant drug product specifications via an in vitro-in vivo link. Modeling and simulation offer a path to establish this link; in this regard, physiologically based modeling has been implemented successfully to support regulatory decision-making and drug product labeling. In this manuscript, case studies of physiologically based biopharmaceutics modeling (PBBM) applied to drug product quality are presented and summarized. These case studies exemplify a possible path to achieve an in vitro-in vivo link and encompass (a) development of biopredictive dissolution methods to support biowaivers, (b) model-informed formulation selection, (c) predicting clinical formulation performance, and (d) defining a safe space for regulatory flexibility via virtual bioequivalence (BE). Workflows for the development and verification of absorption models/PBBM and for the establishment of a safe space using dissolution as an input are described with examples. Breakout session discussions on topics, such as current challenges and some best practices in model development and verification, are included as part of the Supplementary material.


Subject(s)
Biological Products/pharmacokinetics , Biopharmaceutics/methods , Drug Development/methods , Models, Biological , Absorption, Physiological , Biopharmaceutics/standards , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Congresses as Topic , Drug Development/standards , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Drug Labeling/standards , Drug Liberation , Humans , Solubility , Therapeutic Equivalency
3.
J Pharm Sci ; 105(10): 3088-3096, 2016 10.
Article in English | MEDLINE | ID: mdl-27449228

ABSTRACT

A Food and Drug Administration-approved generic oral product of venlafaxine hydrochloride (HCl) extended-release (ER) tablets has used a release mechanism based on an openable matrix, which is different from the push-pull osmotic pump system of its reference-listed drug. In an extreme case, a delay in the bursting of the openable matrix may be considered a product failure mode that alters the intended profile of systemic exposure. A physiologically based pharmacokinetic absorption model was established and verified to simulate the pharmacokinetic profiles after a single-dose oral administration of ER venlafaxine HCl tablets based on an osmotic pump or openable matrix design. This model adequately predicted the observed human mean pharmacokinetic metrics with <20% difference between the predicted and observed data. Based on the modeling and simulation results, Cmax and AUCt of the venlafaxine openable matrix tablets were entirely within the bioequivalence acceptance limits (i.e., 80%-125%) when the lag time varied from 0 to 4 h and using drug-release profiles under most dissolution conditions. The results indicated that a bioinequivalence risk is minimal for a delayed onset of drug release from the approved generic venlafaxine HCl ER tablets with an openable matrix design, supporting its substitutability to the reference product.


Subject(s)
Drug Compounding/methods , Infusion Pumps, Implantable , Venlafaxine Hydrochloride/administration & dosage , Venlafaxine Hydrochloride/pharmacokinetics , Administration, Oral , Capsules , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Infusions, Intravenous , Osmosis/drug effects , Serotonin and Noradrenaline Reuptake Inhibitors/administration & dosage , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacokinetics , Tablets
4.
Pharmacotherapy ; 35(7): 670-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26172282

ABSTRACT

OBJECTIVE: Neonatal abstinence syndrome (NAS)--a clinical entity of infants from in utero exposure to psychoactive xenobiotic and buprenorphine--has been successfully used to treat NAS. However, nothing is known about the pharmacokinetics (PK) of buprenorphine in neonates with NAS. To our knowledge, this is the first study to investigate the population pharmacokinetic of sublingual buprenorphine in neonates with NAS. DESIGN: A retrospective population PK analysis of: (1) neonates with NAS treated with sublingual buprenorphine in randomized, double blinded clinical study and (2) data from healthy adults from a previously published pharmacokinetic study. SETTING: Neonatal intensive care unit and general clinical research unit. PATIENTS: Twenty-four neonates with NAS and five healthy adults. INTERVENTIONS: All participants received sublingual buprenorphine per study protocol. MEASUREMENTS AND MAIN RESULTS: A total of 303 PK data from 29 neonates and adults were used for model development. A population pharmacokinetic analysis was conducted using a first order conditional estimation with interaction in the NONMEM software program. A two-compartment linear PK model with first-order absorption process best described the pharmacokinetics of sublingual buprenorphine in neonates. The apparent clearance (CL) of buprenorphine was linearly related to body weight and matured with increasing age via two distinct saturated pathways. A typical neonate with NAS (body weight, 2.9 kg; postnatal age; 5.4 days) had a CL of 3.5 L/kg/hour and elimination half-life of 11 hours. Phenobarbital did not affect the clearance of buprenorphine compared to neonates of similar age and weight. CONCLUSIONS: This is the first study to investigate the population PK of sublingual buprenorphine in neonatal NAS. To our knowledge, this is also the first report to describe the age-dependent changes of buprenorphine PK in this patient population. No buprenorphine dose adjustment is needed for neonates with NAS treated with buprenorphine and concurrent phenobarbital.


Subject(s)
Buprenorphine/pharmacokinetics , Neonatal Abstinence Syndrome/drug therapy , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Administration, Sublingual , Adult , Buprenorphine/therapeutic use , Clinical Trials, Phase I as Topic , Female , Humans , Infant, Newborn , Male , Models, Biological , Randomized Controlled Trials as Topic
5.
Cell Stress Chaperones ; 20(2): 333-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25366568

ABSTRACT

Interaction of the Alzheimer's Aß peptides with the plasma membrane of cells in culture results in chronic increases in cytosolic [Ca(2+)]. Such increases can cause a variety of secondary effects leading to impaired cell growth or cell degeneration. In this investigation, we made a comprehensive study of the changes in cytosolic [Ca(2+)] in single PC12 cells and human neurons stressed by continuous exposure to a medium containing Aß42 for several days. The differential timing and magnitude of the Aß42-induced increase in [Ca(2+)] reveal subpopulations of cells with differential sensitivity to Aß42. These results suggest that the effect produced by Aß on the level of cytosolic [Ca(2+)] depends on the type of cell being monitored. Moreover, the results obtained of using potent inhibitors of Aß cation channels such as Zn(2+) and the small peptide NA7 add further proof to the suggestion that the long-term increases in cytosolic [Ca(2+)] in cells stressed by continuous exposure to Aß is the result of Aß ion channel activity.


Subject(s)
Amyloid beta-Peptides/toxicity , Calcium/metabolism , Ion Channels/metabolism , Ion Transport/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cell Membrane/metabolism , Cells, Cultured , Fluorescent Dyes/chemistry , Humans , Ion Channels/antagonists & inhibitors , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , PC12 Cells , Peptides/pharmacology , Rats , Zinc/pharmacology
6.
Biol Cell ; 102(9): 525-37, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20590528

ABSTRACT

BACKGROUND INFORMATION: Cell fusion is known to underlie key developmental processes in humans and is postulated to contribute to tissue maintenance and even carcinogenesis. The mechanistic details of cell fusion, especially between different cell types, have been difficult to characterize because of the dynamic nature of the process and inadequate means to track fusion products over time. Here we introduce an inducible system for detecting and tracking live cell fusion products in vitro and potentially in vivo. This system is based on BiFC (bimolecular fluorescence complementation) analysis. In this approach, two proteins that can interact with each other are joined to fragments of a fluorescent protein and are expressed in separate cells. The interaction of said proteins after cell fusion produces a fluorescent signal, enabling the identification and tracking of fusion products over time. RESULTS: Long-term tracking of fused p53-deficient cells revealed that hybrid cells were capable of proliferation. In some cases, proliferation was preceded by nuclear fusion and division was asymmetric (69%+/-2% of proliferating hybrids), suggesting chromosomal instability. In addition, asymmetric division following proliferation could give rise to progeny indistinguishable from unfused counterparts. CONCLUSIONS: These results support the possibility that the chromosomal instability characteristic of tumour cells may be incurred as a consequence of cell fusion and suggest that the role of cell fusion in carcinogenesis may have been masked to this point for lack of an inducible method to track cell fusion. In sum, the BiFC-based approach described here allows for comprehensive studies of the mechanism and biological impact of cell fusion in nature.


Subject(s)
Cell Fusion , Chromosomal Instability , Hybrid Cells/physiology , Luminescent Measurements/methods , Microscopy, Fluorescence/methods , Animals , COS Cells , Cell Division , Cell Proliferation , Chlorocebus aethiops , Fluorescence , Hybrid Cells/cytology , Luminescent Proteins/metabolism , Protein Binding , Recombinant Fusion Proteins/metabolism
7.
J Biol Chem ; 280(46): 38879-87, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16186112

ABSTRACT

Despite advances in understanding the role of histone deacetylases (HDACs) in tumorigenesis, the mechanism by which HDAC inhibitors mediate antineoplastic effects remains elusive. Modifications of the histone code alone are not sufficient to account for the antitumor effect of HDAC inhibitors. The present study demonstrates a novel histone acetylation-independent mechanism by which HDAC inhibitors cause Akt dephosphorylation in U87MG glioblastoma and PC-3 prostate cancer cells by disrupting HDAC-protein phosphatase 1 (PP1) complexes. Of four HDAC inhibitors examined, trichostatin A (TSA) and HDAC42 exhibit the highest activity in down-regulating phospho-Akt, followed by suberoylanilide hydroxamic acid, whereas MS-275 shows only a marginal effect at 5 microm. This differential potency parallels the respective activities in inducing tubulin acetylation, a non-histone substrate for HDAC6. Evidence indicates that this Akt dephosphorylation is not mediated through deactivation of upstream kinases or activation of downstream phosphatases. However, the effect of TSA on phospho-Akt can be rescued by PP1 inhibition but not that of protein phosphatase 2A. Immunochemical analyses reveal that TSA blocks specific interactions of PP1 with HDACs 1 and 6, resulting in increased PP1-Akt association. Moreover, we used isozyme-specific small interfering RNAs to confirm the role of HDACs 1 and 6 as key mediators in facilitating Akt dephosphorylation. The selective action of HDAC inhibitors on HDAC-PP1 complexes represents the first example of modulating specific PP1 interactions by small molecule agents. From a clinical perspective, identification of this PP1-facilitated dephosphorylation mechanism underscores the potential use of HDAC inhibitors in lowering the apoptosis threshold for other therapeutic agents through Akt down-regulation.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/chemistry , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Survival , Dose-Response Relationship, Drug , Down-Regulation , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Marine Toxins , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Phosphatidylinositols/chemistry , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Binding , Protein Isoforms , Protein Phosphatase 1 , Protein Phosphatase 2 , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Pyrans/pharmacology , RNA, Small Interfering/metabolism , Serine/chemistry , Signal Transduction , Spiro Compounds/pharmacology , Subcellular Fractions/metabolism , Threonine/chemistry , Time Factors
8.
Blood ; 105(10): 4021-7, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15665113

ABSTRACT

Resistance to the Ableson protein tyrosine (Abl) kinase inhibitor imatinib mesylate has become a critical issue for patients in advanced phases of chronic myelogenous leukemia. Imatinib-resistant tumor cells develop, in part, as a result of point mutations within the Abl kinase domain. As protein kinase B (Akt) plays a pivotal role in Abl oncogene-mediated cell survival, we hypothesize that concurrent inhibition of Akt will sensitize resistant cells to the residual apoptotic activity of imatinib mesylate, thereby overcoming the resistance. Here, we examined the effect of OSU-03012, a celecoxib-derived phosphoinositide-dependent kinase-1 (PDK-1) inhibitor, on imatinib mesylate-induced apoptosis in 2 clinically relevant breakpoint cluster region (Bcr)-Abl mutant cell lines, Ba/F3p210(E255K) and Ba/F3p210(T315I). The 50% inhibitory concentration (IC50) values of imatinib mesylate to inhibit the proliferation of Ba/F3p210(E255K) and Ba/F3p210(T315I) were 14 +/- 4 and 30 +/- 2 microM, respectively. There was no cross-resistance to OSU-03012 in these mutant cells with an IC50 of 5 microM irrespective of mutations. Nevertheless, in the presence of OSU-03012 the susceptibility of these mutant cells to imatinib-induced apoptosis was significantly enhanced. This synergistic action was, at least in part, mediated through the concerted effect on phospho-Akt. Together these data provide a novel therapeutic strategy to overcome imatinib mesylate resistance, especially with the Abl mutant T315I.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Drug Synergism , Enzyme Activation/drug effects , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Imatinib Mesylate , Mice , Mutation/genetics , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects
9.
Biochemistry ; 43(37): 11701-8, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15362854

ABSTRACT

We previously reported that phosphatidylinositol 3,4,5-trisphosphate (PIP(3)), a lipid product of phosphoinositide 3-kinase (PI3K), induced Ca(2+) influx via a noncapacitative pathway in platelets, Jurkat T cells, and RBL-2H3 mast cells. The identity of this Ca(2+) influx system, however, remains unclear. Here, we investigate a potential link between PIP(3)-sensitive Ca(2+) entry and the canonical transient receptor potential (TRPC) channels by developing stable human embryonic kidney (HEK) 293 cell lines expressing TRPC1, TRPC3, TRPC5, and TRPC6. Two lines of evidence support TRPC6 as a putative target by which PIP(3) induces Ca(2+) influx. First, Fura-2 fluorometric Ca(2+) analysis shows the ability of PIP(3) to selectively stimulate [Ca(2+)](i) increase in TRPC6-expressing cells. Second, pull-down analysis indicates specific interactions between biotin-PIP(3) and TRPC6 protein. Our data indicate that PIP(3) activates store-independent Ca(2+) entry in TRPC6 cells via a nonselective cation channel. Although the activating effect of PIP(3) on TRPC6 is reminiscent to that of 1-oleoyl-2-acetyl-sn-glycerol, this activation is not attributable to the diacylglycerol substructure of PIP(3) since other phosphoinositides failed to trigger Ca(2+) responses. The PIP(3)-activated Ca(2+) entry is inhibited by known TRPC6 inhibitors such as Gd(3+) and SKF96365 and is independent of IP(3) production. Furthermore, we demonstrated that TRPC6 overexpression or antisense downregulation significantly alters the amplitude of PIP(3)- and anti-CD3-activated Ca(2+) responses in Jurkat T cells. Consequently, the link between TRPC6 and PIP(3)-mediated Ca(2+) entry provides a framework to account for an intimate relationship between PI3K and PLCgamma in initiating Ca(2+) response to agonist stimulation in T lymphocytes.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Isoforms/metabolism , Biological Transport/physiology , Calcium Channels/genetics , Carbachol/metabolism , Cell Line , Chelating Agents/metabolism , Fluorescent Dyes/metabolism , Fura-2/metabolism , Humans , Molecular Structure , Phosphatidylinositol Phosphates/chemistry , Protein Isoforms/genetics , TRPC Cation Channels , Type C Phospholipases/metabolism
10.
Mol Cancer Ther ; 3(12): 1671-80, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15634661

ABSTRACT

Evidence suggests that the angiogenic endothelium represents an important target through which celecoxib mediates in vivo antitumor effects. Nevertheless, the pharmacologic basis for celecoxib-caused growth inhibition in endothelial cells in vitro remains to be defined. Previously, we showed that celecoxib-induced apoptosis in PC-3 prostate cancer cells was mediated in part through the inhibition of 3-phosphoinositide-dependent kinase-1/Akt signaling. Our present findings show that celecoxib inhibits the growth of human umbilical vein endothelial cells (HUVEC) with pharmacologic profiles reminiscent of those of PC-3 cells. The underlying antiproliferative mechanism, however, may differ between these two cell types considering differences in the functional status of many tumor suppressors, including PTEN, p53, and retinoblastoma, all of which play integral roles in regulating cell cycle progression and survival. From a mechanistic perspective, the genomic integrity of the HUVEC system presents a vastly different intracellular context to examine how celecoxib acts to induce growth inhibition. Here, we obtain evidence that the antiproliferative effects of celecoxib and its close, cyclooxygenase-2-inactive analogue 4-[5-(2,5-dimethylphenyl)-3(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide (DMC) in HUVECs at pharmacologically attainable concentrations (10-20 micromol/L) are attributable to the inhibition of phosphoinositide-dependent kinase-1/Akt signaling and cyclin-dependent kinase. Especially, celecoxib- and DMC-mediated G1 arrest is associated with attenuated retinoblastoma phosphorylation through the inhibition of multiple cyclin-dependent kinases (IC50, 10-35 micromol/L). Moreover, both celecoxib and DMC reduce neovascularization in the chicken chorioallantoic membrane assay, suggesting the involvement of a cyclooxygenase-2-independent mechanism in the in vivo antiangiogenic effects of celecoxib.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Endothelial Cells/drug effects , G1 Phase/drug effects , Pyrazoles/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Umbilical Veins/drug effects , Animals , Celecoxib , Chickens , Chorioallantoic Membrane , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Membrane Proteins , Phosphorylation/drug effects , Prostaglandin-Endoperoxide Synthases , Retinoblastoma/metabolism , Umbilical Veins/metabolism
11.
J Biol Chem ; 278(28): 25872-8, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12738789

ABSTRACT

Among various molecular strategies by which prostate cancer cells evade apoptosis, phosphoinositide 3-kinase (PI3K)/Akt signaling represents a dominant survival pathway. However, different prostate cancer cell lines such as LNCaP and PC-3 display differential sensitivity to the apoptotic effect of PI3K inhibition in serum-free media, reflecting the heterogeneous nature of prostate cancer in apoptosis regulation. Whereas both cell lines are equally susceptible to LY294002-mediated Akt dephosphorylation, only LNCaP cells default to apoptosis, as evidenced by DNA fragmentation and cytochrome c release. In PC-3 cells, Akt deactivation does not lead to cytochrome c release, suggesting that the intermediary signaling pathway is short-circuited by an antiapoptotic factor. This study presents evidence that Bcl-xL overexpression provides a distinct survival mechanism that protects PC-3 cells from apoptotic signals emanating from PI3K inhibition. First, the Bcl-xL/BAD ratio in PC-3 cells is at least an order of magnitude greater than that of LNCaP cells. Second, ectopic expression of Bcl-xL protects LNCaP cells against LY294002-induced apoptosis. Third, antisense down-regulation of Bcl-xL sensitizes PC-3 cells to the apoptotic effect of LY294002. The physiological relevance of this Bcl-xL-mediated survival mechanism is further underscored by the protective effect of serum on LY294002-induced cell death in LNCaP cells, which is correlated with a multifold increase in Bcl-xL expression. In contrast to Bcl-xL, Bcl-2 expression levels are similar in both cells lines, and do not respond to serum stimulation, suggesting that Bcl-2 may not play a physiological role in antagonizing apoptosis signals pertinent to BAD activation in prostate cancer cells.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins/metabolism , Apoptosis , Blotting, Western , Cell Survival , Chromones/pharmacology , Culture Media, Serum-Free/pharmacology , Cytochrome c Group/metabolism , Cytoplasm/metabolism , DNA Fragmentation , Down-Regulation , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Male , Mitochondria/metabolism , Morpholines/pharmacology , Nucleosomes/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation , bcl-X Protein
12.
J Natl Cancer Inst ; 94(23): 1745-57, 2002 Dec 04.
Article in English | MEDLINE | ID: mdl-12464646

ABSTRACT

BACKGROUND: The cyclooxygenase-2 (COX-2) inhibitor celecoxib is thought to act as a chemopreventive agent by sensitizing cancer cells to apoptotic signals. Other COX-2 inhibitors, such as rofecoxib, are two orders of magnitude less potent than celecoxib at inducing apoptosis. The molecular structures of celecoxib and rofecoxib were used as starting points to examine the structural features that contribute to this discrepancy. METHODS: We used a systematic chemical approach to modify the structures of celecoxib and rofecoxib to produce a series of compounds that were tested for their effects on the viability of human prostate cancer PC-3 cells and their ability to induce apoptosis in these cells. Cell viability was measured by the trypan blue dye exclusion assay, and apoptosis was measured by an enzyme-linked immunosorbent assay that quantifies DNA cleavage and by western blot detection of poly(ADP-ribose) polymerase (PARP) cleavage. Western blotting was used to monitor the effects of the compounds on phosphorylation of the serine/threonine kinase Akt and extracellular signal-regulated kinase 2 (ERK2), two components of celecoxib-induced apoptosis signaling. Monte Carlo simulations were used to molecularly model the surface electrostatic potential and electron density of selected compounds. All statistical tests were two-sided. RESULTS: The structural requirements for the induction of apoptosis in PC-3 cells were different from those for COX-2 inhibition. Structure-function analysis indicated that the induction of apoptosis by compounds derived from COX-2 inhibitors required a bulky terminal phenyl ring, a heterocyclic system with negative electrostatic potential, and a benzenesulfonamide or benzenecarboxamide moiety. These derivatives mediated apoptosis by facilitating the dephosphorylation of Akt and ERK2, irrespective of their COX-2 inhibitory activities. CONCLUSION: A new class of compounds that induce apoptosis by targeting Akt and ERK2 signaling pathways in human prostate cancer cells can be synthesized by modifying existing COX-2 inhibitors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anticarcinogenic Agents/chemical synthesis , Apoptosis/drug effects , Enzyme Inhibitors/chemical synthesis , Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Blotting, Western , Celecoxib , Cyclooxygenase 2 , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Lactones/chemical synthesis , Male , Membrane Proteins , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Prostaglandin-Endoperoxide Synthases , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pyrazoles , Signal Transduction/drug effects , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfones , Tumor Cells, Cultured
13.
Biochem J ; 366(Pt 3): 831-7, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12076251

ABSTRACT

Substantial evidence indicates that the cyclo-oxygenase-2 (COX-2) inhibitor celecoxib, a widely prescribed anti-inflammatory agent, displays anti-tumour effect by sensitizing cancer cells to apoptosis. As part of our effort to understand the mechanism by which celecoxib mediates apoptosis in androgen-independent prostate cancer cells, we investigated its effect on intracellular calcium concentration ([Ca(2+)](i)). Digital ratiometric imaging analysis indicates that exposure of PC-3 cells to celecoxib stimulates an immediate [Ca(2+)](i) rise in a dose- and time-dependent manner. Kinetic data show that this Ca(2+) signal arises from internal Ca(2+) release in conjunction with external Ca(2+) influx. Examinations of the biochemical mechanism responsible for this Ca(2+) mobilization indicate that celecoxib blocks endoplasmic reticulum (ER) Ca(2+)-ATPases. Consequently, inhibition of this Ca(2+) reuptake mechanism results in Ca(2+) mobilization from ER stores followed by capacitative calcium entry, leading to [Ca(2+)](i) elevation. In view of the important role of Ca(2+) in apoptosis regulation, this Ca(2+) perturbation may represent part of the signalling mechanism that celecoxib uses to trigger rapid apoptotic death in cancer cells. This Ca(2+)-ATPase inhibitory activity is highly specific for celecoxib, and is not noted with other COX inhibitors tested, including aspirin, ibuprofen, naproxen, rofecoxib (Vioxx), DuP697 and NS398. Moreover, it is noteworthy that this activity is also observed in many other cell lines examined, including A7r5 smooth muscle cells, NIH 3T3 fibroblast cells and Jurkat T cells. Consequently, this Ca(2+)-perturbing effect may provide a plausible link with the reported toxicities of celecoxib such as increased cardiovascular risks in long-term anti-inflammatory therapy.


Subject(s)
Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Cyclooxygenase Inhibitors/pharmacology , Endoplasmic Reticulum/enzymology , Sulfonamides/pharmacology , Adenosine Triphosphatases/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Cardiovascular System/metabolism , Celecoxib , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Heparin/pharmacology , Humans , Jurkat Cells , Pyrazoles , Ryanodine/metabolism , Spectrometry, Fluorescence , Time Factors , Tumor Cells, Cultured
14.
J Natl Cancer Inst ; 94(8): 585-91, 2002 Apr 17.
Article in English | MEDLINE | ID: mdl-11959891

ABSTRACT

BACKGROUND: The antitumor activity of cyclooxygenase-2 (COX-2) inhibitors is thought to involve COX-2 enzyme inhibition and apoptosis induction, but it is unclear whether COX-2 inhibition is required for apoptosis. Different COX-2 inhibitors have similar IC(50) values (concentration for 50% inhibition) for COX-2 inhibition but differ considerably in their abilities to induce apoptosis, suggesting the involvement of a COX-2-independent pathway in apoptosis. To test this hypothesis, we investigated the effect of COX-2 depletion on apoptosis and performed a structure-activity analysis of the COX-2 inhibitor celecoxib in the androgen-independent prostate cancer cell line PC-3. METHODS: Tetracycline-inducible (Tet-On) COX-2 antisense clones were isolated to assess the effect of COX-2 expression on cell viability and sensitivity to apoptosis induced by COX-2 inhibitors. Untreated Tet-On clones differentially expressed COX-2, and doxycycline-treated clones were depleted of COX-2. We synthesized and characterized various celecoxib derivatives with various COX-2 inhibitory activities and determined their apoptotic activity in PC-3 cells. Apoptosis was assessed with four tests. RESULTS: In contrast to the effect of COX-2 inhibitors, which induced apoptosis, COX-2 depletion did not induce cell death. Susceptibility to COX-2 inhibitor-induced apoptosis was independent of the level of COX-2 expression. Structure-activity analysis found no correlation between apoptosis induction and COX-2 inhibition. Some celecoxib derivatives that lacked COX-2 inhibitory activity facilitated apoptosis and vice versa. Moreover, celecoxib and apoptosis-active celecoxib derivatives mediated cell death by inhibiting the same pathway. CONCLUSION: We have dissociated the apoptosis-inducing activity from the COX-2 inhibitory activity by structural modifications of the COX-2 inhibitor celecoxib. This separation of activities may provide a molecular basis for the development of new classes of apoptosis-inducing agents.


Subject(s)
Apoptosis , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Prostatic Neoplasms/enzymology , Antineoplastic Agents/pharmacology , Blotting, Western , Celecoxib , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Indoles/pharmacology , Inhibitory Concentration 50 , Lactones/pharmacology , Male , Membrane Proteins , Models, Chemical , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Pyrazoles , Structure-Activity Relationship , Sulfonamides/pharmacology , Sulfones , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...