PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 2464912-4 1988 An analysis of fluvoxamine minus placebo difference scores showed a significant correlation between memory functioning and CSF 5HIAA levels. Fluvoxamine 15-26 colony stimulating factor 2 Homo sapiens 123-126 2464912-5 1988 Alcohol amnestic syndrome patients who had the highest blood levels of fluvoxamine demonstrated the largest changes in CSF 5HIAA and improvement in memory performance under fluvoxamine. Fluvoxamine 71-82 colony stimulating factor 2 Homo sapiens 119-122 33602262-7 2021 RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1beta (IL-1beta). Fluvoxamine 86-97 nitric oxide synthase 2 Homo sapiens 258-289 6134546-9 1983 the PRL-releasing effect of an additional acute fluvoxamine administration (same dose) was abolished after 4 days maintenance treatment. Fluvoxamine 48-59 prolactin Rattus norvegicus 4-7 6777458-8 1980 Psychometric tests demonstrated after 50 and 75 mg clovoxamine and 75 mg fluvoxamine an increase in attention, attention variability, concentration, CFF and after-effect in the Archimedean Spiral (indicating central activation), further an improvement in mood and affectivity as compared with placebo, while 125 mg clovoxamine and 75 mg imipramine produced an increase in reaction time, deterioration of mood and affect and psychomotor activity. Fluvoxamine 73-84 host cell factor C1 Homo sapiens 149-152 33524820-0 2021 Multi-spectroscopic and molecular docking studies for binding interaction between fluvoxamine and human serum albumin. Fluvoxamine 82-93 albumin Homo sapiens 104-117 33524820-1 2021 In the present study, different spectroscopic techniques have been used to study the binding interaction between the antidepressant drug fluvoxamine and human serum albumin under simulated physiological conditions (pH 7.4). Fluvoxamine 137-148 albumin Homo sapiens 159-172 33524820-3 2021 The obtained results revealed that the formation of a complex between human serum albumin and fluvoxamine was responsible for quenching the native fluorescence of human serum albumin. Fluvoxamine 94-105 albumin Homo sapiens 76-89 33524820-3 2021 The obtained results revealed that the formation of a complex between human serum albumin and fluvoxamine was responsible for quenching the native fluorescence of human serum albumin. Fluvoxamine 94-105 albumin Homo sapiens 169-182 33524820-4 2021 The results indicated that the quenching mechanism between human serum albumin and fluvoxamine was static. Fluvoxamine 83-94 albumin Homo sapiens 65-78 33524820-7 2021 From the molecular docking results, it could be deduced that fluvoxamine was inserted into sub-domain II A (site I) of human serum albumin and led to a slight change in human serum albumin conformation. Fluvoxamine 61-72 albumin Homo sapiens 125-138 33524820-7 2021 From the molecular docking results, it could be deduced that fluvoxamine was inserted into sub-domain II A (site I) of human serum albumin and led to a slight change in human serum albumin conformation. Fluvoxamine 61-72 albumin Homo sapiens 175-188 34049262-0 2021 Tuning the activity of known drugs via the introduction of halogen atoms, a case study of SERT ligands - Fluoxetine and fluvoxamine. Fluvoxamine 120-131 solute carrier family 6 member 4 Homo sapiens 90-94 34049262-3 2021 To elucidate the role of halogen atoms in the binding of SSRIs to SERT, we designed a series of 22 fluoxetine and fluvoxamine analogs substituted with fluorine, chlorine, bromine, and iodine atoms, differently arranged on the phenyl ring. Fluvoxamine 114-125 solute carrier family 6 member 4 Homo sapiens 66-70 33646885-5 2021 Fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), reduced the BMP-4-stimulated OPG release, whereas a selective and specific norepinephrine reuptake inhibitor, reboxetine failed to affect the OPG release. Fluvoxamine 0-11 bone morphogenetic protein 4 Mus musculus 74-79 33646885-5 2021 Fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), reduced the BMP-4-stimulated OPG release, whereas a selective and specific norepinephrine reuptake inhibitor, reboxetine failed to affect the OPG release. Fluvoxamine 0-11 tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin) Mus musculus 91-94 33646885-10 2021 Both fluvoxamine and sertraline also inhibited the BMP-4-elicited phosphorylation of SMAD1/5/8. Fluvoxamine 5-16 bone morphogenetic protein 4 Mus musculus 51-56 33646885-10 2021 Both fluvoxamine and sertraline also inhibited the BMP-4-elicited phosphorylation of SMAD1/5/8. Fluvoxamine 5-16 SMAD family member 1 Mus musculus 85-94 33602262-7 2021 RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1beta (IL-1beta). Fluvoxamine 86-97 nitric oxide synthase 2 Homo sapiens 291-295 33602262-7 2021 RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1beta (IL-1beta). Fluvoxamine 86-97 interleukin 6 Homo sapiens 344-357 33602262-7 2021 RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1beta (IL-1beta). Fluvoxamine 86-97 interleukin 6 Homo sapiens 359-363 33602262-7 2021 RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1beta (IL-1beta). Fluvoxamine 86-97 interleukin 1 beta Homo sapiens 372-389 33602262-7 2021 RESULTS: All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1beta (IL-1beta). Fluvoxamine 86-97 interleukin 1 alpha Homo sapiens 391-399 33897064-4 2021 Objective: The objective of this study was to investigate the effect of polymorphisms of the CYP3A4, CYP2C9, CYP3A5, ABCB1, CYP2C19, SCL6A4, and 5-HTR2A genes on the concentration/dose indicator of fluvoxamine and on the CYP3A expression level obtained by measuring the miR-27b plasma concentration levels in patients suffering from a recurrent depressive disorder. Fluvoxamine 198-209 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 93-98 33140216-10 2021 Thus, we investigated the effect of fluvoxamine, a Sigma-1 receptor agonist, on Ito and ICa-L. Fluvoxamine enhanced Ito and altered its current kinetics, as shown by acceleration of activation and recovery from inactivation. Fluvoxamine 95-106 sigma non-opioid intracellular receptor 1 Rattus norvegicus 51-67 33897064-3 2021 Previous research revealed that CYP2D6 is involved in the metabolism of fluvoxamine, the activity of which is highly dependent on the polymorphism of the gene encoding it. Fluvoxamine 72-83 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-38 33834720-14 2021 The study shows the efficacy of APAH, fluvoxamine and milnacipran in the presence of CS. Fluvoxamine 38-49 citrate synthase Homo sapiens 85-87 33488945-8 2021 Fluvoxamine stimulation of Sig1R restored autophagic flux in cardiac fibroblasts, indicating that Sig1R appears to play a protective role in the activation of cardiac fibroblasts by inhibiting the IRE1 pathway and restoring autophagic flux. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 27-32 33488945-8 2021 Fluvoxamine stimulation of Sig1R restored autophagic flux in cardiac fibroblasts, indicating that Sig1R appears to play a protective role in the activation of cardiac fibroblasts by inhibiting the IRE1 pathway and restoring autophagic flux. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 98-103 31180700-0 2020 BDNF association study with obsessive-compulsive disorder, its clinical characteristics, and response to fluvoxamine-treatment in Iranian patients. Fluvoxamine 105-116 brain derived neurotrophic factor Homo sapiens 0-4 33302490-0 2020 A Physiologically-Based Pharmacokinetic (PBPK) Model Network for the Prediction of CYP1A2 and CYP2C19 Drug-Drug-Gene Interactions with Fluvoxamine, Omeprazole, S-mephenytoin, Moclobemide, Tizanidine, Mexiletine, Ethinylestradiol, and Caffeine. Fluvoxamine 135-146 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 83-89 33302490-0 2020 A Physiologically-Based Pharmacokinetic (PBPK) Model Network for the Prediction of CYP1A2 and CYP2C19 Drug-Drug-Gene Interactions with Fluvoxamine, Omeprazole, S-mephenytoin, Moclobemide, Tizanidine, Mexiletine, Ethinylestradiol, and Caffeine. Fluvoxamine 135-146 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 94-101 33002595-7 2020 Oppositely, fluvoxamine and sertraline, agents belonging to the class of selective serotonin reuptake inhibitor, upregulated the PGE1-stimulated release of both OPG and IL-6. Fluvoxamine 12-23 tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin) Mus musculus 161-164 33002595-7 2020 Oppositely, fluvoxamine and sertraline, agents belonging to the class of selective serotonin reuptake inhibitor, upregulated the PGE1-stimulated release of both OPG and IL-6. Fluvoxamine 12-23 interleukin 6 Mus musculus 169-173 33002595-10 2020 SB203880, an inhibitor of p38 MAP kinase, suppressed the amplifying effects by duloxetine or fluvoxamine on the PGE1-stimulated release of OPG and IL-6. Fluvoxamine 93-104 tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin) Mus musculus 139-142 33002595-10 2020 SB203880, an inhibitor of p38 MAP kinase, suppressed the amplifying effects by duloxetine or fluvoxamine on the PGE1-stimulated release of OPG and IL-6. Fluvoxamine 93-104 interleukin 6 Mus musculus 147-151 33158023-8 2020 Furthermore, FLV significantly enhanced the levels of glutathione (GSH) and peroxiredoxin 1 (PRDX1) and caused a significant reduction in 3-nitrotyrosine (3-NT) levels, the effects of which were abolished by co-treatment with BD1063. Fluvoxamine 13-16 peroxiredoxin 1 Rattus norvegicus 76-91 33158023-8 2020 Furthermore, FLV significantly enhanced the levels of glutathione (GSH) and peroxiredoxin 1 (PRDX1) and caused a significant reduction in 3-nitrotyrosine (3-NT) levels, the effects of which were abolished by co-treatment with BD1063. Fluvoxamine 13-16 peroxiredoxin 1 Rattus norvegicus 93-98 32629001-3 2020 The S1R agonist fluvoxamine was injected intraperitoneally from the second week to the last week for 21 days in total, and the effects were evaluated by patch clamp, western blot analysis, and Masson staining. Fluvoxamine 16-27 sigma non-opioid intracellular receptor 1 Rattus norvegicus 4-7 32660197-8 2020 Furthermore, in comparison to Sig-1R-/- mice, fluvoxamine significantly increased serum levels of AST, ALT, MPO, and LDH in wildtype animals in a dose-dependent manner at 6 h after IRI. Fluvoxamine 46-57 solute carrier family 17 (anion/sugar transporter), member 5 Mus musculus 98-101 32660197-8 2020 Furthermore, in comparison to Sig-1R-/- mice, fluvoxamine significantly increased serum levels of AST, ALT, MPO, and LDH in wildtype animals in a dose-dependent manner at 6 h after IRI. Fluvoxamine 46-57 glutamic pyruvic transaminase, soluble Mus musculus 103-106 32660197-8 2020 Furthermore, in comparison to Sig-1R-/- mice, fluvoxamine significantly increased serum levels of AST, ALT, MPO, and LDH in wildtype animals in a dose-dependent manner at 6 h after IRI. Fluvoxamine 46-57 myeloperoxidase Mus musculus 108-111 31608449-8 2019 In the neurochemical tests, both thymol and fluvoxamine restored BDNF levels, improving the depressive condition. Fluvoxamine 44-55 brain derived neurotrophic factor Mus musculus 65-69 32802593-3 2020 Fluvoxamine inhibits many cytochrome P450 enzyme (CYP) including CYP2C9, which metabolizes azilsartan. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 31795773-6 2020 As CYP3A4 is an important metabolic pathway for all DOACs except dabigatran, it appears reasonable to recommend avoiding the co-prescription of fluoxetine and fluvoxamine (weak to moderate CYP3A4 inhibitors) and St John"s wort (CYP3A4 inducer). Fluvoxamine 159-170 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 3-9 31172535-5 2019 Following a single oral dose of 3 mg, avadomide exposure when coadministered with the CYP1A2 inhibitor fluvoxamine was 154.81% and 107.59% of that when administered alone, for area under the plasma concentration-time curve from time 0 to infinity (AUC0-inf ) and maximum observed plasma concentration (Cmax ), respectively. Fluvoxamine 103-114 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 86-92 31810748-0 2020 The antidepressant-like effects of fluvoxamine in mice involve the mTOR signaling in the hippocampus and prefrontal cortex. Fluvoxamine 35-46 mechanistic target of rapamycin kinase Mus musculus 67-71 31810748-3 2020 Therefore, this study aims to evaluate whether mTOR underlies the antidepressant-like effects of fluvoxamine. Fluvoxamine 97-108 mechanistic target of rapamycin kinase Mus musculus 47-51 31810748-7 2020 It was found that fluvoxamine treatment fully reversed the effects of CUMS on the mTOR signaling in the hippocampus and PFC, and the usage of rapamycin significantly prevented the antidepressant-like effects of fluvoxamine in the CUMS model of depression. Fluvoxamine 18-29 mechanistic target of rapamycin kinase Mus musculus 82-86 31810748-8 2020 Taken together, the mTOR system is involved in the antidepressant mechanisms of fluvoxamine. Fluvoxamine 80-91 mechanistic target of rapamycin kinase Mus musculus 20-24 31493481-10 2019 SIGNIFICANCE: Our findings indicated that S1R inhibition contributed to atrial electrical remodeling, cardiac autonomic remodeling and atrial fibrosis, which could be attenuated by fluvoxamine, thus providing new insights into the relationship between the S1R and AF. Fluvoxamine 181-192 sigma non-opioid intracellular receptor 1 Rattus norvegicus 42-45 31807102-13 2019 Conclusion: Fluvoxamine improved PSG parameters and ameliorated complaints of insomnia simultaneously during this 8-week study. Fluvoxamine 12-23 pregnancy specific beta-1-glycoprotein 5 Homo sapiens 33-36 31493481-10 2019 SIGNIFICANCE: Our findings indicated that S1R inhibition contributed to atrial electrical remodeling, cardiac autonomic remodeling and atrial fibrosis, which could be attenuated by fluvoxamine, thus providing new insights into the relationship between the S1R and AF. Fluvoxamine 181-192 sigma non-opioid intracellular receptor 1 Rattus norvegicus 256-259 30902656-2 2019 Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. Fluvoxamine 0-11 nerve growth factor Rattus norvegicus 21-40 31182321-2 2019 Smoking induces CYP1A2 thereby increasing clozapine metabolism whereas fluvoxamine inhibits CYP1A2. Fluvoxamine 71-82 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 92-98 30902656-2 2019 Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. Fluvoxamine 0-11 nerve growth factor Rattus norvegicus 42-45 30902656-2 2019 Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 93-109 30902656-2 2019 Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. Fluvoxamine 211-222 nerve growth factor Rattus norvegicus 21-40 30902656-2 2019 Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. Fluvoxamine 211-222 nerve growth factor Rattus norvegicus 42-45 30902656-2 2019 Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. Fluvoxamine 211-222 sigma non-opioid intracellular receptor 1 Rattus norvegicus 93-109 29035919-2 2018 For compulsive-like nest-building behavior, dose-dependent attenuation of nesting by fluvoxamine was observed for the BIG1 compulsive-like strain during the first hour after administration. Fluvoxamine 85-96 contactin 3 Mus musculus 118-122 30762305-0 2019 Physiologically-Based Pharmacokinetic Models for CYP1A2 Drug-Drug Interaction Prediction: A Modeling Network of Fluvoxamine, Theophylline, Caffeine, Rifampicin, and Midazolam. Fluvoxamine 112-123 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 49-55 30762305-1 2019 This study provides whole-body physiologically-based pharmacokinetic models of the strong index cytochrome P450 (CYP)1A2 inhibitor and moderate CYP3A4 inhibitor fluvoxamine and of the sensitive CYP1A2 substrate theophylline. Fluvoxamine 161-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 144-150 30902567-8 2019 Conversely, while fluvoxamine reduced omeprazole metabolism in subjects carrying the CYP2C19*1 allele, it had no impact on omeprazole pharmacokinetics in subjects without this allele. Fluvoxamine 18-29 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 85-92 30730837-0 2019 Selective serotonin reuptake inhibitor fluvoxamine ameliorates stress- and NSAID-induced peptic ulcer possibly by involving Hsp70. Fluvoxamine 39-50 heat shock protein 1B Mus musculus 124-129 30730837-3 2019 However, there is lack of evidence that fluvoxamine recruits Hsp70 to affect stress-induced gastric ulcer. Fluvoxamine 40-51 heat shock protein 1B Mus musculus 61-66 30730837-12 2019 Moreover, fluvoxamine further increased the expression of Hsp70 in the gastric tissue of stress- and indomethacin-treated mice. Fluvoxamine 10-21 heat shock protein 1B Mus musculus 58-63 30730837-14 2019 In addition, the present study suggests the possible involvement of Hsp70 in the amelioration of gastric ulcer by fluvoxamine. Fluvoxamine 114-125 heat shock protein 1B Mus musculus 68-73 30820426-0 2019 Evaluation of the Effect of Antidepressant Drug, Fluvoxamine, on Cyclooxygenase-2 Protein Expression in Lipopolysaccharide-stimulated Macrophages. Fluvoxamine 49-60 prostaglandin-endoperoxide synthase 2 Homo sapiens 65-81 30820426-3 2019 In the line of the indicated study, we sought to evaluate the effect of fluvoxamine on the expression of some inflammatory mediators such as cyclooxygenase-2 (COX-2). Fluvoxamine 72-83 prostaglandin-endoperoxide synthase 2 Homo sapiens 141-157 30820426-3 2019 In the line of the indicated study, we sought to evaluate the effect of fluvoxamine on the expression of some inflammatory mediators such as cyclooxygenase-2 (COX-2). Fluvoxamine 72-83 prostaglandin-endoperoxide synthase 2 Homo sapiens 159-164 30820426-6 2019 Results: The expression of COX-2 significantly decreased by fluvoxamine in U937 macrophages. Fluvoxamine 60-71 prostaglandin-endoperoxide synthase 2 Homo sapiens 27-32 30543144-0 2019 Effects of fluvoxamine on nerve growth factor-induced neurite outgrowth inhibition by dexamethasone in PC12 cells. Fluvoxamine 11-22 nerve growth factor Rattus norvegicus 26-45 30543144-1 2019 In the present study, we examined the effects of fluvoxamine on nerve growth factor (NGF)-induced neurite outgrowth inhibition by dexamethasone (DEX) in PC12 cells. Fluvoxamine 49-60 nerve growth factor Rattus norvegicus 64-83 30543144-1 2019 In the present study, we examined the effects of fluvoxamine on nerve growth factor (NGF)-induced neurite outgrowth inhibition by dexamethasone (DEX) in PC12 cells. Fluvoxamine 49-60 nerve growth factor Rattus norvegicus 85-88 30543144-2 2019 Fluvoxamine increased NGF-induced neurite outgrowth. Fluvoxamine 0-11 nerve growth factor Rattus norvegicus 22-25 30543144-3 2019 Compared with co-treatment with NGF and fluvoxamine, p-Akt levels were higher than the values without fluvoxamine. Fluvoxamine 40-51 AKT serine/threonine kinase 1 Rattus norvegicus 55-58 30543144-5 2019 Fluvoxamine concentration-dependently improved NGF-induced neurite outgrowth inhibition by DEX. Fluvoxamine 0-11 nerve growth factor Rattus norvegicus 47-50 30543144-6 2019 Fluvoxamine also improved the decrease in the NGF-induced p-Akt level caused by DEX. Fluvoxamine 0-11 nerve growth factor Rattus norvegicus 46-49 30543144-6 2019 Fluvoxamine also improved the decrease in the NGF-induced p-Akt level caused by DEX. Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 60-63 30543144-7 2019 Interestingly, the sigma-1 receptor antagonist NE-100 blocked the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX. Fluvoxamine 89-100 sigma non-opioid intracellular receptor 1 Rattus norvegicus 19-35 30543144-7 2019 Interestingly, the sigma-1 receptor antagonist NE-100 blocked the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX. Fluvoxamine 89-100 nerve growth factor Rattus norvegicus 104-107 30543144-9 2019 These results indicate that the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX may be attributable to the phosphorylation of Akt and the sigma-1 receptor. Fluvoxamine 55-66 nerve growth factor Rattus norvegicus 70-73 30543144-9 2019 These results indicate that the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX may be attributable to the phosphorylation of Akt and the sigma-1 receptor. Fluvoxamine 55-66 AKT serine/threonine kinase 1 Rattus norvegicus 164-167 30543144-9 2019 These results indicate that the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX may be attributable to the phosphorylation of Akt and the sigma-1 receptor. Fluvoxamine 55-66 sigma non-opioid intracellular receptor 1 Rattus norvegicus 176-192 30728287-8 2019 These findings could have substantial clinical implications, as we further find that fluvoxamine, an antidepressant therapeutic with high affinity for S1R, protects mice from lethal septic shock and dampens the inflammatory response in human blood leukocytes. Fluvoxamine 85-96 sigma non-opioid intracellular receptor 1 Mus musculus 151-154 29988737-0 2018 Effects of CYP2D6 genetic polymorphisms on the efficacy and safety of fluvoxamine in patients with depressive disorder and comorbid alcohol use disorder. Fluvoxamine 70-81 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 11-17 29988737-2 2018 CYP2D6 is involved in the biotransformation of fluvoxamine. Fluvoxamine 47-58 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 29988737-4 2018 Objective: The primary objective of our study was to investigate the effects of CYP2D6 genetic polymorphisms on the efficacy and safety of fluvoxamine in patients with depressive disorder and comorbid alcohol use disorder, in order to develop the algorithms of optimization of fluvoxamine therapy for reducing the risk of dose-dependent undesirable side effects and pharmacoresistance. Fluvoxamine 139-150 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 80-86 29988737-4 2018 Objective: The primary objective of our study was to investigate the effects of CYP2D6 genetic polymorphisms on the efficacy and safety of fluvoxamine in patients with depressive disorder and comorbid alcohol use disorder, in order to develop the algorithms of optimization of fluvoxamine therapy for reducing the risk of dose-dependent undesirable side effects and pharmacoresistance. Fluvoxamine 277-288 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 80-86 29988737-9 2018 Conclusion: This study demonstrated the lower efficacy and safety of fluvoxamine in patients with depressive disorder and comorbid alcohol use disorders with GA genotype in CYP2D6 1846G>A polymorphic marker. Fluvoxamine 69-80 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 173-179 29785111-11 2018 Conclusion: HTR2A haplotypes are associated with OCD and its treatment response with a fluvoxamine in Iranian patients. Fluvoxamine 87-98 5-hydroxytryptamine receptor 2A Homo sapiens 12-17 29382554-12 2018 Western blot analysis revealed that the expression level of 5-HT2A receptor was specifically decreased in the prefrontal cortex of mice that had been administered yokukansan and fluvoxamine. Fluvoxamine 178-189 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 60-75 28993551-11 2017 DDIs perpetrated by FLV and itraconazole were successfully predicted by use of the present method where two DDI predictors [perpetrator-specific inhibitory activities toward CYP isoforms (pAi, CYPs) and victim-specific fractional CYP-isoform contributions to the clearance (vfm, CYPs)] are determined successively as shown in the graphical abstract. Fluvoxamine 20-23 serpin family E member 1 Homo sapiens 188-191 29155491-6 2018 The CYP3A4-selective inhibitor ketoconazole (2 muM) impaired CLZ N-oxide formation in all 14 of the livers used in inhibition studies (>=50% inhibition) while the CYP1A2-selective inhibitor fluvoxamine (10 muM) decreased norCLZ formation in nine. Fluvoxamine 193-204 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 4-10 29155491-6 2018 The CYP3A4-selective inhibitor ketoconazole (2 muM) impaired CLZ N-oxide formation in all 14 of the livers used in inhibition studies (>=50% inhibition) while the CYP1A2-selective inhibitor fluvoxamine (10 muM) decreased norCLZ formation in nine. Fluvoxamine 193-204 latexin Homo sapiens 47-50 29155491-8 2018 Similarly, fluvoxamine (10 muM) readily inhibited CLZ oxidation in seven livers with high CYP1A2-mediated 7-ethoxyresorufin O-deethylation activity (at or above the median) and three livers with lower intrinsic CYP1A2 activity. Fluvoxamine 11-22 latexin Homo sapiens 27-30 29155491-8 2018 Similarly, fluvoxamine (10 muM) readily inhibited CLZ oxidation in seven livers with high CYP1A2-mediated 7-ethoxyresorufin O-deethylation activity (at or above the median) and three livers with lower intrinsic CYP1A2 activity. Fluvoxamine 11-22 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 90-96 29155491-8 2018 Similarly, fluvoxamine (10 muM) readily inhibited CLZ oxidation in seven livers with high CYP1A2-mediated 7-ethoxyresorufin O-deethylation activity (at or above the median) and three livers with lower intrinsic CYP1A2 activity. Fluvoxamine 11-22 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 211-217 29379174-2 2018 Here we report X-ray structures of engineered thermostable variants of the human serotonin transporter bound to the antidepressants sertraline, fluvoxamine, and paroxetine. Fluvoxamine 144-155 solute carrier family 6 member 4 Homo sapiens 81-102 29101463-2 2018 This study was performed to investigate the potential drug-drug interaction of dovitinib with the CYP1A2 inhibitor fluvoxamine in patients with advanced solid tumors. Fluvoxamine 115-126 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 98-104 28688742-8 2018 Clozapine-fluvoxamine combined treatment significantly attenuated the increments in body weight, insulin resistance, and levels of insulin, glucose, and triglycerides compared with clozapine monotherapy. Fluvoxamine 10-21 insulin Homo sapiens 97-104 28688742-8 2018 Clozapine-fluvoxamine combined treatment significantly attenuated the increments in body weight, insulin resistance, and levels of insulin, glucose, and triglycerides compared with clozapine monotherapy. Fluvoxamine 10-21 insulin Homo sapiens 131-138 29614681-4 2018 Fluvoxamine is a selective serotonin reuptake inhibitor and a potent SIGMAR1 agonist. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Mus musculus 69-76 28815595-6 2017 Escitalopram and fluvoxamine (100 nM to 1 muM) reversibly inhibited nAChR currents. Fluvoxamine 17-28 latexin Homo sapiens 42-45 28815595-6 2017 Escitalopram and fluvoxamine (100 nM to 1 muM) reversibly inhibited nAChR currents. Fluvoxamine 17-28 cholinergic receptor nicotinic alpha 4 subunit Homo sapiens 68-73 28815595-8 2017 Block of nAChR currents by fluvoxamine and NE-100 was not additive suggesting a common site of action. Fluvoxamine 27-38 cholinergic receptor nicotinic alpha 4 subunit Homo sapiens 9-14 28529139-2 2017 Some SSRIs such as fluvoxamine have agonistic activity towards for the sigma1 receptor, but it is not known whether the effect on the receptor plays a key role in the pharmacological effects. Fluvoxamine 19-30 sigma non-opioid intracellular receptor 1 Mus musculus 71-86 28529139-4 2017 We also suggest that the anti-anhedonic effect of fluvoxamine is mediated by combined activation of the 5-HT1A and sigma1 receptors and it is associated with activation of prefrontal dopaminergic system. Fluvoxamine 50-61 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 104-110 28243095-0 2017 Influence of fluvoxamine on plasma interleukin-6 or clinical improvement in patients with major depressive disorder. Fluvoxamine 13-24 interleukin 6 Homo sapiens 35-48 28410959-2 2017 This study examined the role of brain derived neurotrophic factor (BDNF)-cAMP response element binding (CREB) protein signaling pathways, including protein kinase B (AKT), glycogen synthase kinase (GSK)-3beta and related molecules in the molecular response to haloperidol, fluvoxamine, combined haloperidol+fluvoxamine and clozapine treatments in rat frontal cortex, hippocampus and primary cortical neuronal cultures. Fluvoxamine 273-284 brain-derived neurotrophic factor Rattus norvegicus 67-71 28410959-2 2017 This study examined the role of brain derived neurotrophic factor (BDNF)-cAMP response element binding (CREB) protein signaling pathways, including protein kinase B (AKT), glycogen synthase kinase (GSK)-3beta and related molecules in the molecular response to haloperidol, fluvoxamine, combined haloperidol+fluvoxamine and clozapine treatments in rat frontal cortex, hippocampus and primary cortical neuronal cultures. Fluvoxamine 273-284 cAMP responsive element binding protein 1 Rattus norvegicus 104-108 28410959-2 2017 This study examined the role of brain derived neurotrophic factor (BDNF)-cAMP response element binding (CREB) protein signaling pathways, including protein kinase B (AKT), glycogen synthase kinase (GSK)-3beta and related molecules in the molecular response to haloperidol, fluvoxamine, combined haloperidol+fluvoxamine and clozapine treatments in rat frontal cortex, hippocampus and primary cortical neuronal cultures. Fluvoxamine 307-318 brain-derived neurotrophic factor Rattus norvegicus 67-71 28410959-2 2017 This study examined the role of brain derived neurotrophic factor (BDNF)-cAMP response element binding (CREB) protein signaling pathways, including protein kinase B (AKT), glycogen synthase kinase (GSK)-3beta and related molecules in the molecular response to haloperidol, fluvoxamine, combined haloperidol+fluvoxamine and clozapine treatments in rat frontal cortex, hippocampus and primary cortical neuronal cultures. Fluvoxamine 307-318 cAMP responsive element binding protein 1 Rattus norvegicus 104-108 28410959-3 2017 The effect of fluvoxamine augmentation on BDNF-CREB pathways in peripheral mononuclear cells (PMC s) of medicated schizophrenia patients was also studied. Fluvoxamine 14-25 brain derived neurotrophic factor Homo sapiens 42-46 28410959-3 2017 The effect of fluvoxamine augmentation on BDNF-CREB pathways in peripheral mononuclear cells (PMC s) of medicated schizophrenia patients was also studied. Fluvoxamine 14-25 cAMP responsive element binding protein 1 Homo sapiens 47-51 28410959-4 2017 Chronic haloperidol (1mg/kg) +fluvoxamine (10mg/kg) treatment increased TrkB receptor and BDNF expression levels, and the phosphorylation of AKT/CREB/GSK-3beta, compared to the individual drugs in rat brain. Fluvoxamine 30-41 brain-derived neurotrophic factor Rattus norvegicus 90-94 28410959-4 2017 Chronic haloperidol (1mg/kg) +fluvoxamine (10mg/kg) treatment increased TrkB receptor and BDNF expression levels, and the phosphorylation of AKT/CREB/GSK-3beta, compared to the individual drugs in rat brain. Fluvoxamine 30-41 AKT serine/threonine kinase 1 Rattus norvegicus 141-144 28410959-4 2017 Chronic haloperidol (1mg/kg) +fluvoxamine (10mg/kg) treatment increased TrkB receptor and BDNF expression levels, and the phosphorylation of AKT/CREB/GSK-3beta, compared to the individual drugs in rat brain. Fluvoxamine 30-41 cAMP responsive element binding protein 1 Rattus norvegicus 145-149 28410959-4 2017 Chronic haloperidol (1mg/kg) +fluvoxamine (10mg/kg) treatment increased TrkB receptor and BDNF expression levels, and the phosphorylation of AKT/CREB/GSK-3beta, compared to the individual drugs in rat brain. Fluvoxamine 30-41 glycogen synthase kinase 3 beta Rattus norvegicus 150-159 28410959-6 2017 In primary neuronal cell cultures, pretreatment with a selective PI3K inhibitor abolished the haloperidol+fluvoxamine-induced phosphorylation of AKT and GSK-3beta, but did not affect the upregulation of CREB phosphorylation. Fluvoxamine 106-117 AKT serine/threonine kinase 1 Rattus norvegicus 145-148 28410959-6 2017 In primary neuronal cell cultures, pretreatment with a selective PI3K inhibitor abolished the haloperidol+fluvoxamine-induced phosphorylation of AKT and GSK-3beta, but did not affect the upregulation of CREB phosphorylation. Fluvoxamine 106-117 glycogen synthase kinase 3 beta Rattus norvegicus 153-162 28410959-6 2017 In primary neuronal cell cultures, pretreatment with a selective PI3K inhibitor abolished the haloperidol+fluvoxamine-induced phosphorylation of AKT and GSK-3beta, but did not affect the upregulation of CREB phosphorylation. Fluvoxamine 106-117 cAMP responsive element binding protein 1 Rattus norvegicus 203-207 28687730-3 2017 Nanomolar concentrations of fluvoxamine significantly increased cell viability and proliferation of neural stem cells (NSCs) through increasing mRNA expression of Notch1, Hes1 and Ki-67, and protein levels of NICD. Fluvoxamine 28-39 notch receptor 1 Rattus norvegicus 163-169 28687730-3 2017 Nanomolar concentrations of fluvoxamine significantly increased cell viability and proliferation of neural stem cells (NSCs) through increasing mRNA expression of Notch1, Hes1 and Ki-67, and protein levels of NICD. Fluvoxamine 28-39 hes family bHLH transcription factor 1 Rattus norvegicus 171-175 28687730-6 2017 Moreover, fluvoxamine treated EAE rats showed a decrease in IFN-gamma serum levels and an increase in IL-4, pro- and anti-inflammatory cytokines respectively, compared to untreated EAE rats. Fluvoxamine 10-21 interferon gamma Rattus norvegicus 60-69 28687730-6 2017 Moreover, fluvoxamine treated EAE rats showed a decrease in IFN-gamma serum levels and an increase in IL-4, pro- and anti-inflammatory cytokines respectively, compared to untreated EAE rats. Fluvoxamine 10-21 interleukin 4 Rattus norvegicus 102-106 28687730-7 2017 Furthermore, immune cell infiltration and demyelination plaque significantly decreased in spinal cords of fluvoxamine-treated rats, which was accompanied by an increase in protein expression of MBP and GFAP positive cells and a decrease in lactate serum levels, a new biomarker of MS progression. Fluvoxamine 106-117 myelin basic protein Rattus norvegicus 194-197 28687730-7 2017 Furthermore, immune cell infiltration and demyelination plaque significantly decreased in spinal cords of fluvoxamine-treated rats, which was accompanied by an increase in protein expression of MBP and GFAP positive cells and a decrease in lactate serum levels, a new biomarker of MS progression. Fluvoxamine 106-117 glial fibrillary acidic protein Rattus norvegicus 202-206 28410959-7 2017 In the clinic, PMC s of treated patients showed upregulation of mRNA expression and protein levels of BDNF, CREB and AKT after addition of fluvoxamine. Fluvoxamine 139-150 brain derived neurotrophic factor Homo sapiens 102-106 28410959-7 2017 In the clinic, PMC s of treated patients showed upregulation of mRNA expression and protein levels of BDNF, CREB and AKT after addition of fluvoxamine. Fluvoxamine 139-150 cAMP responsive element binding protein 1 Homo sapiens 108-112 28410959-7 2017 In the clinic, PMC s of treated patients showed upregulation of mRNA expression and protein levels of BDNF, CREB and AKT after addition of fluvoxamine. Fluvoxamine 139-150 AKT serine/threonine kinase 1 Homo sapiens 117-120 28243095-3 2017 The aim of this study is to investigate the effects of fluvoxamine on plasma interleukin-6 (IL-6) levels and on clinical improvement of the depressive state. Fluvoxamine 55-66 interleukin 6 Homo sapiens 77-90 28243095-3 2017 The aim of this study is to investigate the effects of fluvoxamine on plasma interleukin-6 (IL-6) levels and on clinical improvement of the depressive state. Fluvoxamine 55-66 interleukin 6 Homo sapiens 92-96 28243095-11 2017 CONCLUSION: Effect of fluvoxamine on IL-6 is partially associated with its clinical efficacy for MDD. Fluvoxamine 22-33 interleukin 6 Homo sapiens 37-41 27803785-3 2016 In this study, we aimed to evaluate the effect of fluvoxamine on the expression of some inflammatory genes like intercellular adhesion molecule (ICAM1), vascular cell adhesion molecule (VCAM1), cyclooxygenases2 (COX2), and inducible nitric oxide synthase (iNOS). Fluvoxamine 50-61 intercellular adhesion molecule 1 Rattus norvegicus 145-150 27987210-8 2017 Picrotoxin-induced anhedonia was ameliorated by fluvoxamine and S-(+)-fluoxetine, selective serotonin reuptake inhibitors with high affinity for the sigma1 receptor. Fluvoxamine 48-59 sigma non-opioid intracellular receptor 1 Mus musculus 149-164 27987210-9 2017 The effect of fluvoxamine was blocked by a 5-HT1A or a sigma1 receptor antagonist, and co-administration of the sigma1 receptor agonist (+)-SKF-10047 and the 5-HT1A receptor agonist osemozotan mimicked the effect of fluvoxamine. Fluvoxamine 14-25 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 43-49 27987210-9 2017 The effect of fluvoxamine was blocked by a 5-HT1A or a sigma1 receptor antagonist, and co-administration of the sigma1 receptor agonist (+)-SKF-10047 and the 5-HT1A receptor agonist osemozotan mimicked the effect of fluvoxamine. Fluvoxamine 14-25 sigma non-opioid intracellular receptor 1 Mus musculus 55-70 27402157-6 2017 Using this final PBPK model, it was predicted that weak CYP3A inhibitors (fluoxetine and fluvoxamine) are anticipated to have negligible DDI risk with palbociclib, whereas moderate CYP3A inhibitors (diltiazem and verapamil) may increase plasma palbociclib AUC by ~40%. Fluvoxamine 89-100 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-61 27056295-4 2017 In rats, pretreatment with either dehydroepiandrosterone or fluvoxamine, a high-affinity sigma1-receptor agonist, improved survival, renal function and structure, and the inflammatory response after sublethal renal ischemia-reperfusion injury. Fluvoxamine 60-71 sigma non-opioid intracellular receptor 1 Rattus norvegicus 89-104 27056295-5 2017 In human proximal tubular epithelial cells, stimulation by fluvoxamine or oxidative stress caused the sigma1-receptor to translocate from the endoplasmic reticulum to the cytosol and nucleus. Fluvoxamine 59-70 sigma non-opioid intracellular receptor 1 Rattus norvegicus 102-117 27056295-6 2017 Fluvoxamine stimulation in these cells also activated nitric oxide production that was blocked by sigma1-receptor knockdown or Akt inhibition. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 98-113 27056295-6 2017 Fluvoxamine stimulation in these cells also activated nitric oxide production that was blocked by sigma1-receptor knockdown or Akt inhibition. Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 127-130 27056295-7 2017 Similarly, in the postischemic rat kidney, sigma1-receptor activation by fluvoxamine triggered the Akt-nitric oxide synthase signaling pathway, resulting in time- and isoform-specific endothelial and neuronal nitric oxide synthase activation and nitric oxide production. Fluvoxamine 73-84 sigma non-opioid intracellular receptor 1 Rattus norvegicus 43-58 27056295-7 2017 Similarly, in the postischemic rat kidney, sigma1-receptor activation by fluvoxamine triggered the Akt-nitric oxide synthase signaling pathway, resulting in time- and isoform-specific endothelial and neuronal nitric oxide synthase activation and nitric oxide production. Fluvoxamine 73-84 AKT serine/threonine kinase 1 Rattus norvegicus 99-102 27056295-8 2017 Concurrently, intravital two-photon imaging revealed prompt peritubular vasodilation after fluvoxamine treatment, which was blocked by the sigma1-receptor antagonist or various nitric oxide synthase blockers. Fluvoxamine 91-102 sigma non-opioid intracellular receptor 1 Rattus norvegicus 139-154 27821711-6 2017 Using this information, a clinical study using the CYP1A2 inhibitor fluvoxamine was performed, resulting in an AUCi/AUC ratio of 1.60, confirming the role of CYP1A2 and indicating a balanced DDI risk profile. Fluvoxamine 68-79 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 51-57 27821711-6 2017 Using this information, a clinical study using the CYP1A2 inhibitor fluvoxamine was performed, resulting in an AUCi/AUC ratio of 1.60, confirming the role of CYP1A2 and indicating a balanced DDI risk profile. Fluvoxamine 68-79 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 158-164 28361099-9 2017 Further, using a pharmacogenomics genotyping panel, we discovered that the patient had the CYP2D6 nonfunctioning variant genotype *4/*4 that results in very low metabolic activity on a number of psychotropic drugs, including fluvoxamine which he was prescribed. Fluvoxamine 225-236 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 91-97 28315270-5 2017 Several clinical studies showed that sigma-1 receptor agonists such as fluvoxamine and ifenprodil could have beneficial effects in patients with neuropsychiatric disorders. Fluvoxamine 71-82 sigma non-opioid intracellular receptor 1 Rattus norvegicus 37-53 27569183-3 2017 In this study we looked at how Fluvoxamine treatment regulates depressive-like signs, motor impairments and the expression of IL-1beta, IL-6, TNF-alpha, TGF-beta and IL-10 cytokines in the striatum of a stressed Parkinsonian rat model. Fluvoxamine 31-42 interleukin 1 beta Rattus norvegicus 126-134 27569183-3 2017 In this study we looked at how Fluvoxamine treatment regulates depressive-like signs, motor impairments and the expression of IL-1beta, IL-6, TNF-alpha, TGF-beta and IL-10 cytokines in the striatum of a stressed Parkinsonian rat model. Fluvoxamine 31-42 interleukin 6 Rattus norvegicus 136-140 27569183-3 2017 In this study we looked at how Fluvoxamine treatment regulates depressive-like signs, motor impairments and the expression of IL-1beta, IL-6, TNF-alpha, TGF-beta and IL-10 cytokines in the striatum of a stressed Parkinsonian rat model. Fluvoxamine 31-42 tumor necrosis factor Rattus norvegicus 142-151 27569183-3 2017 In this study we looked at how Fluvoxamine treatment regulates depressive-like signs, motor impairments and the expression of IL-1beta, IL-6, TNF-alpha, TGF-beta and IL-10 cytokines in the striatum of a stressed Parkinsonian rat model. Fluvoxamine 31-42 transforming growth factor, beta 1 Rattus norvegicus 153-161 27569183-3 2017 In this study we looked at how Fluvoxamine treatment regulates depressive-like signs, motor impairments and the expression of IL-1beta, IL-6, TNF-alpha, TGF-beta and IL-10 cytokines in the striatum of a stressed Parkinsonian rat model. Fluvoxamine 31-42 interleukin 10 Rattus norvegicus 166-171 27569183-9 2017 Lipid peroxidation, mRNA levels of IL-1beta, IL-6 and TNF-alpha were down-regulated while IL-10 and TGF-beta levels were up-regulated in the lesioned striatum of Fluvoxamine treated rats. Fluvoxamine 162-173 interleukin 1 beta Rattus norvegicus 35-43 27569183-9 2017 Lipid peroxidation, mRNA levels of IL-1beta, IL-6 and TNF-alpha were down-regulated while IL-10 and TGF-beta levels were up-regulated in the lesioned striatum of Fluvoxamine treated rats. Fluvoxamine 162-173 interleukin 6 Rattus norvegicus 45-49 27569183-9 2017 Lipid peroxidation, mRNA levels of IL-1beta, IL-6 and TNF-alpha were down-regulated while IL-10 and TGF-beta levels were up-regulated in the lesioned striatum of Fluvoxamine treated rats. Fluvoxamine 162-173 tumor necrosis factor Rattus norvegicus 54-63 27569183-9 2017 Lipid peroxidation, mRNA levels of IL-1beta, IL-6 and TNF-alpha were down-regulated while IL-10 and TGF-beta levels were up-regulated in the lesioned striatum of Fluvoxamine treated rats. Fluvoxamine 162-173 interleukin 10 Rattus norvegicus 90-95 27569183-9 2017 Lipid peroxidation, mRNA levels of IL-1beta, IL-6 and TNF-alpha were down-regulated while IL-10 and TGF-beta levels were up-regulated in the lesioned striatum of Fluvoxamine treated rats. Fluvoxamine 162-173 transforming growth factor, beta 1 Rattus norvegicus 100-108 27803785-3 2016 In this study, we aimed to evaluate the effect of fluvoxamine on the expression of some inflammatory genes like intercellular adhesion molecule (ICAM1), vascular cell adhesion molecule (VCAM1), cyclooxygenases2 (COX2), and inducible nitric oxide synthase (iNOS). Fluvoxamine 50-61 vascular cell adhesion molecule 1 Rattus norvegicus 186-191 27803785-3 2016 In this study, we aimed to evaluate the effect of fluvoxamine on the expression of some inflammatory genes like intercellular adhesion molecule (ICAM1), vascular cell adhesion molecule (VCAM1), cyclooxygenases2 (COX2), and inducible nitric oxide synthase (iNOS). Fluvoxamine 50-61 nitric oxide synthase 2 Rattus norvegicus 223-254 27803785-3 2016 In this study, we aimed to evaluate the effect of fluvoxamine on the expression of some inflammatory genes like intercellular adhesion molecule (ICAM1), vascular cell adhesion molecule (VCAM1), cyclooxygenases2 (COX2), and inducible nitric oxide synthase (iNOS). Fluvoxamine 50-61 nitric oxide synthase 2 Rattus norvegicus 256-260 27803785-7 2016 RESULTS: The expression of ICAM1, VCAM1, COX2, and iNOS was significantly decreased by fluvoxamine in endothelial cells, macrophages, and in rat carrageenan-induced paw edema. Fluvoxamine 87-98 intercellular adhesion molecule 1 Rattus norvegicus 27-32 27803785-7 2016 RESULTS: The expression of ICAM1, VCAM1, COX2, and iNOS was significantly decreased by fluvoxamine in endothelial cells, macrophages, and in rat carrageenan-induced paw edema. Fluvoxamine 87-98 vascular cell adhesion molecule 1 Rattus norvegicus 34-39 27803785-7 2016 RESULTS: The expression of ICAM1, VCAM1, COX2, and iNOS was significantly decreased by fluvoxamine in endothelial cells, macrophages, and in rat carrageenan-induced paw edema. Fluvoxamine 87-98 nitric oxide synthase 2 Rattus norvegicus 51-55 26809458-3 2016 OBJECTIVES: We studied the dose-dependent efficacy of S1R agonist fluvoxamine (FLU) in the prevention of DM-induced depression and investigated the significance of the S1R-BDNF pathway. Fluvoxamine 66-77 sigma non-opioid intracellular receptor 1 Rattus norvegicus 54-57 27002781-1 2016 BACKGROUND: The aim of this study was to determine the impact of fluvoxamine, an inhibitor of Cytochrome P450 (CYP) 2C19 (CYP2C19), on the pharmacokinetics of escitalopram, a substrate of CYP2C19. Fluvoxamine 65-76 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 94-120 27002781-1 2016 BACKGROUND: The aim of this study was to determine the impact of fluvoxamine, an inhibitor of Cytochrome P450 (CYP) 2C19 (CYP2C19), on the pharmacokinetics of escitalopram, a substrate of CYP2C19. Fluvoxamine 65-76 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 122-129 27002781-1 2016 BACKGROUND: The aim of this study was to determine the impact of fluvoxamine, an inhibitor of Cytochrome P450 (CYP) 2C19 (CYP2C19), on the pharmacokinetics of escitalopram, a substrate of CYP2C19. Fluvoxamine 65-76 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 188-195 27339616-1 2016 RATIONALE: We previously reported that the fluvoxamine-induced increase in prefrontal dopamine levels is enhanced by adrenalectomy/castration (which results in circulating neurosteroid deficiency), via combined activation of serotonin1A (5-HT1A) and sigma1 receptors. Fluvoxamine 43-54 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 238-244 26809458-3 2016 OBJECTIVES: We studied the dose-dependent efficacy of S1R agonist fluvoxamine (FLU) in the prevention of DM-induced depression and investigated the significance of the S1R-BDNF pathway. Fluvoxamine 79-82 sigma non-opioid intracellular receptor 1 Rattus norvegicus 54-57 26809458-10 2016 S1R and BDNF protein levels were decreased in DM, while FLU induced SIR-BDNF production. Fluvoxamine 56-59 brain-derived neurotrophic factor Rattus norvegicus 72-76 26764533-4 2016 Antidepressants with distinct mechanisms of action, such as clomipramine, duloxetine and fluvoxamine, also increased BDNF mRNA expression in astrocytic and microglial cultures. Fluvoxamine 89-100 brain-derived neurotrophic factor Rattus norvegicus 117-121 26845564-0 2016 The ubiquitination of serotonin transporter in lymphoblasts derived from fluvoxamine-resistant depression patients. Fluvoxamine 73-84 solute carrier family 6 member 4 Homo sapiens 22-43 26845564-7 2016 In our study, we have used lymphoblasts derived from the peripheral blood lymphocytes to quantitatively examine SERT protein expression and ubiquitination in fluvoxamine-responsive and fluvoxamine-resistant MDD patients. Fluvoxamine 158-169 solute carrier family 6 member 4 Homo sapiens 112-116 26845564-8 2016 We found that the protein levels of SERT were higher in the fluvoxamine-resistant MDD patients. Fluvoxamine 60-71 solute carrier family 6 member 4 Homo sapiens 36-40 26845564-9 2016 Ubiquitinated protein levels of SERT were lower in the fluvoxamine-resistant MDD patients. Fluvoxamine 55-66 solute carrier family 6 member 4 Homo sapiens 32-36 26845564-11 2016 In sum, these findings suggest that the downregulation of the ubiquitination of SERT protein induces insufficient degradation of SERT by proteasome, which resulted in the upregulation of SERT protein in fluvoxamine-resistant MDD patients. Fluvoxamine 203-214 solute carrier family 6 member 4 Homo sapiens 80-84 26845564-11 2016 In sum, these findings suggest that the downregulation of the ubiquitination of SERT protein induces insufficient degradation of SERT by proteasome, which resulted in the upregulation of SERT protein in fluvoxamine-resistant MDD patients. Fluvoxamine 203-214 solute carrier family 6 member 4 Homo sapiens 129-133 26845564-11 2016 In sum, these findings suggest that the downregulation of the ubiquitination of SERT protein induces insufficient degradation of SERT by proteasome, which resulted in the upregulation of SERT protein in fluvoxamine-resistant MDD patients. Fluvoxamine 203-214 solute carrier family 6 member 4 Homo sapiens 129-133 26988603-6 2016 Fluvoxamine inhibited serum-induced ruffle formation, cell migration, and invasion of human GBM and glioma stem cells in vitro by suppressing both FAK and Akt/mammalian target of rapamycin signaling. Fluvoxamine 0-11 protein tyrosine kinase 2 Homo sapiens 147-150 26988603-6 2016 Fluvoxamine inhibited serum-induced ruffle formation, cell migration, and invasion of human GBM and glioma stem cells in vitro by suppressing both FAK and Akt/mammalian target of rapamycin signaling. Fluvoxamine 0-11 AKT serine/threonine kinase 1 Homo sapiens 155-158 26988603-6 2016 Fluvoxamine inhibited serum-induced ruffle formation, cell migration, and invasion of human GBM and glioma stem cells in vitro by suppressing both FAK and Akt/mammalian target of rapamycin signaling. Fluvoxamine 0-11 mechanistic target of rapamycin kinase Homo sapiens 159-188 26886336-1 2016 Fluvoxamine-perpetrated drug-drug interactions (DDIs) of victims metabolized by multiple cytochrome P450 isoforms (CYP1A2, CYP2C19, and CYP3A4) were simulated using 2 compartment-based tube modeling, assuming a multiple inhibition-constant (Ki) model, as well as a previously reported single Ki model. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 115-121 26886336-1 2016 Fluvoxamine-perpetrated drug-drug interactions (DDIs) of victims metabolized by multiple cytochrome P450 isoforms (CYP1A2, CYP2C19, and CYP3A4) were simulated using 2 compartment-based tube modeling, assuming a multiple inhibition-constant (Ki) model, as well as a previously reported single Ki model. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 123-130 26886336-1 2016 Fluvoxamine-perpetrated drug-drug interactions (DDIs) of victims metabolized by multiple cytochrome P450 isoforms (CYP1A2, CYP2C19, and CYP3A4) were simulated using 2 compartment-based tube modeling, assuming a multiple inhibition-constant (Ki) model, as well as a previously reported single Ki model. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 136-142 26626327-2 2016 Fluvoxamine is a potent CYP1A2 inhibitor and may increase the ratio of clozapine to its primary metabolite N-desmethylclozapine (NDMC). Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 24-30 26712326-4 2016 The results confirmed that the selective 5-HT1A antagonist WAY-100635 (0.05 mg/kg) partially blocked the fluvoxamine-quetiapine synergistic effect (maximum DA increase dropped from 325% to 214%). Fluvoxamine 105-116 5-hydroxytryptamine receptor 1A Homo sapiens 41-47 25316382-9 2015 Taken together, Sig-1R stimulation by SA4503 or fluvoxamine treatment increased hippocampal neurogenesis, which is closely associated with amelioration of depressive-like behaviors in CaMKIV null mice. Fluvoxamine 48-59 sigma non-opioid intracellular receptor 1 Mus musculus 16-22 30620511-5 2016 Flu-voxamine inhibits CYPlA2 and CYP2C9 activity, and paroxetine inhibits CYP2D6 activity. Fluvoxamine 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 30620511-7 2016 Fluvoxamine and paroxetine inhibit not only CYP but also P-glycoprotein. Fluvoxamine 0-11 peptidylprolyl isomerase G Homo sapiens 44-47 25316382-3 2015 Here, we demonstrate that chronic stimulation of Sig-1R by treatment with the agonist SA4503 or the SSRI fluvoxamine for 14 days improves depressive-like behaviors in CaMKIV null mice. Fluvoxamine 105-116 sigma non-opioid intracellular receptor 1 Mus musculus 49-55 25316382-9 2015 Taken together, Sig-1R stimulation by SA4503 or fluvoxamine treatment increased hippocampal neurogenesis, which is closely associated with amelioration of depressive-like behaviors in CaMKIV null mice. Fluvoxamine 48-59 calcium/calmodulin-dependent protein kinase IV Mus musculus 184-190 25316382-3 2015 Here, we demonstrate that chronic stimulation of Sig-1R by treatment with the agonist SA4503 or the SSRI fluvoxamine for 14 days improves depressive-like behaviors in CaMKIV null mice. Fluvoxamine 105-116 calcium/calmodulin-dependent protein kinase IV Mus musculus 167-173 26099559-1 2015 Co-administration of fluvoxamine (FLV) (perpetrator) and ramelteon (victim, high-clearance CYP1A2 substrate) reportedly showed a 130-fold increase in the area under blood-ramelteon-levels curve (AUCR), which is unpredictable by any method assuming the traditional well-stirred hepatic extraction (Eh ) model. Fluvoxamine 21-32 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 91-97 26415980-7 2015 Our results indicate that fluvoxamine suppresses beta-casein expression in MCF-12A cells via inhibition of STAT5 phosphorylation caused by induction of ER stress. Fluvoxamine 26-37 signal transducer and activator of transcription 5A Homo sapiens 107-112 26099559-1 2015 Co-administration of fluvoxamine (FLV) (perpetrator) and ramelteon (victim, high-clearance CYP1A2 substrate) reportedly showed a 130-fold increase in the area under blood-ramelteon-levels curve (AUCR), which is unpredictable by any method assuming the traditional well-stirred hepatic extraction (Eh ) model. Fluvoxamine 34-37 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 91-97 25719307-5 2015 Fluoxetine and norfluoxetine are also potent inhibitors of CYP2D6, and fluvoxamine is a potent inhibitor of both CYP1A2 and CYP2C19. Fluvoxamine 71-82 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 113-119 29124205-5 2015 We recently reported that fluvoxamine (Flv) alleviates ER stress via induction of sigma-1 receptor (Sig-1R). Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Homo sapiens 82-98 29124205-5 2015 We recently reported that fluvoxamine (Flv) alleviates ER stress via induction of sigma-1 receptor (Sig-1R). Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Homo sapiens 100-106 29124205-5 2015 We recently reported that fluvoxamine (Flv) alleviates ER stress via induction of sigma-1 receptor (Sig-1R). Fluvoxamine 39-42 sigma non-opioid intracellular receptor 1 Homo sapiens 82-98 29124205-5 2015 We recently reported that fluvoxamine (Flv) alleviates ER stress via induction of sigma-1 receptor (Sig-1R). Fluvoxamine 39-42 sigma non-opioid intracellular receptor 1 Homo sapiens 100-106 25719307-5 2015 Fluoxetine and norfluoxetine are also potent inhibitors of CYP2D6, and fluvoxamine is a potent inhibitor of both CYP1A2 and CYP2C19. Fluvoxamine 71-82 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 124-131 25590967-0 2015 Fluvoxamine alleviates seizure activity and downregulates hippocampal GAP-43 expression in pentylenetetrazole-kindled mice: role of 5-HT3 receptors. Fluvoxamine 0-11 growth associated protein 43 Mus musculus 70-76 25590967-10 2015 Repeated administration of fluvoxamine (20 mg/kg) in PTZ-kindled mice suppressed seizure scores, protected against hippocampal neuronal loss, and downregulated GAP-43 expression, without producing any signs of the 5-HT syndrome in healthy rats. Fluvoxamine 27-38 growth associated protein 43 Mus musculus 160-166 25756551-2 2015 Recent evidence from animal and clinical studies that adding fluvoxamine to antipsychotics alters the expression of transcripts encoding for the GABA-A receptor and BDNF led us to postulate that fluvoxamine augmentation may improve memory in schizophrenia. Fluvoxamine 61-72 brain derived neurotrophic factor Homo sapiens 165-169 25756551-2 2015 Recent evidence from animal and clinical studies that adding fluvoxamine to antipsychotics alters the expression of transcripts encoding for the GABA-A receptor and BDNF led us to postulate that fluvoxamine augmentation may improve memory in schizophrenia. Fluvoxamine 195-206 brain derived neurotrophic factor Homo sapiens 165-169 25689244-16 2015 CONCLUSION: Fluvoxamine attenuated the laboratory response to clopidogrel, possibly through inhibition of CYP2C19, whereas citalopram did not affect this response. Fluvoxamine 12-23 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 106-113 25159194-6 2015 Co-administration of pomalidomide with fluvoxamine (CYP1A2 inhibitor) in the presence of ketoconazole approximately doubled pomalidomide exposure. Fluvoxamine 39-50 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 52-58 25727963-0 2015 Role of the 5-HT1A autoreceptor in the enhancement of fluvoxamine-induced increases in prefrontal dopamine release by adrenalectomy/castration in mice. Fluvoxamine 54-65 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 12-18 25727963-1 2015 We have found that fluvoxamine-induced increases in prefrontal dopamine release are enhanced by adrenalectomy/castration and 5-HT1A receptors are involved in the enhancement. Fluvoxamine 19-30 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 125-131 24676049-5 2014 RESULTS: The activities of citrate synthase, malate dehydrogenase, and complexes I, II-III, and IV were decreased after prolonged administration of fluvoxamine in rats. Fluvoxamine 148-159 citrate synthase Rattus norvegicus 27-43 25704012-3 2015 Preclinical studies showed that some selective serotonin reuptake inhibitors (SSRIs; fluvoxamine, fluoxetine, excitalopram), donepezil, and ifenprodil act as sigma-1 receptor agonists. Fluvoxamine 85-96 sigma non-opioid intracellular receptor 1 Mus musculus 158-174 25704012-5 2015 A study using positron emission tomography have demonstrated that an oral administration of fluvoxamine or donepezil could bind to sigma-1 receptor in the healthy human brain, suggesting that sigma-1 receptor might be involved in the therapeutic mechanisms of these drugs. Fluvoxamine 92-103 sigma non-opioid intracellular receptor 1 Mus musculus 131-147 25704012-5 2015 A study using positron emission tomography have demonstrated that an oral administration of fluvoxamine or donepezil could bind to sigma-1 receptor in the healthy human brain, suggesting that sigma-1 receptor might be involved in the therapeutic mechanisms of these drugs. Fluvoxamine 92-103 sigma non-opioid intracellular receptor 1 Mus musculus 192-208 25704012-6 2015 Moreover, case reports suggest that sigma-1 receptor agonists, including fluvoxamine, and ifenprodil, may be effective in the treatment of cognitive impairment in schizophrenia, delirium in elderly people, and flashbacks in post-traumatic stress disorder. Fluvoxamine 73-84 sigma non-opioid intracellular receptor 1 Mus musculus 36-52 26514046-6 2015 Some antidepressants, such as paroxetine and fluvoxamine, are strong inhibitors of the CYP system. Fluvoxamine 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 24814141-0 2014 Promoter variation in the catechol-O-methyltransferase gene is associated with remission of symptoms during fluvoxamine treatment for major depression. Fluvoxamine 108-119 catechol-O-methyltransferase Homo sapiens 26-54 24814141-1 2014 We investigated the association between remission of depressive symptoms in fluvoxamine treatment and catechol-O-methyltransferase (COMT) gene. Fluvoxamine 76-87 catechol-O-methyltransferase Homo sapiens 102-130 24814141-1 2014 We investigated the association between remission of depressive symptoms in fluvoxamine treatment and catechol-O-methyltransferase (COMT) gene. Fluvoxamine 76-87 catechol-O-methyltransferase Homo sapiens 132-136 24846087-3 2014 Here we present the case of a 29-year-old female patient with disorganized schizophrenia who exhibited OCS due to fluvoxamine-induced elevation of CLZ serum levels via inhibition of CYP 1A2 und 2C19. Fluvoxamine 114-125 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 182-189 24709917-2 2014 Fluvoxamine, one of the currently known antidepressants, is a sigma-1 receptor agonist; its effectiveness in treating acute cerebral ischemia has not been reported. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 62-78 24709917-8 2014 Moreover, NE-100, a selective sigma-1 receptor antagonist, completely blocked the neuroprotective effect of fluvoxamine. Fluvoxamine 108-119 sigma non-opioid intracellular receptor 1 Rattus norvegicus 30-46 24709917-9 2014 The present findings suggest that the sigma-1 receptor agonist fluvoxamine reduces infarct volume and ameliorates neurological impairment even on postischemic treatment. Fluvoxamine 63-74 sigma non-opioid intracellular receptor 1 Rattus norvegicus 38-54 25089150-1 2014 BACKGROUND: We investigated the association between serum proBDNF, a precursor of brain-derived neurotrophic factor (BDNF), and response to fluvoxamine in patients with major depressive disorder (MDD) using the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR): physically healthy and free of current alcohol or drug abuse, comorbid anxiety, or personality disorders. Fluvoxamine 140-151 brain derived neurotrophic factor Homo sapiens 82-115 25089150-1 2014 BACKGROUND: We investigated the association between serum proBDNF, a precursor of brain-derived neurotrophic factor (BDNF), and response to fluvoxamine in patients with major depressive disorder (MDD) using the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR): physically healthy and free of current alcohol or drug abuse, comorbid anxiety, or personality disorders. Fluvoxamine 140-151 brain derived neurotrophic factor Homo sapiens 61-65 24793403-7 2014 Two notable drug-drug interactions are evident: asenapine (an inhibitor of CYP2D6) can increase plasma levels of paroxetine, and fluvoxamine (a CYP1A2 inhibitor) can increase plasma levels of asenapine. Fluvoxamine 129-140 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 144-150 24423185-8 2014 Estradiol"s or PPT"s inhibition of the fluvoxamine-induced slowing of 5-HT clearance mediated by ERalpha, was blocked after inhibition of either MAPK/ERK1/2 or PI3K/Akt signaling pathways. Fluvoxamine 39-50 tachykinin, precursor 1 Rattus norvegicus 15-18 24423185-8 2014 Estradiol"s or PPT"s inhibition of the fluvoxamine-induced slowing of 5-HT clearance mediated by ERalpha, was blocked after inhibition of either MAPK/ERK1/2 or PI3K/Akt signaling pathways. Fluvoxamine 39-50 estrogen receptor 1 Rattus norvegicus 97-104 24423185-8 2014 Estradiol"s or PPT"s inhibition of the fluvoxamine-induced slowing of 5-HT clearance mediated by ERalpha, was blocked after inhibition of either MAPK/ERK1/2 or PI3K/Akt signaling pathways. Fluvoxamine 39-50 mitogen activated protein kinase 3 Rattus norvegicus 145-149 24423185-8 2014 Estradiol"s or PPT"s inhibition of the fluvoxamine-induced slowing of 5-HT clearance mediated by ERalpha, was blocked after inhibition of either MAPK/ERK1/2 or PI3K/Akt signaling pathways. Fluvoxamine 39-50 mitogen activated protein kinase 3 Rattus norvegicus 150-156 25032855-0 2014 Fluvoxamine alleviates ER stress via induction of Sigma-1 receptor. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Mus musculus 50-66 25032855-3 2014 Fluvoxamine (Flv) is a selective serotonin reuptake inhibitor (SSRI) with a high affinity for Sig-1R. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Mus musculus 94-100 25032855-3 2014 Fluvoxamine (Flv) is a selective serotonin reuptake inhibitor (SSRI) with a high affinity for Sig-1R. Fluvoxamine 13-16 sigma non-opioid intracellular receptor 1 Mus musculus 94-100 24855828-5 2014 Both ticlopidine and fluvoxamine were competitive inhibitors of CYP2C19. Fluvoxamine 21-32 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 64-71 24421401-7 2014 Effects of the SSRI fluvoxamine and 5-HT(1A)R agonist 8-OH-DPAT were also potentiated in RGS6(+/-) mice. Fluvoxamine 20-31 regulator of G-protein signaling 6 Mus musculus 89-93 24582626-0 2014 Fluvoxamine moderates reduced voluntary activity following chronic dexamethasone infusion in mice via recovery of BDNF signal cascades. Fluvoxamine 0-11 brain derived neurotrophic factor Mus musculus 114-118 24582626-10 2014 However, marked expression of the XBP1 gene was observed in cDEX-mice treated with Flu compared with mice given saline and untreated cDEX-mice. Fluvoxamine 83-86 X-box binding protein 1 Mus musculus 34-38 24582626-11 2014 Expression of 5-HT2A and Sigma-1 receptors decreased after cDEX infusion compared with the saline group, and these decreases normalized to control levels upon Flu treatment. Fluvoxamine 159-162 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 14-20 24582626-12 2014 Our results indicate that the Flu moderates reductions in voluntary activity following chronic dexamethasone infusions in mice via recovery of BDNF signal cascades. Fluvoxamine 30-33 brain derived neurotrophic factor Mus musculus 143-147 24508523-5 2014 The order of potency for drugs at the sigma-1 receptor chaperone was as follows: fluvoxamine>sertraline>fluoxetine>escitalopram>citalopram>paroxetine>duoxetine. Fluvoxamine 81-92 sigma non-opioid intracellular receptor 1 Rattus norvegicus 38-54 24508523-7 2014 Furthermore, fluvoxamine, fluoxetine, escitalopram, and mirtazapine significantly potentiated NGF-induced neurite outgrowth in cell assays, and the effects of all these drugs, excluding mirtazapine, were antagonized by NE-100, a selective antagonist of the sigma-1 receptor chaperone. Fluvoxamine 13-24 sigma non-opioid intracellular receptor 1 Rattus norvegicus 257-273 24508523-10 2014 These findings suggest that activation at the sigma-1 receptor chaperone may be involved in the action of some SSRIs, such as fluvoxamine, fluoxetine and escitalopram. Fluvoxamine 126-137 sigma non-opioid intracellular receptor 1 Rattus norvegicus 46-62 24396420-8 2014 These results suggest that sigma1 receptors mediate fluvoxamine-elicited changes in the expression levels of mTOR, Camk2gamma and GSK-3beta in N2a cells, which indicates that sigma1 receptors are likely to be involved in the pharmacological effects of fluvoxamine. Fluvoxamine 52-63 mechanistic target of rapamycin kinase Mus musculus 109-113 24396420-8 2014 These results suggest that sigma1 receptors mediate fluvoxamine-elicited changes in the expression levels of mTOR, Camk2gamma and GSK-3beta in N2a cells, which indicates that sigma1 receptors are likely to be involved in the pharmacological effects of fluvoxamine. Fluvoxamine 52-63 glycogen synthase kinase 3 beta Mus musculus 130-139 24396420-0 2014 Effects of BD1047, a sigma1 receptor antagonist, on the expression of mTOR, Camk2gamma and GSK-3beta in fluvoxamine-treated N2a cells. Fluvoxamine 104-115 sigma non-opioid intracellular receptor 1 Mus musculus 21-36 24396420-0 2014 Effects of BD1047, a sigma1 receptor antagonist, on the expression of mTOR, Camk2gamma and GSK-3beta in fluvoxamine-treated N2a cells. Fluvoxamine 104-115 mechanistic target of rapamycin kinase Mus musculus 70-74 24396420-8 2014 These results suggest that sigma1 receptors mediate fluvoxamine-elicited changes in the expression levels of mTOR, Camk2gamma and GSK-3beta in N2a cells, which indicates that sigma1 receptors are likely to be involved in the pharmacological effects of fluvoxamine. Fluvoxamine 252-263 mechanistic target of rapamycin kinase Mus musculus 109-113 24396420-0 2014 Effects of BD1047, a sigma1 receptor antagonist, on the expression of mTOR, Camk2gamma and GSK-3beta in fluvoxamine-treated N2a cells. Fluvoxamine 104-115 glycogen synthase kinase 3 beta Mus musculus 91-100 24373833-9 2014 These mechanisms likely underlie in part the anti-hypertrophic and cardioprotective action of the sigma1R agonists including fluvoxamine. Fluvoxamine 125-136 sigma non-opioid intracellular receptor 1 Rattus norvegicus 98-105 24396420-3 2014 The present study aimed to observe the effects of fluvoxamine on the expression levels of mammalian target of rapamycin (mTOR), Ca2+/calmodulin-dependent protein kinase 2gamma (Camk2gamma) and glycogen synthase kinase-3beta (GSK-3beta) in fluvoxamine-treated N2a cells and attempted to elucidate whether sigma1 receptors mediate the pharmacological effects of fluvoxamine. Fluvoxamine 50-61 mechanistic target of rapamycin kinase Homo sapiens 90-119 24396420-3 2014 The present study aimed to observe the effects of fluvoxamine on the expression levels of mammalian target of rapamycin (mTOR), Ca2+/calmodulin-dependent protein kinase 2gamma (Camk2gamma) and glycogen synthase kinase-3beta (GSK-3beta) in fluvoxamine-treated N2a cells and attempted to elucidate whether sigma1 receptors mediate the pharmacological effects of fluvoxamine. Fluvoxamine 50-61 mechanistic target of rapamycin kinase Homo sapiens 121-125 24396420-3 2014 The present study aimed to observe the effects of fluvoxamine on the expression levels of mammalian target of rapamycin (mTOR), Ca2+/calmodulin-dependent protein kinase 2gamma (Camk2gamma) and glycogen synthase kinase-3beta (GSK-3beta) in fluvoxamine-treated N2a cells and attempted to elucidate whether sigma1 receptors mediate the pharmacological effects of fluvoxamine. Fluvoxamine 50-61 glycogen synthase kinase 3 beta Homo sapiens 193-223 24396420-3 2014 The present study aimed to observe the effects of fluvoxamine on the expression levels of mammalian target of rapamycin (mTOR), Ca2+/calmodulin-dependent protein kinase 2gamma (Camk2gamma) and glycogen synthase kinase-3beta (GSK-3beta) in fluvoxamine-treated N2a cells and attempted to elucidate whether sigma1 receptors mediate the pharmacological effects of fluvoxamine. Fluvoxamine 50-61 glycogen synthase kinase 3 beta Homo sapiens 225-234 24373833-0 2014 Fluvoxamine rescues mitochondrial Ca2+ transport and ATP production through sigma(1)-receptor in hypertrophic cardiomyocytes. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 76-93 24373833-1 2014 AIMS: We previously reported that fluvoxamine, a selective serotonin reuptake inhibitor with high affinity for the sigma1-receptor (sigma1R), ameliorates cardiac hypertrophy and dysfunction via sigma1R stimulation. Fluvoxamine 34-45 sigma non-opioid intracellular receptor 1 Rattus norvegicus 115-130 24373833-1 2014 AIMS: We previously reported that fluvoxamine, a selective serotonin reuptake inhibitor with high affinity for the sigma1-receptor (sigma1R), ameliorates cardiac hypertrophy and dysfunction via sigma1R stimulation. Fluvoxamine 34-45 sigma non-opioid intracellular receptor 1 Rattus norvegicus 132-139 24373833-1 2014 AIMS: We previously reported that fluvoxamine, a selective serotonin reuptake inhibitor with high affinity for the sigma1-receptor (sigma1R), ameliorates cardiac hypertrophy and dysfunction via sigma1R stimulation. Fluvoxamine 34-45 sigma non-opioid intracellular receptor 1 Rattus norvegicus 194-201 24373833-5 2014 Interestingly, sigma1R stimulation with fluvoxamine rescued impaired mitochondrial Ca(2+) mobilization and ATP production, an effect abolished by treatment of cells with the sigma1R antagonist, NE-100. Fluvoxamine 40-51 sigma non-opioid intracellular receptor 1 Rattus norvegicus 15-22 24373833-5 2014 Interestingly, sigma1R stimulation with fluvoxamine rescued impaired mitochondrial Ca(2+) mobilization and ATP production, an effect abolished by treatment of cells with the sigma1R antagonist, NE-100. Fluvoxamine 40-51 sigma non-opioid intracellular receptor 1 Rattus norvegicus 174-181 24373833-8 2014 SIGNIFICANCE: These results suggest that sigma1R stimulation with fluvoxamine promotes SR-mitochondrial Ca(2+) transport and mitochondrial ATP production, whereas sigma1R stimulation suppresses intracellular Ca(2+) overload through IP3Rs and RyRs. Fluvoxamine 66-77 sigma non-opioid intracellular receptor 1 Rattus norvegicus 41-48 23716885-7 2013 CONCLUSIONS: There would be an accumulation of aripiprazole when coadministered with fluvoxamine, a known inhibitor of CYP3A4, leading to hepatic damage and reduction in aripiprazole when administered along with carbamazepine. Fluvoxamine 85-96 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-125 25252797-4 2014 Fluvoxamine (10 - 100 muM) concentration-dependently suppressed both monosynaptic A-fiber- and C-fiber-mediated EPSCs, which were attenuated by the selective 5-HT1A receptor antagonist WAY100635. Fluvoxamine 0-11 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 158-164 25252797-8 2014 These results suggest that fluvoxamine reduces excitatory synaptic transmission from primary afferent fibers via presynaptic mechanisms involving 5-HT1A and/or 5-HT3 receptors, which may contribute to its analgesic effects. Fluvoxamine 27-38 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 146-152 23896940-4 2013 Here, the effects of diclofenac, different antidepressants (sertraline, venlafaxine, and fluvoxamine), and vitamin B6 (pyridoxine) on IL-10 and tumor necrosis factor-alpha (TNF-alpha) change with and without immune challenges with lipopolysaccharide (LPS) were investigated in in vitro culture of astrocytes from 2-day-old Swiss-Albino mice. Fluvoxamine 89-100 interleukin 10 Mus musculus 134-139 23896940-4 2013 Here, the effects of diclofenac, different antidepressants (sertraline, venlafaxine, and fluvoxamine), and vitamin B6 (pyridoxine) on IL-10 and tumor necrosis factor-alpha (TNF-alpha) change with and without immune challenges with lipopolysaccharide (LPS) were investigated in in vitro culture of astrocytes from 2-day-old Swiss-Albino mice. Fluvoxamine 89-100 tumor necrosis factor Mus musculus 144-171 23851260-4 2013 Systemic administration of fluvoxamine, a selective 5-HT reuptake inhibitor with agonistic activity towards the sigma1 receptor, increased prefrontal dopamine (DA) levels, and adrenalectomy/castration potentiated this fluvoxamine-induced increase in DA. Fluvoxamine 27-38 sigma non-opioid intracellular receptor 1 Mus musculus 112-127 23851260-4 2013 Systemic administration of fluvoxamine, a selective 5-HT reuptake inhibitor with agonistic activity towards the sigma1 receptor, increased prefrontal dopamine (DA) levels, and adrenalectomy/castration potentiated this fluvoxamine-induced increase in DA. Fluvoxamine 218-229 sigma non-opioid intracellular receptor 1 Mus musculus 112-127 23728612-0 2013 Histamine H1 receptor occupancy by the new-generation antidepressants fluvoxamine and mirtazapine: a positron emission tomography study in healthy volunteers. Fluvoxamine 70-81 histamine receptor H1 Homo sapiens 0-21 25358426-0 2014 Bcl1 polymorphism of the glucocorticoid receptor gene and treatment response to milnacipran and fluvoxamine in Japanese patients with depression. Fluvoxamine 96-107 cyclin D1 Homo sapiens 0-4 25358426-0 2014 Bcl1 polymorphism of the glucocorticoid receptor gene and treatment response to milnacipran and fluvoxamine in Japanese patients with depression. Fluvoxamine 96-107 nuclear receptor subfamily 3 group C member 1 Homo sapiens 25-48 25358426-2 2014 The purpose of this study was to investigate whether Bcl1 polymorphisms of GRs are associated with the antidepressant effect of milnacipran, a serotonin noradrenaline reuptake inhibitor (SNRI), and fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), in Japanese patients with depression. Fluvoxamine 198-209 cyclin D1 Homo sapiens 53-57 25358426-7 2014 However, when milnacipran- and fluvoxamine-treated subjects were analyzed independently, patients with G allele in Bcl1 polymorphism had a significantly better response to fluvoxamine than those with C/C genotype. Fluvoxamine 31-42 cyclin D1 Homo sapiens 115-119 25358426-7 2014 However, when milnacipran- and fluvoxamine-treated subjects were analyzed independently, patients with G allele in Bcl1 polymorphism had a significantly better response to fluvoxamine than those with C/C genotype. Fluvoxamine 172-183 cyclin D1 Homo sapiens 115-119 25069239-7 2013 Some SSRIs (fluoxetine and fluvoxamine), which have sigma-1 receptor agonistic activity, inhibited the development of methamphetamine-induced behavioral sensitization. Fluvoxamine 27-38 sigma non-opioid intracellular receptor 1 Mus musculus 52-68 25069239-10 2013 These results suggest that sigma-1 receptor agonistic activity might be involved in the attenuating effects of fluoxetine and fluvoxamine on methamphetamine-induced behavioral sensitization and rewarding effects. Fluvoxamine 126-137 sigma non-opioid intracellular receptor 1 Mus musculus 27-43 23044468-8 2013 Moreover, sigma-1 receptor agonists, including the endogenous neurosteroid dehydroepiandosterone (DHEA) and the selective serotonin reuptake inhibitor (SSRI) fluvoxamine, show potent cardioprotective and antidepressive effects in rodents, via sigma-1 receptor stimulation. Fluvoxamine 158-169 sigma non-opioid intracellular receptor 1 Mus musculus 10-26 23288236-0 2013 5-HTTLPR rs25531A > G differentially influence paroxetine and fluvoxamine antidepressant efficacy: a randomized, controlled trial. Fluvoxamine 65-76 solute carrier family 6 member 4 Homo sapiens 0-8 23001793-5 2013 Ketoconazole (inhibitor of CYP3A4/5) inhibited metabolism of (-)-THP or (+)-THP at same degree, whereas the inhibition of fluvoxamine (inhibitor of CYP1A2) on metabolism of (+)-THP was greater than that of (-)-THP; moreover, the metabolic rate of (+)-THP was 5.3-fold of (-)-THP in recombinant human CYP1A2. Fluvoxamine 122-133 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 148-154 23001793-5 2013 Ketoconazole (inhibitor of CYP3A4/5) inhibited metabolism of (-)-THP or (+)-THP at same degree, whereas the inhibition of fluvoxamine (inhibitor of CYP1A2) on metabolism of (+)-THP was greater than that of (-)-THP; moreover, the metabolic rate of (+)-THP was 5.3-fold of (-)-THP in recombinant human CYP1A2. Fluvoxamine 122-133 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 300-306 23428811-6 2013 Our findings suggest that SSRIs such as fluvoxamine protect against PO- and TAC-induced cardiac dysfunction by upregulating sigma-1 receptor expression and stimulating sigma-1 receptor-mediated Akt-eNOS signaling. Fluvoxamine 40-51 sigma non-opioid intracellular receptor 1 Rattus norvegicus 124-140 23428811-6 2013 Our findings suggest that SSRIs such as fluvoxamine protect against PO- and TAC-induced cardiac dysfunction by upregulating sigma-1 receptor expression and stimulating sigma-1 receptor-mediated Akt-eNOS signaling. Fluvoxamine 40-51 sigma non-opioid intracellular receptor 1 Rattus norvegicus 168-184 23044468-8 2013 Moreover, sigma-1 receptor agonists, including the endogenous neurosteroid dehydroepiandosterone (DHEA) and the selective serotonin reuptake inhibitor (SSRI) fluvoxamine, show potent cardioprotective and antidepressive effects in rodents, via sigma-1 receptor stimulation. Fluvoxamine 158-169 sigma non-opioid intracellular receptor 1 Mus musculus 243-259 23408363-5 2013 RESULTS: Addition of 25 or 50 mg fluvoxamine induces a mean rise of plasma concentrations of clozapine with a factor 2-3, probably even higher with the addition of 100 mg. Fluvoxamine 33-44 transcription termination factor 2 Homo sapiens 110-120 21989809-3 2012 Previously, we have reported that acute haloperidol (HALO)-fluvoxamine (FLU) in vivo and in vitro treatment regulated GABA-Abeta2/3 receptor subunits, and protein kinase C (PKC) and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) signaling pathways. Fluvoxamine 72-75 protein kinase C, gamma Rattus norvegicus 173-176 23041149-0 2012 Involvement of the sigma1 receptor in the antidepressant-like effects of fluvoxamine in the forced swimming test in comparison with the effects elicited by paroxetine. Fluvoxamine 73-84 sigma non-opioid intracellular receptor 1 Mus musculus 19-34 23041149-1 2012 We studied the involvement of the sigma(1) receptor in the antidepressant-like effects of the selective serotonin reuptake inhibitor (SSRI) fluvoxamine in DBA/2 mice using the forced swimming test. Fluvoxamine 140-151 sigma non-opioid intracellular receptor 1 Mus musculus 34-51 23041149-6 2012 These results suggested that the sigma(1) receptor was associated with fluvoxamine-induced antidepressant-like effects but not with paroxetine-induced antidepressant-like effects. Fluvoxamine 71-82 sigma non-opioid intracellular receptor 1 Mus musculus 33-50 22859721-0 2012 In silico and intuitive predictions of CYP46A1 inhibition by marketed drugs with subsequent enzyme crystallization in complex with fluvoxamine. Fluvoxamine 131-142 cytochrome P450 family 46 subfamily A member 1 Homo sapiens 39-46 22859721-5 2012 One of the identified tight binders, the widely used antidepressant fluvoxamine, was cocrystallized with CYP46A1. Fluvoxamine 68-79 cytochrome P450 family 46 subfamily A member 1 Homo sapiens 105-112 22859721-7 2012 The NH(2)-containing arm of the Y-shaped fluvoxamine coordinates the CYP46A1 heme iron, whereas the methoxy-containing arm points away from the heme group and has multiple hydrophobic interactions with aliphatic amino acid residues. Fluvoxamine 41-52 cytochrome P450 family 46 subfamily A member 1 Homo sapiens 69-76 22225849-2 2012 Previously, we reported that acute systemic treatment with estradiol or progesterone blocked the ability of the selective serotonin reuptake inhibitor, fluvoxamine, to inhibit serotonin transporter function in ovariectomized rats. Fluvoxamine 152-163 solute carrier family 6 member 4 Rattus norvegicus 176-197 22225849-9 2012 The AD-like effect of estradiol involved ER beta and G-protein coupled receptor 30, whereas its blockade of fluvoxamine"s effects was ER alpha-mediated. Fluvoxamine 108-119 estrogen receptor 1 Rattus norvegicus 134-142 22297159-2 2012 In particular, some selective serotonin (5-HT) reuptake inhibitors, such as citalopram and fluvoxamine, seem to exert part of their antidepressant effects through oxytocin (OT) release. Fluvoxamine 91-102 oxytocin/neurophysin I prepropeptide Homo sapiens 163-171 22297159-2 2012 In particular, some selective serotonin (5-HT) reuptake inhibitors, such as citalopram and fluvoxamine, seem to exert part of their antidepressant effects through oxytocin (OT) release. Fluvoxamine 91-102 oxytocin/neurophysin I prepropeptide Homo sapiens 173-175 21989809-3 2012 Previously, we have reported that acute haloperidol (HALO)-fluvoxamine (FLU) in vivo and in vitro treatment regulated GABA-Abeta2/3 receptor subunits, and protein kinase C (PKC) and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) signaling pathways. Fluvoxamine 72-75 mitogen activated protein kinase 1 Rattus norvegicus 254-257 21989809-6 2012 HALO and FLU each given alone increased PKC phosphorylation levels (29 +- 15% and 40 +- 11.8%, vs. control, respectively) and did not affect ERK2 phosphorylation, while HALO-FLU combined treatment did not alter PKC phosphorylation levels and significantly decreased ERK2 phosphorylation levels (58 +- 4.4% vs. control). Fluvoxamine 9-12 protein kinase C, gamma Rattus norvegicus 40-43 22649406-0 2012 Fluvoxamine attenuated endoplasmic reticulum stress-induced leptin resistance. Fluvoxamine 0-11 leptin Homo sapiens 60-66 24900029-11 2012 CONCLUSION: Anesthesia, body temperature, and fluvoxamine affect the result of [(18)F]FP-CIT PET in mice by altering striatal and bone uptakes. Fluvoxamine 46-57 citron Mus musculus 89-92 22649406-3 2012 In the present study, we found that fluvoxamine, a selective serotonin reuptake inhibitor used for depression, can attenuate ER stress-induced "leptin resistance," i.e., insensitivity to the anti-obesity hormone leptin. Fluvoxamine 36-47 leptin Homo sapiens 144-150 22649406-3 2012 In the present study, we found that fluvoxamine, a selective serotonin reuptake inhibitor used for depression, can attenuate ER stress-induced "leptin resistance," i.e., insensitivity to the anti-obesity hormone leptin. Fluvoxamine 36-47 leptin Homo sapiens 212-218 22649406-6 2012 ER stress caused an impairment of leptin-induced STAT3 phosphorylation which was reversed by fluvoxamine. Fluvoxamine 93-104 leptin Homo sapiens 34-40 22649406-6 2012 ER stress caused an impairment of leptin-induced STAT3 phosphorylation which was reversed by fluvoxamine. Fluvoxamine 93-104 signal transducer and activator of transcription 3 Homo sapiens 49-54 22649406-7 2012 Fluvoxamine would be a novel leptin-sensitizing drug, which targets ER stress. Fluvoxamine 0-11 leptin Homo sapiens 29-35 22288409-5 2012 Among SSRIs, fluvoxamine, a potent sigma-1 receptor agonist, has the highest affinity at sigma-1 receptors. Fluvoxamine 13-24 sigma non-opioid intracellular receptor 1 Rattus norvegicus 35-51 22288409-7 2012 Furthermore, phencyclidine (PCP)-induced cognitive impairment, associated with animal models of schizophrenia is significantly improved by sub-chronic administration of sigma-1 receptor agonists such as fluvoxamine, SA4503 (cutamesine) and donepezil. Fluvoxamine 203-214 sigma non-opioid intracellular receptor 1 Rattus norvegicus 169-185 22288409-9 2012 A positron emission tomography (PET) study using the specific sigma-1 receptor ligand [11C]SA4503 demonstrates that fluvoxamine and donepezil bind to sigma-1 receptors in the healthy human brain. Fluvoxamine 116-127 sigma non-opioid intracellular receptor 1 Rattus norvegicus 62-78 22288409-10 2012 In clinical studies, some sigma-1 receptor agonists, including fluvoxamine, donepezil and neurosteroids, improve cognitive impairment and clinical symptoms in neuropsychiatric diseases. Fluvoxamine 63-74 sigma non-opioid intracellular receptor 1 Rattus norvegicus 26-42 22467103-3 2012 Fluvoxamine (50 mg/kg/day) alone significantly elevated the serum level of adiponectin, but no significant difference was found between other drug-treated groups and the control group. Fluvoxamine 0-11 adiponectin, C1Q and collagen domain containing Rattus norvegicus 75-86 21739267-5 2012 Coadministration of fluvoxamine, a less potent CYP3A4 inhibitor, decreased the CL/F of aripiprazole by 39% in CYP2D6 EMs and 40% in IMs, indicating the same inhibitory effect on CYP enzymes, regardless of the CYP2D6 genotype. Fluvoxamine 20-31 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-53 21739267-5 2012 Coadministration of fluvoxamine, a less potent CYP3A4 inhibitor, decreased the CL/F of aripiprazole by 39% in CYP2D6 EMs and 40% in IMs, indicating the same inhibitory effect on CYP enzymes, regardless of the CYP2D6 genotype. Fluvoxamine 20-31 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 110-116 21739267-5 2012 Coadministration of fluvoxamine, a less potent CYP3A4 inhibitor, decreased the CL/F of aripiprazole by 39% in CYP2D6 EMs and 40% in IMs, indicating the same inhibitory effect on CYP enzymes, regardless of the CYP2D6 genotype. Fluvoxamine 20-31 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 209-215 21739267-6 2012 Percent contribution of CYP2D6 to total CL/F (CYP2D6 plus CYP3A4) of aripiprazole estimated as a reduced percentage of CL/F by CYP enzyme inhibition was 62% for CYP2D6 EMs and 24% for IMs in paroxetine coadministration, and 40% for CYP2D6 EMs and 18% for IMs in fluvoxamine coadministration. Fluvoxamine 262-273 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 24-30 21739267-6 2012 Percent contribution of CYP2D6 to total CL/F (CYP2D6 plus CYP3A4) of aripiprazole estimated as a reduced percentage of CL/F by CYP enzyme inhibition was 62% for CYP2D6 EMs and 24% for IMs in paroxetine coadministration, and 40% for CYP2D6 EMs and 18% for IMs in fluvoxamine coadministration. Fluvoxamine 262-273 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 21739267-6 2012 Percent contribution of CYP2D6 to total CL/F (CYP2D6 plus CYP3A4) of aripiprazole estimated as a reduced percentage of CL/F by CYP enzyme inhibition was 62% for CYP2D6 EMs and 24% for IMs in paroxetine coadministration, and 40% for CYP2D6 EMs and 18% for IMs in fluvoxamine coadministration. Fluvoxamine 262-273 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 21739267-6 2012 Percent contribution of CYP2D6 to total CL/F (CYP2D6 plus CYP3A4) of aripiprazole estimated as a reduced percentage of CL/F by CYP enzyme inhibition was 62% for CYP2D6 EMs and 24% for IMs in paroxetine coadministration, and 40% for CYP2D6 EMs and 18% for IMs in fluvoxamine coadministration. Fluvoxamine 262-273 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 21979923-2 2011 Enzymes involved in duloxetine metabolism are cytochrome P450 isoenzymes (CYP) CYP1A2 and to a lesser extent CYP2D6 whereas the selective serotonin reuptake inhibitor Fluvoxamine is known to be a potent inhibitor of CYP1A2. Fluvoxamine 167-178 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 216-222 22293694-7 2012 By contrast, fluvoxamine administration significantly attenuated TAC-induced myocardial hypertrophy concomitant with recovery of Sig-1R expression in LV. Fluvoxamine 13-24 sigma non-opioid intracellular receptor 1 Mus musculus 129-135 22293694-9 2012 The fluvoxamine cardioprotective effect was nullified by treatment with a Sig-1R antagonist, NE-100 (1 mg/kg). Fluvoxamine 4-15 sigma non-opioid intracellular receptor 1 Mus musculus 74-80 22293694-11 2012 Fluvoxamine treatment significantly restored TAC-induced impaired Akt and eNOS phosphorylation in LV. Fluvoxamine 0-11 thymoma viral proto-oncogene 1 Mus musculus 66-69 22293694-12 2012 Our findings suggest that fluvoxamine protects heart against TAC-induced cardiac dysfunction via upregulation of Sig-1R and stimulation of Sig-1R-mediated Akt-eNOS signaling in mice. Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Mus musculus 113-119 22293694-12 2012 Our findings suggest that fluvoxamine protects heart against TAC-induced cardiac dysfunction via upregulation of Sig-1R and stimulation of Sig-1R-mediated Akt-eNOS signaling in mice. Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Mus musculus 139-145 22293694-12 2012 Our findings suggest that fluvoxamine protects heart against TAC-induced cardiac dysfunction via upregulation of Sig-1R and stimulation of Sig-1R-mediated Akt-eNOS signaling in mice. Fluvoxamine 26-37 thymoma viral proto-oncogene 1 Mus musculus 155-158 22293694-13 2012 This is the first report of a potential role of Sig-1R stimulation by fluvoxamine in preventing cardiac hypertrophy and myocardial injury in TAC mice. Fluvoxamine 70-81 sigma non-opioid intracellular receptor 1 Mus musculus 48-54 21979923-7 2011 Our findings indicate that duloxetine plasma levels can be enhanced by a potent CYP1A2 inhibition by FLX and that DLX, even in higher plasma levels, seems to be well tolerated. Fluvoxamine 101-104 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 80-86 21487652-3 2011 RESULTS: Fluvoxamine-induced increases in the extracellular levels of dopamine (DA), but not of 5-HT and noradrenaline, were significantly higher in adrenalectomized/castrated than in sham-operated mice, and this effect was blocked by BD1047, a selective sigma(1) receptor antagonist. Fluvoxamine 9-20 sigma non-opioid intracellular receptor 1 Mus musculus 255-272 21487652-5 2011 Moreover, fluvoxamine increased c-Fos expression, a marker of neuronal activity, in the prefrontal cortex of adrenalectomized/castrated mice, and this effect was blocked by BD1047. Fluvoxamine 10-21 FBJ osteosarcoma oncogene Mus musculus 32-37 21487652-0 2011 Fluvoxamine enhances prefrontal dopaminergic neurotransmission in adrenalectomized/castrated mice via both 5-HT reuptake inhibition and sigma(1) receptor activation. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Mus musculus 136-153 21487652-7 2011 CONCLUSIONS: These findings suggest that fluvoxamine enhances prefrontal dopaminergic neurotransmission via both 5-HT reuptake inhibition and sigma(1) receptor activation under the circulating neuroactive steroid-deficient conditions. Fluvoxamine 41-52 sigma non-opioid intracellular receptor 1 Mus musculus 142-159 21487652-1 2011 RATIONALE: Fluvoxamine, a selective serotonin (5-HT) reuptake inhibitor (SSRI) and an agonist for the sigma(1) receptors, increases extracellular monoamines in the prefrontal cortex, but it is not known whether the sigma(1) receptor is involved in the neurochemical effect of fluvoxamine. Fluvoxamine 11-22 sigma non-opioid intracellular receptor 1 Mus musculus 102-119 20547595-0 2011 CYP2D6 genotype and smoking influence fluvoxamine steady-state concentration in Japanese psychiatric patients: lessons for genotype-phenotype association study design in translational pharmacogenetics. Fluvoxamine 38-49 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 24250395-10 2011 Our results suggest that IP administration of fluvoxamine produces a noticeable anti-inflammatory effect in the carrageenan-induced paw edema in rats and at least, a part of this effect is mediated through glucocorticoid receptor. Fluvoxamine 46-57 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 206-229 20547595-6 2011 We investigated the joint effect of smoking (an inducer of CYP1A2) and CYP2D6 genotype on interindividual variability in fluvoxamine steady-state concentration. Fluvoxamine 121-132 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 71-77 20547595-8 2011 While CYP2D6 genotype significantly influenced fluvoxamine concentration in all four dose groups (p < 0.05), the percentage variance explained (R2) by CYP2D6 decreased as the dose of fluvoxamine increased. Fluvoxamine 47-58 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 6-12 20547595-8 2011 While CYP2D6 genotype significantly influenced fluvoxamine concentration in all four dose groups (p < 0.05), the percentage variance explained (R2) by CYP2D6 decreased as the dose of fluvoxamine increased. Fluvoxamine 186-197 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 154-160 20547595-10 2011 Together, CYP2D6 genotype and smoking status explained 23% of the variance in fluvoxamine concentration but only at the low 50 mg/d dose group. Fluvoxamine 78-89 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 10-16 20547595-11 2011 These findings contribute to evidence-based and personalized choice of fluvoxamine dose using smoking status and CYP2D6 genetic variation. Fluvoxamine 71-82 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 113-119 21326811-0 2011 Neonatal exposure to fluoxetine and fluvoxamine alteres spine density in mouse hippocampal CA1 pyramidal neurons. Fluvoxamine 36-47 carbonic anhydrase 1 Mus musculus 91-94 21371073-2 2011 The serotonin transporter (SERT) handles serotonin (5-hydroxytryptamine (5-HT)) and is blocked by the antidepressant SERT inhibitors fluoxetine and fluvoxamine. Fluvoxamine 148-159 solute carrier family 6 member 4 Rattus norvegicus 4-25 21371073-2 2011 The serotonin transporter (SERT) handles serotonin (5-hydroxytryptamine (5-HT)) and is blocked by the antidepressant SERT inhibitors fluoxetine and fluvoxamine. Fluvoxamine 148-159 solute carrier family 6 member 4 Rattus norvegicus 27-31 21371073-2 2011 The serotonin transporter (SERT) handles serotonin (5-hydroxytryptamine (5-HT)) and is blocked by the antidepressant SERT inhibitors fluoxetine and fluvoxamine. Fluvoxamine 148-159 solute carrier family 6 member 4 Rattus norvegicus 117-121 21371073-10 2011 In in vitro experiments, SERT affected the contractility of aortas from female rats, as evidenced by an eightfold increase in potency of 5-HT in fluvoxamine (1 mumol/L)-incubated WT aortas compared with control. Fluvoxamine 145-156 solute carrier family 6 member 4 Rattus norvegicus 25-29 21044620-0 2011 Sigma-1 receptor stimulation with fluvoxamine activates Akt-eNOS signaling in the thoracic aorta of ovariectomized rats with abdominal aortic banding. Fluvoxamine 34-45 sigma non-opioid intracellular receptor 1 Rattus norvegicus 0-16 21044620-0 2011 Sigma-1 receptor stimulation with fluvoxamine activates Akt-eNOS signaling in the thoracic aorta of ovariectomized rats with abdominal aortic banding. Fluvoxamine 34-45 AKT serine/threonine kinase 1 Rattus norvegicus 56-59 21044620-0 2011 Sigma-1 receptor stimulation with fluvoxamine activates Akt-eNOS signaling in the thoracic aorta of ovariectomized rats with abdominal aortic banding. Fluvoxamine 34-45 nitric oxide synthase 3 Rattus norvegicus 60-64 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 176-182 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 184-190 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 200-207 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 211-217 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 221-227 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 232-238 21044620-5 2011 Fluvoxamine administration significantly attenuated pressure overload-induced vascular injury with concomitant increase in receptor expression and subsequent decrease in IP3 receptor expression. Fluvoxamine 0-11 inositol 1,4,5-trisphosphate receptor, type 3 Rattus norvegicus 170-182 21044620-6 2011 Fluvoxamine treatment also significantly restored pressure overload-induced impaired Akt phosphorylation and stimulated eNOS protein expression as well as Akt-mediated eNOS phosphorylation (Ser1177). Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 85-88 21044620-6 2011 Fluvoxamine treatment also significantly restored pressure overload-induced impaired Akt phosphorylation and stimulated eNOS protein expression as well as Akt-mediated eNOS phosphorylation (Ser1177). Fluvoxamine 0-11 nitric oxide synthase 3 Rattus norvegicus 120-124 21044620-6 2011 Fluvoxamine treatment also significantly restored pressure overload-induced impaired Akt phosphorylation and stimulated eNOS protein expression as well as Akt-mediated eNOS phosphorylation (Ser1177). Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 155-158 21044620-7 2011 Fluvoxamine"s vasculoprotective effect was nullified by co-administration of a sigma-1 receptor antagonist. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 79-95 22506441-3 2011 We report a case of gynecomastia and galactorrhea in an adolescent male while on a combination of risperidone and fluvoxamine, although the serum prolactin was within normal range. Fluvoxamine 114-125 prolactin Homo sapiens 146-155 21427517-10 2011 The behavioral effects induced by mixed sigma(1)-receptor/SSRI antidepressants, like fluvoxamine or sertraline, may therefore involve a non-selective action at both targets. Fluvoxamine 85-96 sigma non-opioid intracellular receptor 1 Mus musculus 40-57 20619611-0 2010 Effect of catechol-O-methyltransferase Val(108/158)Met polymorphism on antidepressant efficacy of fluvoxamine. Fluvoxamine 98-109 catechol-O-methyltransferase Homo sapiens 10-38 20802134-0 2010 Sigma1-receptor stimulation with fluvoxamine ameliorates transverse aortic constriction-induced myocardial hypertrophy and dysfunction in mice. Fluvoxamine 33-44 sigma non-opioid intracellular receptor 1 Mus musculus 0-15 20802134-3 2010 Here, we investigated the role of sigma(1)-receptor (sigma(1)R) stimulation with fluvoxamine on myocardial hypertrophy and cardiac functional recovery. Fluvoxamine 81-92 sigma non-opioid intracellular receptor 1 Mus musculus 53-62 20802134-7 2010 In contrast, fluvoxamine administration significantly attenuated TAC-induced myocardial hypertrophy concomitant with recovery of sigma(1)R expression in the LV. Fluvoxamine 13-24 sigma non-opioid intracellular receptor 1 Mus musculus 129-138 20802134-9 2010 The fluvoxamine cardioprotective effect was nullified by treatment with a sigma(1)R antagonist [NE-100 (1 mg/kg)]. Fluvoxamine 4-15 sigma non-opioid intracellular receptor 1 Mus musculus 74-83 20802134-11 2010 Fluvoxamine treatment significantly restored TAC-induced impaired Akt and endothelial nitric oxide synthase (eNOS) phosphorylation in the LV. Fluvoxamine 0-11 thymoma viral proto-oncogene 1 Mus musculus 66-69 20802134-11 2010 Fluvoxamine treatment significantly restored TAC-induced impaired Akt and endothelial nitric oxide synthase (eNOS) phosphorylation in the LV. Fluvoxamine 0-11 nitric oxide synthase 3, endothelial cell Mus musculus 74-107 20802134-12 2010 Our findings suggest that fluvoxamine protects against TAC-induced cardiac dysfunction via upregulated sigma(1)R expression and stimulation of sigma(1)R-mediated Akt-eNOS signaling in mice. Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Mus musculus 103-112 20802134-12 2010 Our findings suggest that fluvoxamine protects against TAC-induced cardiac dysfunction via upregulated sigma(1)R expression and stimulation of sigma(1)R-mediated Akt-eNOS signaling in mice. Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Mus musculus 143-152 20802134-12 2010 Our findings suggest that fluvoxamine protects against TAC-induced cardiac dysfunction via upregulated sigma(1)R expression and stimulation of sigma(1)R-mediated Akt-eNOS signaling in mice. Fluvoxamine 26-37 thymoma viral proto-oncogene 1 Mus musculus 162-165 20802134-13 2010 This is the first report of a potential role for sigma(1)R stimulation by fluvoxamine in attenuating cardiac hypertrophy and restoring contractility in TAC mice. Fluvoxamine 74-85 sigma non-opioid intracellular receptor 1 Mus musculus 49-58 21273663-3 2010 The kinetic analysis of chlorpromazine metabolism carried out in the absence or presence of antidepressants showed that fluvoxamine potently inhibited chlorpromazine 5-sulfoxidation (K(i) = 2.8 muM), mono-N-demethylation (K(i) = 1.4 muM) and di-N-demethylation (K(i) = 1.1 muM) via a competitive mechanism at therapeutic antidepressant concentrations. Fluvoxamine 120-131 latexin Homo sapiens 194-197 21273663-3 2010 The kinetic analysis of chlorpromazine metabolism carried out in the absence or presence of antidepressants showed that fluvoxamine potently inhibited chlorpromazine 5-sulfoxidation (K(i) = 2.8 muM), mono-N-demethylation (K(i) = 1.4 muM) and di-N-demethylation (K(i) = 1.1 muM) via a competitive mechanism at therapeutic antidepressant concentrations. Fluvoxamine 120-131 latexin Homo sapiens 233-236 21273663-3 2010 The kinetic analysis of chlorpromazine metabolism carried out in the absence or presence of antidepressants showed that fluvoxamine potently inhibited chlorpromazine 5-sulfoxidation (K(i) = 2.8 muM), mono-N-demethylation (K(i) = 1.4 muM) and di-N-demethylation (K(i) = 1.1 muM) via a competitive mechanism at therapeutic antidepressant concentrations. Fluvoxamine 120-131 latexin Homo sapiens 233-236 20722474-0 2010 Targeting sigma-1 receptor with fluvoxamine ameliorates pressure-overload-induced hypertrophy and dysfunctions. Fluvoxamine 32-43 sigma non-opioid intracellular receptor 1 Rattus norvegicus 10-26 20722474-5 2010 The fluvoxamine administration significantly attenuated PO-induced myocardial hypertrophy with concomitant increase in the expression of Sig-1R in LV. Fluvoxamine 4-15 sigma non-opioid intracellular receptor 1 Rattus norvegicus 137-143 20722474-8 2010 Fluvoxamine treatment significantly restored PO-induced impaired eNOS and Akt activity in the LV. Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 74-77 20722474-1 2010 OBJECTIVE: We here investigated the effect of sigma-1 receptor (Sig-1R) stimulation with fluvoxamine on myocardial hypertrophy, cardiac functional recovery and defined mechanisms underlying its cardioprotective action. Fluvoxamine 89-100 sigma non-opioid intracellular receptor 1 Rattus norvegicus 46-62 20722474-10 2010 Fluvoxamine treatment protects PO-induced cardiac injury via upregulation of Sig-1R and stimulation of Sig-1R-mediated Akt-eNOS signaling in ovariectomized rats. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 77-83 20722474-10 2010 Fluvoxamine treatment protects PO-induced cardiac injury via upregulation of Sig-1R and stimulation of Sig-1R-mediated Akt-eNOS signaling in ovariectomized rats. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 103-109 20722474-1 2010 OBJECTIVE: We here investigated the effect of sigma-1 receptor (Sig-1R) stimulation with fluvoxamine on myocardial hypertrophy, cardiac functional recovery and defined mechanisms underlying its cardioprotective action. Fluvoxamine 89-100 sigma non-opioid intracellular receptor 1 Rattus norvegicus 64-70 20722474-10 2010 Fluvoxamine treatment protects PO-induced cardiac injury via upregulation of Sig-1R and stimulation of Sig-1R-mediated Akt-eNOS signaling in ovariectomized rats. Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 119-122 20435083-3 2010 All three activities were substantially inhibited (>or=75%) by the CYP1 inhibitor alpha-naphthoflavone, whereas only MROD and CECOD were substantially inhibited by the CYP1A2-preferential inhibitor fluvoxamine. Fluvoxamine 201-212 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 70-74 20435083-3 2010 All three activities were substantially inhibited (>or=75%) by the CYP1 inhibitor alpha-naphthoflavone, whereas only MROD and CECOD were substantially inhibited by the CYP1A2-preferential inhibitor fluvoxamine. Fluvoxamine 201-212 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 171-177 19004511-3 2010 METHODS: We assessed the effect of SSRIs, (citalopram, fluvoxamine and fluoxetine), on TNF-alpha-induced expression of VCAM-1 and ICAM-1 in human aorta endothelial cells and adhesiveness to U937 monocytes. Fluvoxamine 55-66 tumor necrosis factor Homo sapiens 87-96 20846463-6 2010 One micromolar of paroxetine, fluvoxamine or sertraline increased cPLA2a expression during chronic treatment; citalopram had a similar effect at 0.1-0.5 muM; these are therapeutically relevant concentrations. Fluvoxamine 30-41 phospholipase A2, group V Mus musculus 66-71 20491280-9 2010 The authors investigated whether 5-HTTLPR was associated with the antidepressant response to fluvoxamine, a selective serotonin reuptake inhibitor. Fluvoxamine 93-104 solute carrier family 6 member 4 Homo sapiens 33-41 19958758-0 2010 Involvement of the 5-HT(1A) receptor in the anti-immobility effects of fluvoxamine in the forced swimming test and mouse strain differences in 5-HT(1A) receptor binding. Fluvoxamine 71-82 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 19-36 19958758-4 2010 The anti-immobility effects of fluvoxamine in DBA/2 mice were inhibited by the 5-HT(1A) receptor antagonist N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-(2-pyridinyl)cyclohexanecarboxamide (WAY 100635). Fluvoxamine 31-42 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 79-96 19958758-6 2010 These results suggest that fluvoxamine-induced antidepressant-like effects in DBA/2 mice are mediated by the 5-HT(1A) receptor. Fluvoxamine 27-38 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 109-126 19958758-10 2010 These results suggest that, although the anti-immobility effects of fluvoxamine in DBA/2 mice are mediated by the 5-HT(1A) receptor, strain differences in 5-HT(1A) receptor binding are not related to variation in immobility time and responses to fluvoxamine. Fluvoxamine 68-79 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 114-131 19004511-3 2010 METHODS: We assessed the effect of SSRIs, (citalopram, fluvoxamine and fluoxetine), on TNF-alpha-induced expression of VCAM-1 and ICAM-1 in human aorta endothelial cells and adhesiveness to U937 monocytes. Fluvoxamine 55-66 vascular cell adhesion molecule 1 Homo sapiens 119-125 19004511-3 2010 METHODS: We assessed the effect of SSRIs, (citalopram, fluvoxamine and fluoxetine), on TNF-alpha-induced expression of VCAM-1 and ICAM-1 in human aorta endothelial cells and adhesiveness to U937 monocytes. Fluvoxamine 55-66 intercellular adhesion molecule 1 Homo sapiens 130-136 19004511-10 2010 ICAM-1 expression was decreased in the presence of fluvoxamine and fluoxetine at concentrations from 10(-7) M to 10(-4) M (p<0.05) and in the presence of citalopram at concentrations from 10(-7) M to 10(-5) M (p<0.05). Fluvoxamine 51-62 intercellular adhesion molecule 1 Homo sapiens 0-6 20007294-7 2010 The minor caffeine-fluvoxamine interaction (1.78-fold) was slightly higher than predicted values based on determination of a moderate f(m) value for CYP1A1, although CYP1A2 may also be involved in caffeine metabolism. Fluvoxamine 19-30 Cytochrome P450 1A1 Canis lupus familiaris 149-155 20007294-7 2010 The minor caffeine-fluvoxamine interaction (1.78-fold) was slightly higher than predicted values based on determination of a moderate f(m) value for CYP1A1, although CYP1A2 may also be involved in caffeine metabolism. Fluvoxamine 19-30 cytochrome P450 family 1 subfamily A member 2 Canis lupus familiaris 166-172 19760522-6 2010 In addition, we performed an association analysis between TSNAX and the efficacy of fluvoxamine treatment in 120 Japanese patients with MDD. Fluvoxamine 84-95 translin associated factor X Homo sapiens 58-63 20128812-6 2010 Further, the selective 5-HT reuptake inhibitor fluvoxamine increased WAY 100635 induced head twitches in SERT +/+ and +/- mice. Fluvoxamine 47-58 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 105-109 19995565-13 2010 SIGNIFICANCE: This study demonstrates that fluvoxamine treatment rapidly increased phosphorylation of Akt-1. Fluvoxamine 43-54 AKT serine/threonine kinase 1 Rattus norvegicus 102-107 19995565-0 2010 Fluvoxamine and sigma-1 receptor agonists dehydroepiandrosterone (DHEA)-sulfate induces the Ser473-phosphorylation of Akt-1 in PC12 cells. Fluvoxamine 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 118-123 19995565-7 2010 Here, we examined whether fluvoxamine (FLV) activated Akt-1 and increased phosphorylation of Akt-1 via sigma-1 receptor in PC12 cells. Fluvoxamine 26-37 AKT serine/threonine kinase 1 Rattus norvegicus 54-59 19995565-7 2010 Here, we examined whether fluvoxamine (FLV) activated Akt-1 and increased phosphorylation of Akt-1 via sigma-1 receptor in PC12 cells. Fluvoxamine 26-37 AKT serine/threonine kinase 1 Rattus norvegicus 93-98 19995565-7 2010 Here, we examined whether fluvoxamine (FLV) activated Akt-1 and increased phosphorylation of Akt-1 via sigma-1 receptor in PC12 cells. Fluvoxamine 26-37 sigma non-opioid intracellular receptor 1 Rattus norvegicus 103-119 19995565-7 2010 Here, we examined whether fluvoxamine (FLV) activated Akt-1 and increased phosphorylation of Akt-1 via sigma-1 receptor in PC12 cells. Fluvoxamine 39-42 AKT serine/threonine kinase 1 Rattus norvegicus 54-59 19995565-7 2010 Here, we examined whether fluvoxamine (FLV) activated Akt-1 and increased phosphorylation of Akt-1 via sigma-1 receptor in PC12 cells. Fluvoxamine 39-42 AKT serine/threonine kinase 1 Rattus norvegicus 93-98 19995565-7 2010 Here, we examined whether fluvoxamine (FLV) activated Akt-1 and increased phosphorylation of Akt-1 via sigma-1 receptor in PC12 cells. Fluvoxamine 39-42 sigma non-opioid intracellular receptor 1 Rattus norvegicus 103-119 19843060-5 2009 RESULTS: (i) The CYP3A4 and CYP1A2 inhibitors ketoconazole and fluvoxamine inhibited CLZ oxidation to varying extents in individual microsomal fractions. Fluvoxamine 63-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 17-23 19875396-1 2010 A patient with an SCN5A p.W822X nonsense mutation, localized in the transmembrane region DII-S4 of the Na(v)1.5 sodium channel and leading to a non-expression of the mutant allele, was prescribed the selective serotonin reuptake inhibitor (SSRI) fluvoxamine (Floxyfral), 100 mg per day. Fluvoxamine 246-257 sodium voltage-gated channel alpha subunit 5 Homo sapiens 18-23 19875396-1 2010 A patient with an SCN5A p.W822X nonsense mutation, localized in the transmembrane region DII-S4 of the Na(v)1.5 sodium channel and leading to a non-expression of the mutant allele, was prescribed the selective serotonin reuptake inhibitor (SSRI) fluvoxamine (Floxyfral), 100 mg per day. Fluvoxamine 246-257 immunoglobulin lambda variable 2-18 Homo sapiens 103-111 19875396-1 2010 A patient with an SCN5A p.W822X nonsense mutation, localized in the transmembrane region DII-S4 of the Na(v)1.5 sodium channel and leading to a non-expression of the mutant allele, was prescribed the selective serotonin reuptake inhibitor (SSRI) fluvoxamine (Floxyfral), 100 mg per day. Fluvoxamine 259-268 sodium voltage-gated channel alpha subunit 5 Homo sapiens 18-23 19875396-1 2010 A patient with an SCN5A p.W822X nonsense mutation, localized in the transmembrane region DII-S4 of the Na(v)1.5 sodium channel and leading to a non-expression of the mutant allele, was prescribed the selective serotonin reuptake inhibitor (SSRI) fluvoxamine (Floxyfral), 100 mg per day. Fluvoxamine 259-268 immunoglobulin lambda variable 2-18 Homo sapiens 103-111 20118554-0 2010 Effects of cigarette smoking and cytochrome P450 2D6 genotype on fluvoxamine concentration in plasma of Japanese patients. Fluvoxamine 65-76 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 33-52 20118554-7 2010 Among non-smoker patients, the C/D ratios of fluvoxamine in those with one and two mutated alleles of CYP2D6 were 1.6- and 1.4-fold higher, respectively, than those with no mutated alleles, though the differences among those three genotype groups were not significant. Fluvoxamine 45-56 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 102-108 20016223-0 2010 Genetic association analysis of functional polymorphisms in neuronal nitric oxide synthase 1 gene (NOS1) and mood disorders and fluvoxamine response in major depressive disorder in the Japanese population. Fluvoxamine 128-139 nitric oxide synthase 1 Homo sapiens 69-92 20016223-0 2010 Genetic association analysis of functional polymorphisms in neuronal nitric oxide synthase 1 gene (NOS1) and mood disorders and fluvoxamine response in major depressive disorder in the Japanese population. Fluvoxamine 128-139 nitric oxide synthase 1 Homo sapiens 99-103 20016223-6 2010 In addition, we performed an association analysis between NOS1 and the efficacy of fluvoxamine treatment in 117 MDD patients. Fluvoxamine 83-94 nitric oxide synthase 1 Homo sapiens 58-62 21179660-7 2010 In addition, responders to fluvoxamine, paroxetine, milnacipran, and sertraline all increased serum BDNF levels. Fluvoxamine 27-38 brain derived neurotrophic factor Homo sapiens 100-104 19679463-3 2009 Anti-cancer effects of a selective sigma-1 agonist, 4-(N-benzylpiperidin-4-yl)-4-iodobenzamide (4-IBP), in glioblastoma were shown previously, leading to the present work where the effects on glioblastoma cells of 17 agonists or antagonists of sigma-1 receptors were studied, including currently marketed drugs fluvoxamine, dextromethorphan, donepezil, memantine and haloperidol. Fluvoxamine 311-322 translocator protein Homo sapiens 98-101 19928541-5 2009 GAF scales for depressive symptoms including the "waiting-for-instruction" behavior improved in 7 of these 9 cases with fluvoxamine. Fluvoxamine 120-131 fibroblast growth factor 9 Homo sapiens 0-3 19843060-5 2009 RESULTS: (i) The CYP3A4 and CYP1A2 inhibitors ketoconazole and fluvoxamine inhibited CLZ oxidation to varying extents in individual microsomal fractions. Fluvoxamine 63-74 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 28-34 20021354-7 2009 Moreover, a recent study using the specific sigma-1 receptor ligand [(11)C] SA4503 and positron emission tomography (PET) have demonstrated that an oral administration of fluvoxamine, but not paroxetine, could bind to sigma-1 receptors in the healthy human brain, in a dose-dependent manner. Fluvoxamine 171-182 sigma non-opioid intracellular receptor 1 Rattus norvegicus 44-60 19386277-8 2009 In addition, we performed an association analysis of GRM2 and GRM3 and the efficacy of fluvoxamine treatment in 117 Japanese patients with MDD. Fluvoxamine 87-98 glutamate metabotropic receptor 2 Homo sapiens 53-57 19386277-0 2009 Association analysis of group II metabotropic glutamate receptor genes (GRM2 and GRM3) with mood disorders and fluvoxamine response in a Japanese population. Fluvoxamine 111-122 glutamate metabotropic receptor 2 Homo sapiens 72-76 20021354-5 2009 In a cell culture system, we demonstrated that fluvoxamine, but not sertraline or paroxetine, significantly potentiated nerve-growth factor (NGF)-induced neurite outgrowth in PC12 cells, and that the effect of fluvoxamine on NGF-induced neurite outgrowth was significantly antagonized by treatment with the selective sigma-1 receptor antagonist NE-100. Fluvoxamine 47-58 nerve growth factor Rattus norvegicus 120-139 20021354-5 2009 In a cell culture system, we demonstrated that fluvoxamine, but not sertraline or paroxetine, significantly potentiated nerve-growth factor (NGF)-induced neurite outgrowth in PC12 cells, and that the effect of fluvoxamine on NGF-induced neurite outgrowth was significantly antagonized by treatment with the selective sigma-1 receptor antagonist NE-100. Fluvoxamine 47-58 nerve growth factor Rattus norvegicus 141-144 20021354-5 2009 In a cell culture system, we demonstrated that fluvoxamine, but not sertraline or paroxetine, significantly potentiated nerve-growth factor (NGF)-induced neurite outgrowth in PC12 cells, and that the effect of fluvoxamine on NGF-induced neurite outgrowth was significantly antagonized by treatment with the selective sigma-1 receptor antagonist NE-100. Fluvoxamine 47-58 nerve growth factor Rattus norvegicus 225-228 20021354-5 2009 In a cell culture system, we demonstrated that fluvoxamine, but not sertraline or paroxetine, significantly potentiated nerve-growth factor (NGF)-induced neurite outgrowth in PC12 cells, and that the effect of fluvoxamine on NGF-induced neurite outgrowth was significantly antagonized by treatment with the selective sigma-1 receptor antagonist NE-100. Fluvoxamine 47-58 sigma non-opioid intracellular receptor 1 Rattus norvegicus 317-333 19593168-4 2009 In patients comedicated with fluvoxamine, a strong CYP1A2 inhibitor, clozapine and norclozapine concentrations correlate with CYP3A activity (r = 0.44, P = 0.075; r = 0.63, P = 0.007, respectively). Fluvoxamine 29-40 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 51-57 19593168-4 2009 In patients comedicated with fluvoxamine, a strong CYP1A2 inhibitor, clozapine and norclozapine concentrations correlate with CYP3A activity (r = 0.44, P = 0.075; r = 0.63, P = 0.007, respectively). Fluvoxamine 29-40 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-131 19225771-0 2009 Effect of smoking and CYP2D6 polymorphisms on the extent of fluvoxamine-alprazolam interaction in patients with psychosomatic disease. Fluvoxamine 60-71 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 22-28 19386277-0 2009 Association analysis of group II metabotropic glutamate receptor genes (GRM2 and GRM3) with mood disorders and fluvoxamine response in a Japanese population. Fluvoxamine 111-122 glutamate metabotropic receptor 3 Homo sapiens 81-85 19386277-6 2009 Therefore, we studied the association between Group II mGluR genes (GRM2 and GRM3) and mood disorders and the efficacy of fluvoxamine treatment in Japanese MDD patients. Fluvoxamine 122-133 glutamate metabotropic receptor 2 Homo sapiens 68-72 19386277-6 2009 Therefore, we studied the association between Group II mGluR genes (GRM2 and GRM3) and mood disorders and the efficacy of fluvoxamine treatment in Japanese MDD patients. Fluvoxamine 122-133 glutamate metabotropic receptor 3 Homo sapiens 77-81 19225771-1 2009 PURPOSE: Fluvoxamine (FVX) is metabolized by cytochrome P450 (CYP) 2D6 and CYP1A2 and inhibits CYP3A4. Fluvoxamine 9-20 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 45-70 19225771-1 2009 PURPOSE: Fluvoxamine (FVX) is metabolized by cytochrome P450 (CYP) 2D6 and CYP1A2 and inhibits CYP3A4. Fluvoxamine 9-20 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 75-81 19225771-1 2009 PURPOSE: Fluvoxamine (FVX) is metabolized by cytochrome P450 (CYP) 2D6 and CYP1A2 and inhibits CYP3A4. Fluvoxamine 9-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-101 19225771-1 2009 PURPOSE: Fluvoxamine (FVX) is metabolized by cytochrome P450 (CYP) 2D6 and CYP1A2 and inhibits CYP3A4. Fluvoxamine 22-25 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 45-70 19225771-1 2009 PURPOSE: Fluvoxamine (FVX) is metabolized by cytochrome P450 (CYP) 2D6 and CYP1A2 and inhibits CYP3A4. Fluvoxamine 22-25 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 75-81 19225771-1 2009 PURPOSE: Fluvoxamine (FVX) is metabolized by cytochrome P450 (CYP) 2D6 and CYP1A2 and inhibits CYP3A4. Fluvoxamine 22-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-101 18976545-0 2009 Fluvoxamine exerts anorexic effect in 5-HT2C receptor mutant mice with heterozygous mutation of beta-endorphin gene. Fluvoxamine 0-11 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 38-53 19457229-16 2009 In addition, fluvoxamine and ranitidine decreased the levels of the oxidant parameters, myeloperoxidase and malondialdeyhyde, in the stomach tissues of the rats when compared to indomethacin group. Fluvoxamine 13-24 myeloperoxidase Rattus norvegicus 88-103 18976545-0 2009 Fluvoxamine exerts anorexic effect in 5-HT2C receptor mutant mice with heterozygous mutation of beta-endorphin gene. Fluvoxamine 0-11 pro-opiomelanocortin-alpha Mus musculus 96-110 18976545-2 2009 We previously reported that fluvoxamine, a selective serotonin reuptake inhibitor, together with pharmacological inactivation of 5-HT2C receptors exert feeding suppression through activation of 5-HT1B receptors in mice. Fluvoxamine 28-39 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 194-200 18976545-3 2009 Here, we report that fluvoxamine exerted anorexic effects in 5-HT2C receptor mutant mice with heterozygous mutation of beta-endorphin gene (2CREnd mice), whereas fluvoxamine had no effect on food intake in age-matched wild-type mice and 5-HT2C receptor mutant mice, which are associated with decreases in hypothalamic proopiomelanocortin (POMC) expression. Fluvoxamine 21-32 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 61-76 18976545-3 2009 Here, we report that fluvoxamine exerted anorexic effects in 5-HT2C receptor mutant mice with heterozygous mutation of beta-endorphin gene (2CREnd mice), whereas fluvoxamine had no effect on food intake in age-matched wild-type mice and 5-HT2C receptor mutant mice, which are associated with decreases in hypothalamic proopiomelanocortin (POMC) expression. Fluvoxamine 21-32 pro-opiomelanocortin-alpha Mus musculus 119-133 18976545-5 2009 These results suggest that fluvoxamine-induced feeding suppression requires a perturbation of 5-HT2C receptor and beta-endorphin signalling plus functional hypothalamic POMC activity, whereas mCPP-induced feeding suppression does not always require functional 5-HT2C receptor, beta-endorphin, and POMC activity in mice. Fluvoxamine 27-38 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 94-109 18976545-5 2009 These results suggest that fluvoxamine-induced feeding suppression requires a perturbation of 5-HT2C receptor and beta-endorphin signalling plus functional hypothalamic POMC activity, whereas mCPP-induced feeding suppression does not always require functional 5-HT2C receptor, beta-endorphin, and POMC activity in mice. Fluvoxamine 27-38 pro-opiomelanocortin-alpha Mus musculus 114-128 18976545-5 2009 These results suggest that fluvoxamine-induced feeding suppression requires a perturbation of 5-HT2C receptor and beta-endorphin signalling plus functional hypothalamic POMC activity, whereas mCPP-induced feeding suppression does not always require functional 5-HT2C receptor, beta-endorphin, and POMC activity in mice. Fluvoxamine 27-38 pro-opiomelanocortin-alpha Mus musculus 169-173 18976545-5 2009 These results suggest that fluvoxamine-induced feeding suppression requires a perturbation of 5-HT2C receptor and beta-endorphin signalling plus functional hypothalamic POMC activity, whereas mCPP-induced feeding suppression does not always require functional 5-HT2C receptor, beta-endorphin, and POMC activity in mice. Fluvoxamine 27-38 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 260-275 18976545-5 2009 These results suggest that fluvoxamine-induced feeding suppression requires a perturbation of 5-HT2C receptor and beta-endorphin signalling plus functional hypothalamic POMC activity, whereas mCPP-induced feeding suppression does not always require functional 5-HT2C receptor, beta-endorphin, and POMC activity in mice. Fluvoxamine 27-38 pro-opiomelanocortin-alpha Mus musculus 277-291 18976545-5 2009 These results suggest that fluvoxamine-induced feeding suppression requires a perturbation of 5-HT2C receptor and beta-endorphin signalling plus functional hypothalamic POMC activity, whereas mCPP-induced feeding suppression does not always require functional 5-HT2C receptor, beta-endorphin, and POMC activity in mice. Fluvoxamine 27-38 pro-opiomelanocortin-alpha Mus musculus 297-301 18949539-0 2009 Spectrofluorimetric determination of fluvoxamine in dosage forms and plasma via derivatization with 4-chloro-7-nitrobenzo-2-oxa-1,3-diazole. Fluvoxamine 37-48 OXA1L mitochondrial inner membrane protein Homo sapiens 124-129 19571598-4 2009 Therefore, we examined the association between the orphan nuclear receptor Rev-erbalpha gene (NR1D1) and the efficacy of fluvoxamine treatment in 118 Japanese patients with major depressive disorder. Fluvoxamine 121-132 nuclear receptor subfamily 1 group D member 1 Homo sapiens 94-99 18322468-0 2009 Impact of ovarian hormones on the modulation of the serotonin transporter by fluvoxamine. Fluvoxamine 77-88 solute carrier family 6 member 4 Rattus norvegicus 52-73 18322468-4 2009 Local application of the SSRI, fluvoxamine, directly into the CA3 area of hippocampus increased significantly 5-HT clearance time parameters in male rats and female rats in estrus or diestrus, but not in proestrus. Fluvoxamine 31-42 carbonic anhydrase 3 Rattus norvegicus 62-65 19347611-0 2009 CLOCK may predict the response to fluvoxamine treatment in Japanese major depressive disorder patients. Fluvoxamine 34-45 clock circadian regulator Homo sapiens 0-5 19347611-3 2009 Therefore, we examined the association between CLOCK and the efficacy of fluvoxamine treatment in 121 patients with Japanese major depressive disorder (MDD). Fluvoxamine 73-84 clock circadian regulator Homo sapiens 47-52 19347611-11 2009 Our results indicate that CLOCK genotype may be a predictor of fluvoxamine treatment response in Japanese MDD. Fluvoxamine 63-74 clock circadian regulator Homo sapiens 26-31 18493251-1 2008 BACKGROUND AND PURPOSE: The pharmacokinetic-pharmacodynamic (PK-PD) correlation of fluvoxamine 5-HT transporter (SERT) occupancy was determined in rat frontal cortex ex vivo. Fluvoxamine 83-94 solute carrier family 6 member 4 Rattus norvegicus 113-117 18817806-0 2009 Intracerebroventricular fluvoxamine administration inhibited pain behavior but increased Fos expression in affective pain pathways. Fluvoxamine 24-35 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 89-92 18817806-3 2009 A 5-HT1 receptor antagonist (WAY-100635) and a 5-HT2 receptor antagonist (ketanserin), injected i.c.v., induced hyperalgesia and inhibited fluvoxamine"s anti-nociceptive effects. Fluvoxamine 139-150 5-hydroxytryptamine receptor 2A Homo sapiens 47-61 18817806-7 2009 Moreover, fluvoxamine alone increased Fos in the BL and PFC. Fluvoxamine 10-21 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 38-41 18817806-9 2009 The results indicated that 5-HT2 receptor activities participate minimally in Fos induction by fluvoxamine in the PFC and BL. Fluvoxamine 95-106 5-hydroxytryptamine receptor 2A Homo sapiens 27-41 18817806-9 2009 The results indicated that 5-HT2 receptor activities participate minimally in Fos induction by fluvoxamine in the PFC and BL. Fluvoxamine 95-106 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 78-81 18817806-10 2009 In contrast, WAY-100635 affected the Fos expression produced by fluvoxamine. Fluvoxamine 64-75 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 37-40 18817806-11 2009 In the portion of the brain with affectional pain pathways, 5-HT1 receptor activities induced anti-nociceptive effects and decreased Fos expression with fluvoxamine, while 5-HT2 receptor activation affected to anti-nociceptive effects but did not induce Fos expression. Fluvoxamine 153-164 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 133-136 18655223-5 2008 The calibration line for fluvoxamine showed good linearity in the range of 5-5000 ng/mL (r > 0.999) with the limit of quantification of 5 ng/mL (RSD = 6.51%). Fluvoxamine 25-36 calcium binding protein, spermatid associated 1 Rattus norvegicus 148-155 18978520-0 2008 Dose-dependent effect of the CYP2D6 genotype on the steady-state fluvoxamine concentration. Fluvoxamine 65-76 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 29-35 18978520-3 2008 This study investigated the dose-dependent effect of CYP2D6-variant alleles on the steady-state fluvoxamine concentration. Fluvoxamine 96-107 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 53-59 18978520-9 2008 The present study suggests that the effect of the CYP2D6 genotype on fluvoxamine metabolism is greater at lower doses of fluvoxamine. Fluvoxamine 69-80 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 50-56 18978520-9 2008 The present study suggests that the effect of the CYP2D6 genotype on fluvoxamine metabolism is greater at lower doses of fluvoxamine. Fluvoxamine 121-132 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 50-56 18596927-8 2008 Fluvoxamine (but not sertraline or paroxetine) and the sigma-1 receptor agonists SA4503, PPBP, and DHEA-sulfate significantly potentiated NGF-induced neurite outgrowth in PC12 cells in a concentration-dependent manner. Fluvoxamine 0-11 nerve growth factor Rattus norvegicus 138-141 18596927-9 2008 The potentiation by fluvoxamine and the three sigma-1 receptor agonists was blocked by co-administration of the selective sigma-1 receptor antagonist NE-100, suggesting that sigma-1 receptors play a role in blocking the enhancement of NGF-induced neurite outgrowth. Fluvoxamine 20-31 sigma non-opioid intracellular receptor 1 Rattus norvegicus 122-138 18596927-9 2008 The potentiation by fluvoxamine and the three sigma-1 receptor agonists was blocked by co-administration of the selective sigma-1 receptor antagonist NE-100, suggesting that sigma-1 receptors play a role in blocking the enhancement of NGF-induced neurite outgrowth. Fluvoxamine 20-31 nerve growth factor Rattus norvegicus 235-238 18596927-12 2008 CONCLUSION: These findings suggest that stimulation of sigma-1 receptors and subsequent interaction with IP(3) receptors, PLC-gamma, PI3K, p38MAPK, JNK, and the Ras/Raf/MAPK signaling pathways are involved in the mechanisms of action of sigma-1 receptor agonists such as fluvoxamine and SA4503. Fluvoxamine 271-282 sigma non-opioid intracellular receptor 1 Rattus norvegicus 55-71 18609433-2 2009 Fluvoxamine, a SSRI, is mainly metabolized by cytochrome P450 (CYP) 2D6 and 1A2. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-79 19043208-7 2008 Fluvoxamine-treated rats showed significantly lower neuropeptide Y (NPY) immunostaining levels in the paraventricular nucleus (PVN) and dorsomedial hypothalamic nucleus (DMH) than untreated controls. Fluvoxamine 0-11 neuropeptide Y Rattus norvegicus 52-66 19043208-7 2008 Fluvoxamine-treated rats showed significantly lower neuropeptide Y (NPY) immunostaining levels in the paraventricular nucleus (PVN) and dorsomedial hypothalamic nucleus (DMH) than untreated controls. Fluvoxamine 0-11 neuropeptide Y Rattus norvegicus 68-71 19043208-8 2008 Hypophagic and weight-inhibiting effects of fluvoxamine might be mediated via decreased NPY in PVN and DMH. Fluvoxamine 44-55 neuropeptide Y Rattus norvegicus 88-91 18655786-13 2008 Furthermore, differences in 5-HT transporter binding may cause variations in responses to fluvoxamine. Fluvoxamine 90-101 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 28-44 18816299-3 2008 In vitro, fluvoxamine, tolfenamic acid, mefenamic acid and rofecoxib potently inhibited CYP1A2 [the 50% inhibitory concentration (IC(50)) < 10 microM]. Fluvoxamine 10-21 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 88-94 18596927-3 2008 The SSRI fluvoxamine, with sigma-1 receptor agonism, is shown to potentiate nerve-growth factor (NGF)-induced neurite outgrowth in PC 12 cells. Fluvoxamine 9-20 sigma non-opioid intracellular receptor 1 Rattus norvegicus 27-43 18596927-3 2008 The SSRI fluvoxamine, with sigma-1 receptor agonism, is shown to potentiate nerve-growth factor (NGF)-induced neurite outgrowth in PC 12 cells. Fluvoxamine 9-20 nerve growth factor Rattus norvegicus 76-95 18596927-3 2008 The SSRI fluvoxamine, with sigma-1 receptor agonism, is shown to potentiate nerve-growth factor (NGF)-induced neurite outgrowth in PC 12 cells. Fluvoxamine 9-20 nerve growth factor Rattus norvegicus 97-100 18596927-5 2008 In this study, we examined the roles of cellular signaling pathways in the potentiation of NGF-induced neurite outgrowth by fluvoxamine and sigma-1 receptor agonists. Fluvoxamine 124-135 nerve growth factor Rattus norvegicus 91-94 18691982-11 2008 Fluoxetine and paroxetine are potent inhibitors of CYP2D6, fluvoxamine markedly inhibits CYP1A2 and CYP2C19, and nefazodone is a substantial inhibitor of CYP3A4. Fluvoxamine 59-70 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 51-57 18691982-11 2008 Fluoxetine and paroxetine are potent inhibitors of CYP2D6, fluvoxamine markedly inhibits CYP1A2 and CYP2C19, and nefazodone is a substantial inhibitor of CYP3A4. Fluvoxamine 59-70 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 89-95 18691982-11 2008 Fluoxetine and paroxetine are potent inhibitors of CYP2D6, fluvoxamine markedly inhibits CYP1A2 and CYP2C19, and nefazodone is a substantial inhibitor of CYP3A4. Fluvoxamine 59-70 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 100-107 16959056-0 2007 Fluvoxamine, a selective serotonin reuptake inhibitor, and 5-HT2C receptor inactivation induce appetite-suppressing effects in mice via 5-HT1B receptors. Fluvoxamine 0-11 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 136-142 18482738-5 2008 Sertraline, fluoxetine, fluvoxamine, citalopram and paroxetine all significantly shortened the period of Period1-bioluminescence rhythms in rat-1 fibroblasts. Fluvoxamine 24-35 period circadian regulator 1 Rattus norvegicus 105-112 18690879-2 2008 Recent human studies have shown that the magnitude of inhibitory interactions caused by the reversible CYP1A2 inhibitor fluvoxamine decreases as liver function worsens, and virtually vanishes in patients with more advanced hepatocellular insufficiency. Fluvoxamine 120-131 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 103-109 17709099-5 2007 The structure-activity relationship readily distinguished the pro-apoptotic and growth inhibitory effect of SSRIs from their eponymous action (blockage of the serotonin transporter): acetylation of the SSRIs fluvoxamine and paroxetine abrogated the ability of these compounds to inhibit 5HT-uptake, but did not impair their cytotoxic action. Fluvoxamine 208-219 solute carrier family 6 member 4 Homo sapiens 159-180 18438654-7 2008 CONCLUSION: Co-administration of the CYP1A2 inhibitor fluvoxamine with ramosetron resulted in an interaction. Fluvoxamine 54-65 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-43 17920044-8 2007 Other SSRIs, sertraline and fluvoxamine, also inhibited Kir4.1 channel currents whereas the tetracyclic (mianserin) or the 5-HT1A receptor-related (buspirone) antidepressants did not. Fluvoxamine 28-39 potassium inwardly rectifying channel subfamily J member 10 Homo sapiens 56-62 16959056-2 2007 Here, we report that fluvoxamine (3-30 mg/kg), a selective serotonin reuptake inhibitor (SSRI), in the presence of SB 242084 (1-2 mg/kg), a selective 5-HT2C receptor antagonist, exerts appetite-suppressing effects while fluvoxamine or SB 242084 alone has no effect. Fluvoxamine 21-32 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 150-165 16959056-5 2007 These results suggest that fluvoxamine and inactivation of 5-HT2C receptors exert feeding suppression through activation of 5-HT1B receptors, and that 5-HT1B receptors up-regulate hypothalamic POMC and CART gene expression and down-regulate hypothalamic orexin gene expression in mice. Fluvoxamine 27-38 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 124-130 16959056-5 2007 These results suggest that fluvoxamine and inactivation of 5-HT2C receptors exert feeding suppression through activation of 5-HT1B receptors, and that 5-HT1B receptors up-regulate hypothalamic POMC and CART gene expression and down-regulate hypothalamic orexin gene expression in mice. Fluvoxamine 27-38 hypocretin Mus musculus 254-260 17823102-5 2007 The primary pharmacokinetic interactions with smoking occur with drugs that are CYP1A2 substrates, such as caffeine, clozapine, fluvoxamine, olanzapine, tacrine, and theophylline. Fluvoxamine 128-139 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 80-86 17662512-7 2007 The mRNA expression of chemokine receptors, IL8RA and CCR1, and of RGS7 was significantly down-regulated following fluvoxamine augmentation. Fluvoxamine 115-126 C-X-C motif chemokine receptor 1 Homo sapiens 44-49 17662512-7 2007 The mRNA expression of chemokine receptors, IL8RA and CCR1, and of RGS7 was significantly down-regulated following fluvoxamine augmentation. Fluvoxamine 115-126 C-C motif chemokine receptor 1 Homo sapiens 54-58 17662512-7 2007 The mRNA expression of chemokine receptors, IL8RA and CCR1, and of RGS7 was significantly down-regulated following fluvoxamine augmentation. Fluvoxamine 115-126 regulator of G protein signaling 7 Homo sapiens 67-71 17092970-0 2007 The G196A polymorphism of the brain-derived neurotrophic factor gene and the antidepressant effect of milnacipran and fluvoxamine. Fluvoxamine 118-129 brain derived neurotrophic factor Homo sapiens 30-63 17092970-2 2007 The purpose of the present study was to investigate whether the G196A polymorphism of the brain-derived neurotrophic factor (BDNF) gene is associated with the antidepressant effect of milnacipran, a serotonin norepinephrine reuptake inhibitor, and fluvoxamine, a selective serotonin reuptake inhibitor. Fluvoxamine 248-259 brain derived neurotrophic factor Homo sapiens 90-123 17092970-2 2007 The purpose of the present study was to investigate whether the G196A polymorphism of the brain-derived neurotrophic factor (BDNF) gene is associated with the antidepressant effect of milnacipran, a serotonin norepinephrine reuptake inhibitor, and fluvoxamine, a selective serotonin reuptake inhibitor. Fluvoxamine 248-259 brain derived neurotrophic factor Homo sapiens 125-129 17092970-7 2007 In all subjects receiving milnacipran or fluvoxamine, the G/A genotype of the BDNF G196A polymorphism was associated with a significantly better therapeutic effect in the MADRS scores during this study. Fluvoxamine 41-52 brain derived neurotrophic factor Homo sapiens 78-82 17092970-9 2007 These results suggest that the BDNF G196A polymorphism in part determines the antidepressant effect of both milnacipran and fluvoxamine. Fluvoxamine 124-135 brain derived neurotrophic factor Homo sapiens 31-35 17318509-9 2007 Furthermore, when 5-HTnergic neuronal function was examined using antibodies against tryptophan hydroxylase (TPH) following the behavioral tests, fluvoxamine significantly reversed the loss of TPH-positive cells produced by OBX in the dorsal raphe. Fluvoxamine 146-157 tryptophan hydroxylase 1 Rattus norvegicus 85-107 17431033-9 2007 Fluvoxamine, an inhibitor of CYP2C19, inhibited timolol metabolism to a lesser extent, confirming its minor contribution. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 29-36 17481608-5 2007 Pretreatment with the selective serotonin reuptake inhibitor fluvoxamine, the relatively selective noradrenaline reuptake inhibitor reboxetine, or the non-selective monoaminergic reuptake inhibitor imipramine, significantly inhibited IL-6 and NO production in a dose-dependent manner. Fluvoxamine 61-72 interleukin 6 Mus musculus 234-238 17504220-6 2007 Interactions mediated by potent CYP1A2 inhibitors (such as fluvoxamine) or inducers (like cigarette smoke) appear to be consistent, predictable and usually clinically significant. Fluvoxamine 59-70 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 32-38 17516189-4 2007 The likely cause of the prolonged delirium was the interaction of promethazine and fluvoxamine through the inhibition of the CYP2D6 enzyme. Fluvoxamine 83-94 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 125-131 17254018-10 2007 The selective 5-HT reuptake inhibitor fluvoxamine (50 and 100 pmol) significantly decreased 5-HT clearance in BDNF+/+ mice, but not in BDNF+/- mice. Fluvoxamine 38-49 brain derived neurotrophic factor Mus musculus 110-114 17318509-9 2007 Furthermore, when 5-HTnergic neuronal function was examined using antibodies against tryptophan hydroxylase (TPH) following the behavioral tests, fluvoxamine significantly reversed the loss of TPH-positive cells produced by OBX in the dorsal raphe. Fluvoxamine 146-157 tryptophan hydroxylase 1 Rattus norvegicus 109-112 17318509-9 2007 Furthermore, when 5-HTnergic neuronal function was examined using antibodies against tryptophan hydroxylase (TPH) following the behavioral tests, fluvoxamine significantly reversed the loss of TPH-positive cells produced by OBX in the dorsal raphe. Fluvoxamine 146-157 tryptophan hydroxylase 1 Rattus norvegicus 193-196 17318509-10 2007 CONCLUSION: We demonstrated that chronic treatment with milnacipran or fluvoxamine was effective to improve both the hyperemotional behavior and the loss of TPH-positive cells seen in OBX rats. Fluvoxamine 71-82 tryptophan hydroxylase 1 Rattus norvegicus 157-160 17417072-0 2007 Dose-dependent effects of the 3435 C>T genotype of ABCB1 gene on the steady-state plasma concentration of fluvoxamine in psychiatric patients. Fluvoxamine 109-120 ATP binding cassette subfamily B member 1 Homo sapiens 54-59 17307149-2 2007 In order to gain insights for developing personalized drugs, the 3D (dimensional) structure of CYP2C19 has been developed based on the crystal structure of CYP2C9 (PDB code 1R90), and its structure-activity relationship with the ligands of CEC, Fluvoxamine, Lescol, and Ticlopidine investigated through the structure-activity relationship approach. Fluvoxamine 245-256 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 95-102 17307149-2 2007 In order to gain insights for developing personalized drugs, the 3D (dimensional) structure of CYP2C19 has been developed based on the crystal structure of CYP2C9 (PDB code 1R90), and its structure-activity relationship with the ligands of CEC, Fluvoxamine, Lescol, and Ticlopidine investigated through the structure-activity relationship approach. Fluvoxamine 245-256 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 17417072-1 2007 UNLABELLED: This study investigated effects of the 3435 C>T genotype of the adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1, MDR1) gene on the steady-state plasma concentration of fluvoxamine (FLV). Fluvoxamine 203-214 ATP binding cassette subfamily B member 1 Homo sapiens 79-139 17417072-1 2007 UNLABELLED: This study investigated effects of the 3435 C>T genotype of the adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1, MDR1) gene on the steady-state plasma concentration of fluvoxamine (FLV). Fluvoxamine 203-214 ATP binding cassette subfamily B member 1 Homo sapiens 141-146 17417072-1 2007 UNLABELLED: This study investigated effects of the 3435 C>T genotype of the adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1, MDR1) gene on the steady-state plasma concentration of fluvoxamine (FLV). Fluvoxamine 203-214 ATP binding cassette subfamily B member 1 Homo sapiens 148-152 17417072-1 2007 UNLABELLED: This study investigated effects of the 3435 C>T genotype of the adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1, MDR1) gene on the steady-state plasma concentration of fluvoxamine (FLV). Fluvoxamine 216-219 ATP binding cassette subfamily B member 1 Homo sapiens 79-139 17417072-1 2007 UNLABELLED: This study investigated effects of the 3435 C>T genotype of the adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1, MDR1) gene on the steady-state plasma concentration of fluvoxamine (FLV). Fluvoxamine 216-219 ATP binding cassette subfamily B member 1 Homo sapiens 141-146 17417072-1 2007 UNLABELLED: This study investigated effects of the 3435 C>T genotype of the adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1, MDR1) gene on the steady-state plasma concentration of fluvoxamine (FLV). Fluvoxamine 216-219 ATP binding cassette subfamily B member 1 Homo sapiens 148-152 16495935-4 2007 Furthermore, the effect of fluvoxamine on PCP-induced cognitive deficits was antagonized by co-administration of the selective sigma-1 receptor antagonist NE-100 (1 mg/kg/day). Fluvoxamine 27-38 sigma non-opioid intracellular receptor 1 Mus musculus 127-143 16495935-9 2007 Therefore, sigma-1 receptor agonists such as fluvoxamine would be potential therapeutic drugs for the treatment of the cognitive deficits of schizophrenia. Fluvoxamine 45-56 sigma non-opioid intracellular receptor 1 Mus musculus 11-27 16814933-4 2006 Subchronic treatment of naive rats with imipramine or fluvoxamine significantly decreased the expression of synapsin I. Fluvoxamine 54-65 synapsin I Rattus norvegicus 108-118 17166674-6 2007 The fact that fluvoxamine, a potent cytochrome P450 (CYP) 1A2 inhibitor, reduced tacrine toxicity and the expression of the CYP 1A2 gene was maintained in gel entrapped hepatocytes, but not in hepatocyte monolayers, could illustrate a close association between CYP 1A2 expression levels and tacrine toxicity. Fluvoxamine 14-25 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 36-61 17166674-6 2007 The fact that fluvoxamine, a potent cytochrome P450 (CYP) 1A2 inhibitor, reduced tacrine toxicity and the expression of the CYP 1A2 gene was maintained in gel entrapped hepatocytes, but not in hepatocyte monolayers, could illustrate a close association between CYP 1A2 expression levels and tacrine toxicity. Fluvoxamine 14-25 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 124-131 17166674-6 2007 The fact that fluvoxamine, a potent cytochrome P450 (CYP) 1A2 inhibitor, reduced tacrine toxicity and the expression of the CYP 1A2 gene was maintained in gel entrapped hepatocytes, but not in hepatocyte monolayers, could illustrate a close association between CYP 1A2 expression levels and tacrine toxicity. Fluvoxamine 14-25 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 261-268 17596106-1 2007 OBJECTIVE: To investigate the effects of steady-state dosing of fluvoxamine, an inhibitor of cytochrome P450 (CYP) 1A2 and CYP2C19, on the pharmacokinetics of roflumilast, an oral, once-daily phosphodiesterase 4 (PDE4) inhibitor and its pharmacodynamically active metabolite roflumilast N-oxide. Fluvoxamine 64-75 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 93-118 17596106-1 2007 OBJECTIVE: To investigate the effects of steady-state dosing of fluvoxamine, an inhibitor of cytochrome P450 (CYP) 1A2 and CYP2C19, on the pharmacokinetics of roflumilast, an oral, once-daily phosphodiesterase 4 (PDE4) inhibitor and its pharmacodynamically active metabolite roflumilast N-oxide. Fluvoxamine 64-75 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 123-130 17596106-1 2007 OBJECTIVE: To investigate the effects of steady-state dosing of fluvoxamine, an inhibitor of cytochrome P450 (CYP) 1A2 and CYP2C19, on the pharmacokinetics of roflumilast, an oral, once-daily phosphodiesterase 4 (PDE4) inhibitor and its pharmacodynamically active metabolite roflumilast N-oxide. Fluvoxamine 64-75 phosphodiesterase 4A Homo sapiens 213-217 17596106-8 2007 The combined effect of fluvoxamine co-administration on roflumilast and roflumilast N-oxide exposures resulted in a moderate (i.e. 59%) increase in total PDE4 inhibitory activity. Fluvoxamine 23-34 phosphodiesterase 4A Homo sapiens 154-158 17596106-9 2007 CONCLUSION: Co-administration of roflumilast and fluvoxamine affects the disposition of roflumilast and its active metabolite roflumilast N-oxide most likely via a potent dual pathway inhibition of CYP1A2 and CYP2C19 by fluvoxamine. Fluvoxamine 49-60 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 198-204 17596106-9 2007 CONCLUSION: Co-administration of roflumilast and fluvoxamine affects the disposition of roflumilast and its active metabolite roflumilast N-oxide most likely via a potent dual pathway inhibition of CYP1A2 and CYP2C19 by fluvoxamine. Fluvoxamine 49-60 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 209-216 17045169-5 2006 Dynamic PET studies using were performed on three controls and two patients, one treated with the antidepressant and inhibitor of cytochrome CYP1A2 fluvoxamine, the other suffering from liver cirrhosis. Fluvoxamine 148-159 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 141-147 17484519-0 2007 Identification of human cytochrome P450 enzymes involved in the major metabolic pathway of fluvoxamine. Fluvoxamine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 17484519-4 2007 The formation of fluvoxamino alcohol from fluvoxamine in pooled human liver microsomes was significantly inhibited by quinidine, a relatively specific CYP2D6 inhibitor, with a Ki value of 2.2 microM, whereas other several relatively specific CYP inhibitors did not inhibit the formation of fluvoxamino alcohol. Fluvoxamine 42-53 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 151-157 17484519-4 2007 The formation of fluvoxamino alcohol from fluvoxamine in pooled human liver microsomes was significantly inhibited by quinidine, a relatively specific CYP2D6 inhibitor, with a Ki value of 2.2 microM, whereas other several relatively specific CYP inhibitors did not inhibit the formation of fluvoxamino alcohol. Fluvoxamine 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 17484519-7 2007 These data suggest that CYP2D6 is the only enzyme predominantly responsible for the first-step oxidation of fluvoxamine to fluvoxamino alcohol, and alcohol dehydrogenase is involved in the second-step oxidation of fluvoxamino alcohol to the corresponding carbolic acid. Fluvoxamine 108-119 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 24-30 17484519-7 2007 These data suggest that CYP2D6 is the only enzyme predominantly responsible for the first-step oxidation of fluvoxamine to fluvoxamino alcohol, and alcohol dehydrogenase is involved in the second-step oxidation of fluvoxamino alcohol to the corresponding carbolic acid. Fluvoxamine 108-119 aldo-keto reductase family 1 member A1 Homo sapiens 148-169 17214606-12 2007 Of a particular clinical significance is the interaction between fluvoxamine, a potent CYP1A2 inhibitor, and clozapine. Fluvoxamine 65-76 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 87-93 17596106-9 2007 CONCLUSION: Co-administration of roflumilast and fluvoxamine affects the disposition of roflumilast and its active metabolite roflumilast N-oxide most likely via a potent dual pathway inhibition of CYP1A2 and CYP2C19 by fluvoxamine. Fluvoxamine 220-231 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 198-204 17596106-10 2007 The exposure increases observed for roflumilast N-oxide are suggested to be attributable to CYP2C19 co-inhibition by fluvoxamine and thus, are not to be expected to occur when roflumilast is co-administered with more selective CYP1A2 inhibitors. Fluvoxamine 117-128 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 92-99 16856883-11 2006 Fluvoxamine increased the AUC(0-infinity) and t(1/2) of rosiglitazone moderately and hence may be a weak inhibitor of CYP2C8. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 8 Homo sapiens 118-124 16985100-8 2006 Fluvoxamine decreased the rofecoxib-caused inactivation of CYP1A2 in a concentration-dependent manner. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 59-65 17240844-3 2006 We recently reported that chronic treatment with fluvoxamine or paroxetine desensitized 5-HT2C receptor function. Fluvoxamine 49-60 5-hydroxytryptamine receptor 2C Homo sapiens 88-94 16846619-0 2006 Fluvoxamine, a selective serotonin reuptake inhibitor, exerts its antiallodynic effects on neuropathic pain in mice via 5-HT2A/2C receptors. Fluvoxamine 0-11 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 120-126 16846619-6 2006 The antiallodynic effects of systemic fluvoxamine were also reduced by both systemic and intrathecal administration of ketanserin, a 5-HT2A/2C receptor antagonist. Fluvoxamine 38-49 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 133-139 16846619-8 2006 These results indicate that fluvoxamine exerts its antiallodynic effects on neuropathic pain via descending 5-HT fibers and spinal 5-HT2A or 5-HT2C receptors, and the antinociception on acute mechanical pain via 5-HT3 receptors. Fluvoxamine 28-39 5-hydroxytryptamine (serotonin) receptor 2A Mus musculus 131-137 16846619-8 2006 These results indicate that fluvoxamine exerts its antiallodynic effects on neuropathic pain via descending 5-HT fibers and spinal 5-HT2A or 5-HT2C receptors, and the antinociception on acute mechanical pain via 5-HT3 receptors. Fluvoxamine 28-39 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 141-147 16814933-6 2006 Subchronic treatment of naive rats with fluvoxamine but not imipramine showed a tendency to decrease the expression of synapsin I. Fluvoxamine 40-51 synapsin I Rattus norvegicus 119-129 16874005-0 2006 Effects of the serotonin type 2A, 3A and 3B receptor and the serotonin transporter genes on paroxetine and fluvoxamine efficacy and adverse drug reactions in depressed Japanese patients. Fluvoxamine 107-118 solute carrier family 6 member 4 Homo sapiens 61-82 16918719-2 2006 We studied the effect of co-administration of fluvoxamine (CYP1A2 inhibitor) and erythromycin (CYP3A4 inhibitor) on the pharmacokinetics of lidocaine in a double-blind, randomized, three-way cross-over study. Fluvoxamine 46-57 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 59-65 16918719-10 2006 We conclude that inhibition of CYP1A2 by fluvoxamine considerably reduces the presystemic metabolism of oral lidocaine and may increase the risk of lidocaine toxicity if lidocaine is ingested. Fluvoxamine 41-52 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 31-37 16522641-12 2006 We found that the BDNF-stimulated PLC-gamma activation and the ensued increase in intracellular Ca2+ ([Ca2+]i) were potentiated by pretreatment with imipramine or fluvoxamine, so was the BDNF-induced glutamate release. Fluvoxamine 163-174 brain derived neurotrophic factor Homo sapiens 18-22 16522641-12 2006 We found that the BDNF-stimulated PLC-gamma activation and the ensued increase in intracellular Ca2+ ([Ca2+]i) were potentiated by pretreatment with imipramine or fluvoxamine, so was the BDNF-induced glutamate release. Fluvoxamine 163-174 brain derived neurotrophic factor Homo sapiens 187-191 16678550-0 2006 Liver dysfunction markedly decreases the inhibition of cytochrome P450 1A2-mediated theophylline metabolism by fluvoxamine. Fluvoxamine 111-122 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 55-74 16678550-1 2006 BACKGROUND AND OBJECTIVES: In vivo inhibition of cytochrome P450 (CYP) 1A2 by fluvoxamine causes a reduction in the clearance of the high-extraction drug lidocaine, which decreases in proportion to the degree of liver dysfunction. Fluvoxamine 78-89 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 49-74 16678550-12 2006 Two mechanisms, as follows in order of importance, are responsible for the effect of liver dysfunction: (1) decreased sensitivity to fluvoxamine of CYP1A2-mediated biotransformations in the cirrhotic liver, probably resulting from reduced uptake of the inhibitory drug, and (2) reduced hepatic expression of CYP1A2, which makes its contribution to overall drug elimination less important. Fluvoxamine 133-144 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 148-154 16678550-12 2006 Two mechanisms, as follows in order of importance, are responsible for the effect of liver dysfunction: (1) decreased sensitivity to fluvoxamine of CYP1A2-mediated biotransformations in the cirrhotic liver, probably resulting from reduced uptake of the inhibitory drug, and (2) reduced hepatic expression of CYP1A2, which makes its contribution to overall drug elimination less important. Fluvoxamine 133-144 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 308-314 16487224-0 2006 Different effects of fluvoxamine on rabeprazole pharmacokinetics in relation to CYP2C19 genotype status. Fluvoxamine 21-32 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 80-87 16487224-8 2006 There was a significant difference in fluvoxamine-mediated percentage increase in AUC(0,infinity) of rabeprazole and rabeprazole thioether between CYP2C19 genotypes. Fluvoxamine 38-49 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 147-154 16487224-9 2006 CONCLUSIONS: The present study indicates that there are significant drug interactions between rabeprazole and fluvoxamine in EMs of CYP2C19. Fluvoxamine 110-121 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 132-139 16488409-3 2006 Administration of the non-selective MAO inhibitor, pargyline, and the selective MAO-A inhibitor, clorgyline, resulted in 5-HT syndrome in 5-HTP-treated rats, and subchronic co-administration of fluvoxamine intensified the syndrome. Fluvoxamine 194-205 monoamine oxidase A Rattus norvegicus 80-85 16360124-6 2006 These results suggest that the desensitization of 5-HT2A receptor function occurs in the same way as that of 5-HT2C receptor function through chronic treatment with either fluvoxamine or paroxetine as a consequence of prolonged exposure to elevated levels of serotonin. Fluvoxamine 172-183 5-hydroxytryptamine receptor 2A Homo sapiens 50-65 16360124-6 2006 These results suggest that the desensitization of 5-HT2A receptor function occurs in the same way as that of 5-HT2C receptor function through chronic treatment with either fluvoxamine or paroxetine as a consequence of prolonged exposure to elevated levels of serotonin. Fluvoxamine 172-183 5-hydroxytryptamine receptor 2C Rattus norvegicus 109-115 16546220-4 2006 We examined the effects of chronic administration of imipramine, fluvoxamine, maprotiline and, as a negative control, cocaine on the level of G(olf) protein in the rat striatum. Fluvoxamine 65-76 G protein subunit alpha L Rattus norvegicus 142-148 16546220-8 2006 Chronic fluvoxamine and maprotiline treatment (20 mg/kg for 2 weeks) also significantly increased the level of G(olf) (by 9% and 25%, respectively), but cocaine did not alter it significantly. Fluvoxamine 8-19 G protein subunit alpha L Rattus norvegicus 111-117 16633150-0 2006 Time course of in vivo 5-HTT transporter occupancy by fluvoxamine. Fluvoxamine 54-65 huntingtin Homo sapiens 25-28 16633150-2 2006 In this study, we measured the time course of the selective serotonin reuptake inhibitor fluvoxamine in the human brain based on serotonin transporter (5-HTT) occupancy by positron emission tomography. Fluvoxamine 89-100 huntingtin Homo sapiens 154-157 16633150-6 2006 The relationship between the plasma concentration of fluvoxamine and 5-HTT occupancy at these different time points was fitted to the law of mass action. Fluvoxamine 53-64 huntingtin Homo sapiens 71-74 16205777-0 2006 Polymorphisms in the 5-hydroxytryptamine 2A receptor and CytochromeP4502D6 genes synergistically predict fluvoxamine-induced side effects in japanese depressed patients. Fluvoxamine 105-116 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 57-74 16205777-2 2006 CytochromeP450 (CYP) 2D6 may also be associated with the side effects induced by fluvoxamine, since the plasma fluvoxamine concentration depends on a CYP2D6 gene polymorphism. Fluvoxamine 81-92 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-24 16205777-2 2006 CytochromeP450 (CYP) 2D6 may also be associated with the side effects induced by fluvoxamine, since the plasma fluvoxamine concentration depends on a CYP2D6 gene polymorphism. Fluvoxamine 81-92 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 150-156 16205777-2 2006 CytochromeP450 (CYP) 2D6 may also be associated with the side effects induced by fluvoxamine, since the plasma fluvoxamine concentration depends on a CYP2D6 gene polymorphism. Fluvoxamine 111-122 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-24 16205777-2 2006 CytochromeP450 (CYP) 2D6 may also be associated with the side effects induced by fluvoxamine, since the plasma fluvoxamine concentration depends on a CYP2D6 gene polymorphism. Fluvoxamine 111-122 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 150-156 16205777-12 2006 5-HT2A receptor and CYP2D6 gene polymorphisms had a synergistic effect for the prediction of fluvoxamine-induced gastrointestinal side effects. Fluvoxamine 93-104 5-hydroxytryptamine receptor 2A Homo sapiens 0-15 16205777-12 2006 5-HT2A receptor and CYP2D6 gene polymorphisms had a synergistic effect for the prediction of fluvoxamine-induced gastrointestinal side effects. Fluvoxamine 93-104 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 20-26 16893613-0 2006 Co-administration of ramelton and fluvoxamine to increase levels of interleukin-2. Fluvoxamine 34-45 interleukin 2 Homo sapiens 68-81 16893613-5 2006 It turns out that the medication fluvoxamine approved by the FDA for the treatment of obsessive compulsive disorder is a potent inhibitor of the CYP1A2 enzyme, with the effect that co-administration of ramelton and fluvoxamine increases blood levels of ramelton by 100-200 fold. Fluvoxamine 33-44 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 145-151 16893613-5 2006 It turns out that the medication fluvoxamine approved by the FDA for the treatment of obsessive compulsive disorder is a potent inhibitor of the CYP1A2 enzyme, with the effect that co-administration of ramelton and fluvoxamine increases blood levels of ramelton by 100-200 fold. Fluvoxamine 215-226 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 145-151 16893613-7 2006 Thus, here we point out that co-administration of ramelton and modest doses of fluvoxamine may be able to smoothly produce increased levels of Il-2, this may be useful in diseases and conditions such as metastatic cancer and maintenance of suppression of the HIV virus. Fluvoxamine 79-90 interleukin 2 Homo sapiens 143-147 17356306-0 2006 Association study between vesicle-associated membrane protein 2 gene polymorphisms and fluvoxamine response in Japanese major depressive patients. Fluvoxamine 87-98 vesicle associated membrane protein 2 Homo sapiens 26-63 17356306-3 2006 The purpose of this study was to investigate whether the VAMP2 gene is associated with clinical responses to a specific antidepressant, fluvoxamine. Fluvoxamine 136-147 vesicle associated membrane protein 2 Homo sapiens 57-62 16874005-1 2006 In this study, we tested the influence of the serotonin type 2A, 3A and 3B receptor genes (HTR2A, HTR3A, HTR3B) in addition to a polymorphism in the promoter region of the serotonin transporter (SERTPR), and investigated the different characteristics of clinical responses to paroxetine and fluvoxamine. Fluvoxamine 291-302 solute carrier family 6 member 4 Homo sapiens 172-193 16874005-4 2006 Patients with the l allele of SERTPR showed a better response to SSRIs than s/s genotype carriers (p = 0.015-0.042), more significantly to fluvoxamine. Fluvoxamine 139-150 solute carrier family 6 member 4 Homo sapiens 30-36 16874005-5 2006 The -1438G/G genotype of HTR2A was associated with a good response to SSRIs (p = 0.010-0.039), especially to fluvoxamine, and significantly with severe nausea in paroxetine-treated patients (p = 0.013). Fluvoxamine 109-120 5-hydroxytryptamine receptor 2A Homo sapiens 25-30 16226034-8 2005 However, higher doses of fluvoxamine may elevate plasma risperidone levels, presumably as a result of a dose-dependent inhibitory effect of fluvoxamine on CYP2D6-and/or CYP3A4-mediated 9-hydroxylation of risperidone. Fluvoxamine 25-36 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 155-161 16282844-2 2005 The antidepressant fluvoxamine is an inhibitor of cytochrome P450 3A4. Fluvoxamine 19-30 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 50-69 16226034-8 2005 However, higher doses of fluvoxamine may elevate plasma risperidone levels, presumably as a result of a dose-dependent inhibitory effect of fluvoxamine on CYP2D6-and/or CYP3A4-mediated 9-hydroxylation of risperidone. Fluvoxamine 25-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 169-175 16226034-8 2005 However, higher doses of fluvoxamine may elevate plasma risperidone levels, presumably as a result of a dose-dependent inhibitory effect of fluvoxamine on CYP2D6-and/or CYP3A4-mediated 9-hydroxylation of risperidone. Fluvoxamine 140-151 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 155-161 16226034-8 2005 However, higher doses of fluvoxamine may elevate plasma risperidone levels, presumably as a result of a dose-dependent inhibitory effect of fluvoxamine on CYP2D6-and/or CYP3A4-mediated 9-hydroxylation of risperidone. Fluvoxamine 140-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 169-175 16236038-1 2005 BACKGROUND: Coadministration of fluvoxamine impairs the clearance of caffeine and prolongs its elimination half-life, which is attributable to inhibition of CYP1A2 by fluvoxamine. Fluvoxamine 32-43 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 157-163 16359723-6 2005 Furthermore, as with selective serotonin reuptake inhibitors such as fluvoxamine and paroxetine, SIV was also reduced by the serotonin and noradrenaline reuptake inhibitor milnacipran and the metabotropic glutamate receptor 5 antagonist MPEP, while desipramine was without effect. Fluvoxamine 69-80 glutamate metabotropic receptor 5 Rattus norvegicus 192-225 16236038-1 2005 BACKGROUND: Coadministration of fluvoxamine impairs the clearance of caffeine and prolongs its elimination half-life, which is attributable to inhibition of CYP1A2 by fluvoxamine. Fluvoxamine 167-178 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 157-163 15963095-0 2005 Enantioselective disposition of lansoprazole in relation to CYP2C19 genotypes in the presence of fluvoxamine. Fluvoxamine 97-108 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 60-67 16079297-9 2005 Furthermore, the time-dependent change of SERT mRNA expression and uptake activity in the midbrain is suggested to be the mechanism underlying the 24-h rhythm of anti-immobility effect of fluvoxamine. Fluvoxamine 188-199 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 42-46 15963095-2 2005 The aim of this study was to examine the effects of fluvoxamine, a CYP2C19 inhibitor, on the pharmacokinetics of each lansoprazole enantiomer among three different CYP2C19 genotype groups. Fluvoxamine 52-63 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 67-74 15963095-7 2005 The mean fluvoxamine-mediated percent increase in the AUC(0, infinity) of (R)-lansoprazole in the homEMs compared with the PMs was significant (P = 0.0117); however, C(max) did not differ among the three CYP2C19 genotypes. Fluvoxamine 9-20 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 204-211 16119478-0 2005 Clinical efficacy of fluvoxamine and functional polymorphism in a serotonin transporter gene on childhood autism. Fluvoxamine 21-32 solute carrier family 6 member 4 Homo sapiens 66-87 16119478-1 2005 We studied the correlation between response to fluvoxamine and serotonin transporter gene promoter region polymorphism (5-HTTLPR). Fluvoxamine 47-58 solute carrier family 6 member 4 Homo sapiens 63-84 16119478-1 2005 We studied the correlation between response to fluvoxamine and serotonin transporter gene promoter region polymorphism (5-HTTLPR). Fluvoxamine 47-58 solute carrier family 6 member 4 Homo sapiens 120-128 16119478-7 2005 5-HTTLPR may influence the individual responses to fluvoxamine administration. Fluvoxamine 51-62 solute carrier family 6 member 4 Homo sapiens 0-8 15845683-2 2005 We studied the effect of coadministration of the antidepressant fluvoxamine (CYP1A2 inhibitor) and antimicrobial drug erythromycin (CYP3A4 inhibitor) on lidocaine pharmacokinetics in a double-blind, randomized, three-way crossover study. Fluvoxamine 64-75 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 77-83 15845683-8 2005 We conclude that inhibition of CYP1A2 by fluvoxamine considerably reduces elimination of lidocaine and may increase the risk of lidocaine toxicity. Fluvoxamine 41-52 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 31-37 15383832-12 2005 These findings suggest that the combination of prefrontal DA-D2/3 receptor blockade and 5-HT1A receptor activation in regions other than the cortex plays an important role in sulpiride and fluvoxamine-induced increase in prefrontal DA release. Fluvoxamine 189-200 5-hydroxytryptamine receptor 1A Rattus norvegicus 88-94 15719219-3 2005 OBJECTIVES: This study was conducted to examine the effects of chronic pretreatment with the SSRIs fluvoxamine and paroxetine on the facilitation of ejaculation induced by the 5-HT1A receptor agonist 8-OH-DPAT. Fluvoxamine 99-110 5-hydroxytryptamine receptor 1A Rattus norvegicus 176-182 15738749-5 2005 The present prospective study tested the hypothesis that olanzapine steady-state doses can be significantly decreased by coadministration of a low subclinical dose of fluvoxamine, a potent inhibitor of cytochrome P450 1A2. Fluvoxamine 167-178 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 202-221 15738749-11 2005 4"-N-desmethylolanzapine/olanzapine metabolic ratio decreased from 0.45 +/- 0.20 at baseline to 0.25 +/- 0.11 at week 6, suggesting inhibition of the cytochrome P450 1A2-mediated olanzapine 4"-N-demethylation by fluvoxamine (P < 0.05). Fluvoxamine 212-223 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 150-169 15678084-3 2005 Oral administration of fluvoxamine, fluoxetine, paroxetine and sertraline at pharmacologically relevant doses exerted dose- and time-dependent binding activity of brain SERT as revealed by significant increases in KD for specific [3H]paroxetine binding, and the in vivo SERT-binding potency was in the order of paroxetine>>fluoxetine, sertraline>fluvoxamine. Fluvoxamine 23-34 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 169-173 15678084-3 2005 Oral administration of fluvoxamine, fluoxetine, paroxetine and sertraline at pharmacologically relevant doses exerted dose- and time-dependent binding activity of brain SERT as revealed by significant increases in KD for specific [3H]paroxetine binding, and the in vivo SERT-binding potency was in the order of paroxetine>>fluoxetine, sertraline>fluvoxamine. Fluvoxamine 23-34 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 270-274 15678084-3 2005 Oral administration of fluvoxamine, fluoxetine, paroxetine and sertraline at pharmacologically relevant doses exerted dose- and time-dependent binding activity of brain SERT as revealed by significant increases in KD for specific [3H]paroxetine binding, and the in vivo SERT-binding potency was in the order of paroxetine>>fluoxetine, sertraline>fluvoxamine. Fluvoxamine 355-366 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 169-173 15678084-11 2005 In conclusion, the present study has provided the first in vivo evidences to support that fluvoxamine, fluoxetine, paroxetine and sertraline orally administered bind to the pharmacologically relevant brain SERT in mice and that their SERT-binding characteristics is closely associated with the pharmacokinetics and inhibition of marble-burying behaviour. Fluvoxamine 90-101 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 206-210 15678084-11 2005 In conclusion, the present study has provided the first in vivo evidences to support that fluvoxamine, fluoxetine, paroxetine and sertraline orally administered bind to the pharmacologically relevant brain SERT in mice and that their SERT-binding characteristics is closely associated with the pharmacokinetics and inhibition of marble-burying behaviour. Fluvoxamine 90-101 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 234-238 15458611-3 2004 The aim of this study is to investigate a possible association between this 5-HT1A receptor variant and antidepressant response to fluvoxamine in a sample of 262 mood-disorder subjects (151 major depressed and 111 bipolars) treated with fluvoxamine for 6 wk. Fluvoxamine 131-142 5-hydroxytryptamine receptor 1A Homo sapiens 76-91 15458611-3 2004 The aim of this study is to investigate a possible association between this 5-HT1A receptor variant and antidepressant response to fluvoxamine in a sample of 262 mood-disorder subjects (151 major depressed and 111 bipolars) treated with fluvoxamine for 6 wk. Fluvoxamine 237-248 5-hydroxytryptamine receptor 1A Homo sapiens 76-91 15458611-6 2004 In bipolars, 5-HT1A*C/C genotype carriers showed a better response to fluvoxamine (p=0.036), independently from clinical variables. Fluvoxamine 70-81 5-hydroxytryptamine receptor 1A Homo sapiens 13-19 15545309-2 2004 Administration of a potent CYP1A2 inhibitor (eg, fluvoxamine) may alter the pharmacokinetics of olanzapine. Fluvoxamine 49-60 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 27-33 15301922-0 2004 Chronic treatment with fluvoxamine desensitizes 5-HT2C receptor-mediated hypolocomotion in rats. Fluvoxamine 23-34 5-hydroxytryptamine receptor 2C Rattus norvegicus 48-54 15496639-2 2004 The aim of this study was to compare the inhibitory effects of fluvoxamine, an inhibitor of CYP2C19, on the metabolism of lansoprazole between CYP2C19 genotypes. Fluvoxamine 63-74 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 92-99 15496639-2 2004 The aim of this study was to compare the inhibitory effects of fluvoxamine, an inhibitor of CYP2C19, on the metabolism of lansoprazole between CYP2C19 genotypes. Fluvoxamine 63-74 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 143-150 15496639-7 2004 There was a significant difference in the fluvoxamine-mediated percentage increase in the AUC(0-infinity) of lansoprazole between CYP2C19 genotypes. Fluvoxamine 42-53 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 130-137 15319333-10 2004 Experiments with thiotepa and ketoconazole suggested inhibition of microsomal CYP2B6 and CYP3A4 activity, whereas studies with fluvoxamine (a substrate of CYP2C19) were inconclusive due to lack of specificity. Fluvoxamine 127-138 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 155-162 15301922-3 2004 In the present study, we investigated whether chronic administration of fluvoxamine desensitizes 5-HT2C receptors using a putative in vivo rat model of 5-HT2C receptor function. Fluvoxamine 72-83 5-hydroxytryptamine receptor 2C Rattus norvegicus 97-103 15301922-3 2004 In the present study, we investigated whether chronic administration of fluvoxamine desensitizes 5-HT2C receptors using a putative in vivo rat model of 5-HT2C receptor function. Fluvoxamine 72-83 5-hydroxytryptamine receptor 2C Rattus norvegicus 152-158 15301922-5 2004 This effect of fluvoxamine was reversed by treatment with a selective 5-HT2C receptor antagonist, SB 242084. Fluvoxamine 15-26 5-hydroxytryptamine receptor 2C Rattus norvegicus 70-76 15301922-8 2004 These results suggest that 5-HT2C receptors are desensitized by chronic treatment with fluvoxamine, as well as paroxetine. Fluvoxamine 87-98 5-hydroxytryptamine receptor 2C Rattus norvegicus 27-33 15301922-9 2004 Thus, the clinical efficacy of fluvoxamine on anxiety disorders might involve the normalization of the 5-HT2C receptor function. Fluvoxamine 31-42 5-hydroxytryptamine receptor 2C Rattus norvegicus 103-109 15167270-6 2004 5-HT-induced contraction of normal aorta was potentiated by the 5-HTT inhibitor fluvoxamine. Fluvoxamine 80-91 solute carrier family 6 member 4 Rattus norvegicus 64-69 15251034-5 2004 METHODS: Fatigued patients were randomised to receive fluvoxamine (75 mg BID) or placebo for a six-week period. Fluvoxamine 54-65 BH3 interacting domain death agonist Homo sapiens 73-76 15167270-7 2004 A change in arterial 5-HTT function occurs in deoxycorticosterone (DOCA)-salt hypertension as the potency and threshold of 5-HT in contracting aorta from the DOCA-salt rat was increased by fluoxetine and fluvoxamine (1 micromol/L; DOCA fluvoxamine -log EC50 [mol/L] = 6.85 +/- 0.08, DOCA-control = 6.44 +/- 0.08); expression of transporter was significantly increased in aorta of DOCA salt rats (145% Sham). Fluvoxamine 204-215 solute carrier family 6 member 4 Rattus norvegicus 21-26 15025747-0 2004 Different inhibitory effect of fluvoxamine on omeprazole metabolism between CYP2C19 genotypes. Fluvoxamine 31-42 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 76-83 15170064-6 2004 The author also examined polymorphisms of the serotonin transporter/biosynthetic or metabolizing enzymes in depressive patients treated with fluvoxamine, a selective serotonin reuptake inhibitor, and the relationship between clinical efficacy and polymorphisms was investigated. Fluvoxamine 141-152 solute carrier family 6 member 4 Homo sapiens 46-67 15025747-2 2004 The inhibitory effect of fluvoxamine, an inhibitor of CYP2C19 as well as CYP1A2, on the metabolism of omeprazole was compared between different genotypes for CYP2C19. Fluvoxamine 25-36 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 54-61 15025747-12 2004 These findings confirm a potent inhibitory effect of fluvoxamine on CYP2C19 activity. Fluvoxamine 53-64 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 68-75 14749694-10 2004 The extent of fluvoxamine-lidocaine interaction decreases as liver function worsens, most likely because of the concomitant decrease in the hepatic level of CYP1A2. Fluvoxamine 14-25 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 157-163 15060511-12 2004 Inhibition of tizanidine-metabolizing enzyme(s), mainly CYP1A2, by fluvoxamine seems to explain the observed interaction. Fluvoxamine 67-78 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 56-62 15164615-0 2004 [Postherpetic neuralgia alleviated by an SSRI fluvoxamine: two cases of PHN accompanied with depression were treated with fluvoxamine]. Fluvoxamine 46-57 carbamoyl-phosphate synthase 1 Homo sapiens 72-75 15164615-0 2004 [Postherpetic neuralgia alleviated by an SSRI fluvoxamine: two cases of PHN accompanied with depression were treated with fluvoxamine]. Fluvoxamine 122-133 carbamoyl-phosphate synthase 1 Homo sapiens 72-75 15164615-8 2004 It would be concluded that fluvoxamine alleviates PHN by restoration of the descending serotonergic inhibition of primary afferent activity that carries pain impulses. Fluvoxamine 27-38 carbamoyl-phosphate synthase 1 Homo sapiens 50-53 15148501-1 2004 We investigated the effects of a 5-hydroxytryptamine (5-HT) 1A receptor gene polymorphism on the clinical response to fluvoxamine (FLV) in 65 depressed outpatients who gave written consent to participate in the study. Fluvoxamine 118-129 5-hydroxytryptamine receptor 1A Homo sapiens 33-71 15148501-1 2004 We investigated the effects of a 5-hydroxytryptamine (5-HT) 1A receptor gene polymorphism on the clinical response to fluvoxamine (FLV) in 65 depressed outpatients who gave written consent to participate in the study. Fluvoxamine 131-134 5-hydroxytryptamine receptor 1A Homo sapiens 33-71 12883230-0 2003 Effects of the CYP 2D6 genotype and cigarette smoking on the steady-state plasma concentrations of fluvoxamine and its major metabolite fluvoxamino acid in Japanese depressed patients. Fluvoxamine 99-110 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 15-22 15199661-3 2004 Thus, fluoxetine, fluvoxamine and paroxetine are partially metabolised by CYP2D6, citalopram by CYP2C19 and sertraline by at least five different CYPs. Fluvoxamine 18-29 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 74-80 15199661-3 2004 Thus, fluoxetine, fluvoxamine and paroxetine are partially metabolised by CYP2D6, citalopram by CYP2C19 and sertraline by at least five different CYPs. Fluvoxamine 18-29 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 96-103 14642738-7 2003 Fluvoxamine and diphenytriazol inhibited zolmitriptan N-demethylase activity catalyzed by CYP1A2 (K(i)=3.8+/-0.3 and 3.2+/-0.1 microM, respectively). Fluvoxamine 0-11 cytochrome P450, family 1, subfamily a, polypeptide 2 Rattus norvegicus 90-96 14642738-13 2003 This study aso demonstrated that fluvoxamine may be a mechanism-based inactivator of CYP1A2. Fluvoxamine 33-44 cytochrome P450, family 1, subfamily a, polypeptide 2 Rattus norvegicus 85-91 14501158-4 2003 The compound was as effective as SERT inhibitors such as fluoxetine and fluvoxamine in a 5-hydroxytryptophan-enhancing test in mice. Fluvoxamine 72-83 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 33-37 12721776-0 2003 Blockade of serotonin 5-HT1B and 5-HT2A receptors suppresses the induction of locomotor activity by 5-HT reuptake inhibitors, citalopram and fluvoxamine, in NMRI mice exposed to a novel environment: a comparison to other 5-HT receptor subtypes. Fluvoxamine 141-152 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 22-28 15199661-5 2004 Fluvoxamine is a potent inhibitor of CYP1A2 and CYP2C19 and a moderate inhibitor of CYP2C9. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-43 15199661-5 2004 Fluvoxamine is a potent inhibitor of CYP1A2 and CYP2C19 and a moderate inhibitor of CYP2C9. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 48-55 15199661-5 2004 Fluvoxamine is a potent inhibitor of CYP1A2 and CYP2C19 and a moderate inhibitor of CYP2C9. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 14703714-4 2003 Fluoxetine and paroxetine are potent inhibitors of CYP2D6, fluvoxamine markedly inhibits CYP1A2 and CYP2C19, while nefazodone is a potent inhibitor of CYP3A4. Fluvoxamine 59-70 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 51-57 14703714-4 2003 Fluoxetine and paroxetine are potent inhibitors of CYP2D6, fluvoxamine markedly inhibits CYP1A2 and CYP2C19, while nefazodone is a potent inhibitor of CYP3A4. Fluvoxamine 59-70 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 89-95 14703714-4 2003 Fluoxetine and paroxetine are potent inhibitors of CYP2D6, fluvoxamine markedly inhibits CYP1A2 and CYP2C19, while nefazodone is a potent inhibitor of CYP3A4. Fluvoxamine 59-70 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 100-107 12969751-2 2003 Local administration by reversed microdialysis of the selective 5-HT reuptake inhibitor, fluvoxamine (0.1-10 microM), concentration dependently increased 5-HT to the same extent in wildtype and in 5-HT1B knockout (KO) mice. Fluvoxamine 89-100 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 197-203 12644890-7 2003 In the presence of fluvoxamine, the increased 5-HT release evoked by KCl depolarization was augmented by NAS-181, supporting the idea that blockade of 5-HT transporters is necessary to measure an effect of 5-HT(1B) receptor blockade. Fluvoxamine 19-30 5-hydroxytryptamine receptor 1B Rattus norvegicus 206-213 12755054-2 2003 To create better treatment, we studied here clinical adverse effects of fluvoxamine and correlated them with genetic polymorphism of two genes, the promoter region of serotonin transporter gene (5-HTTLPR) and serotonin 2A receptor gene (5-HT2AR). Fluvoxamine 72-83 solute carrier family 6 member 4 Homo sapiens 167-188 12755054-2 2003 To create better treatment, we studied here clinical adverse effects of fluvoxamine and correlated them with genetic polymorphism of two genes, the promoter region of serotonin transporter gene (5-HTTLPR) and serotonin 2A receptor gene (5-HT2AR). Fluvoxamine 72-83 solute carrier family 6 member 4 Homo sapiens 195-203 12755054-2 2003 To create better treatment, we studied here clinical adverse effects of fluvoxamine and correlated them with genetic polymorphism of two genes, the promoter region of serotonin transporter gene (5-HTTLPR) and serotonin 2A receptor gene (5-HT2AR). Fluvoxamine 72-83 5-hydroxytryptamine receptor 2A Homo sapiens 209-230 12839860-0 2003 Drug binding to HERG channels: evidence for a "non-aromatic" binding site for fluvoxamine. Fluvoxamine 78-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 12839862-0 2003 Blockade of HERG potassium currents by fluvoxamine: incomplete attenuation by S6 mutations at F656 or Y652. Fluvoxamine 39-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 12839862-3 2003 The objectives of this study were (i) to identify and characterise any inhibitory action on HERG of the selective-serotonin re-uptake inhibitor fluvoxamine, (ii) to then determine whether fluvoxamine shared the consensus molecular determinants of HERG blockade of those drugs so far tested. Fluvoxamine 144-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 12839862-6 2003 I(HERG) tails, following repolarisation from +20 to -40 mV, were blocked by fluvoxamine with an IC(50) of 3.8 micro M. 3. Fluvoxamine 76-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 2-6 12839862-7 2003 Blockade of wild-type HERG was of extremely rapid onset (within 10 ms) and showed voltage dependence, with fluvoxamine also inducing a leftward shift in voltage-dependent activation of I(HERG). Fluvoxamine 107-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 12839862-7 2003 Blockade of wild-type HERG was of extremely rapid onset (within 10 ms) and showed voltage dependence, with fluvoxamine also inducing a leftward shift in voltage-dependent activation of I(HERG). Fluvoxamine 107-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 187-191 12839862-11 2003 The S6 mutations, Y652A and F656A, and the pore helix mutant S631A only partially attenuated blockade by fluvoxamine at concentrations causing profound blockade of wild-type HERG. Fluvoxamine 105-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 12828569-10 2003 Inclusion of the CYP1A2 inhibitor fluvoxamine in the incubation mixture with human liver microsomes resulted in potent inhibition of tangeretin and genistein metabolism. Fluvoxamine 34-45 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 17-23 12695344-0 2003 Comparison of in vitro and in vivo inhibition potencies of fluvoxamine toward CYP2C19. Fluvoxamine 59-70 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 78-85 12695344-1 2003 A previous study suggested that fluvoxamine inhibition potency toward CYP1A2 is 10 times greater in vivo than in vitro. Fluvoxamine 32-43 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 70-76 12695344-2 2003 The present study was designed to determine whether the same gap exists for CYP2C19, another isozyme inhibited by fluvoxamine. Fluvoxamine 114-125 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 76-83 12695344-3 2003 In vitro studies examined the effect of nonspecific binding on the determination of inhibition constant (K(i)) values of fluvoxamine toward CYP2C19 in human liver microsomes and in a cDNA-expressed microsomal (Supersomes) system using (S)-mephenytoin as a CYP2C19 probe. Fluvoxamine 121-132 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 140-147 12695344-3 2003 In vitro studies examined the effect of nonspecific binding on the determination of inhibition constant (K(i)) values of fluvoxamine toward CYP2C19 in human liver microsomes and in a cDNA-expressed microsomal (Supersomes) system using (S)-mephenytoin as a CYP2C19 probe. Fluvoxamine 121-132 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 256-263 12618595-4 2003 The production of 5-HT from 5-MT catalyzed by CYP2D6 was inhibited by selective serotonin reuptake inhibitors, and their inhibition potency (K(i), micromol/l) decreased in the order of fluoxetine (0.411) > norfluoxetine (1.38) > fluvoxamine (10.1) > citalopram (10.9). Fluvoxamine 235-246 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 12649369-6 2003 In L-MDR1 cells fluoxetine, norfluoxetine, fluvoxamine, reboxetine, and paroxetine-M revealed intermediate Pgp inhibition and citalopram, desmethylcitalopram, venlafaxine, and N-desmethylvenlafaxine were only weak inhibitors. Fluvoxamine 43-54 ATP binding cassette subfamily B member 1 Homo sapiens 107-110 12695316-0 2003 High levels of serotonin transporter occupancy with low-dose clomipramine in comparative occupancy study with fluvoxamine using positron emission tomography. Fluvoxamine 110-121 solute carrier family 6 member 4 Homo sapiens 15-36 12695316-14 2003 CONCLUSIONS: Clinical doses of clomipramine and fluvoxamine occupied approximately 80% of 5-HTT, and dose escalation would have minimal effect on 5-HTT blockade. Fluvoxamine 48-59 solute carrier family 6 member 4 Homo sapiens 90-95 12640215-8 2003 These results are consistent with two previously published observations, which indicate that the initial affinity of the 5-HTT predicted response to fluvoxamine or fluoxetine in the same way. Fluvoxamine 149-160 solute carrier family 6 member 4 Homo sapiens 121-126 12886034-2 2003 The authors first investigated whether a functional polymorphism in the monoamine oxidase A (MAOA-VNTR) and a -1438G/A polymorphism in the promoter region of the 5-HT(2A) gene were associated with the incidence of nausea induced by fluvoxamine. Fluvoxamine 232-243 5-hydroxytryptamine receptor 2A Homo sapiens 162-169 12610741-1 2003 OBJECTIVE: The purpose of the present study was to investigate whether plasma fluvoxamine (FV) concentration is associated with CYP2D6*10 allele polymorphisms. Fluvoxamine 78-89 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 128-134 12496948-4 2003 Both 6- and 12-week fluvoxamine treatments were associated with a significant and robust reduction of the adrenocorticotrophic hormone (ACTH) and cortisol response to the DEX/CRH test. Fluvoxamine 20-31 corticotropin releasing hormone Homo sapiens 175-178 12480170-0 2003 Fluvoxamine suppresses the long-term potentiation in the hippocampal CA1 field of anesthetized rats: an effect mediated via 5-HT1A receptors. Fluvoxamine 0-11 carbonic anhydrase 1 Rattus norvegicus 69-72 12480170-0 2003 Fluvoxamine suppresses the long-term potentiation in the hippocampal CA1 field of anesthetized rats: an effect mediated via 5-HT1A receptors. Fluvoxamine 0-11 5-hydroxytryptamine receptor 1A Rattus norvegicus 124-130 12856820-2 2003 It was decided to evaluate the influence of imipramine (IMI), amitriptyline (AMI), fluvoxamine (FLU), mianserin (MIA) and tianeptine (TIA) on PLA2 activity after an acute and long-term (4 weeks) drug administration. Fluvoxamine 83-94 phospholipase A2 group IB Rattus norvegicus 142-146 12607287-0 2003 [Serotonin 2A receptor gene polymorphism and clinical efficacy of fluvoxamine in children with autistic disorder]. Fluvoxamine 66-77 5-hydroxytryptamine receptor 2A Homo sapiens 1-22 12856820-8 2003 It seems that FLU was the only antidepressant, which induced either inhibition or activation of PLA2 depending on time of administration. Fluvoxamine 14-17 phospholipase A2 group IB Rattus norvegicus 96-100 12856820-2 2003 It was decided to evaluate the influence of imipramine (IMI), amitriptyline (AMI), fluvoxamine (FLU), mianserin (MIA) and tianeptine (TIA) on PLA2 activity after an acute and long-term (4 weeks) drug administration. Fluvoxamine 96-99 phospholipase A2 group IB Rattus norvegicus 142-146 12452751-8 2002 Fluvoxamine has the potential to inhibit CYP1A2, CYP2C9, CYP2C19, and CYP3A4 to a significant degree. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 41-47 12452751-8 2002 Fluvoxamine has the potential to inhibit CYP1A2, CYP2C9, CYP2C19, and CYP3A4 to a significant degree. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 12452751-8 2002 Fluvoxamine has the potential to inhibit CYP1A2, CYP2C9, CYP2C19, and CYP3A4 to a significant degree. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 57-64 12452751-8 2002 Fluvoxamine has the potential to inhibit CYP1A2, CYP2C9, CYP2C19, and CYP3A4 to a significant degree. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 70-76 12502014-5 2002 The present study fails to demonstrate that the genetic polymorphisms of MAOA-VNTR and TPH-A218C affect the antidepressant effect of fluvoxamine in Japanese patients with major depressive disorder. Fluvoxamine 133-144 monoamine oxidase A Homo sapiens 73-77 12444499-10 2002 Thus, fluvoxamine-induced facilitatory effects appear to be mediated via 5-HT(1A) and 5-HT(4)/5-HT(7) receptors in an inhibitory and a stimulatory manner, respectively. Fluvoxamine 6-17 5-hydroxytryptamine receptor 1A Rattus norvegicus 73-80 12444499-11 2002 In the CA3 field, excitability produced by fluvoxamine was abolished by either NAN 190 or DR 4004, but not by GR 113808, suggesting that 5-HT(1A) and 5-HT(7) receptors contribute to this facilitation. Fluvoxamine 43-54 carbonic anhydrase 3 Rattus norvegicus 7-10 12444499-11 2002 In the CA3 field, excitability produced by fluvoxamine was abolished by either NAN 190 or DR 4004, but not by GR 113808, suggesting that 5-HT(1A) and 5-HT(7) receptors contribute to this facilitation. Fluvoxamine 43-54 5-hydroxytryptamine receptor 1A Rattus norvegicus 137-144 12523495-8 2002 The obtained results suggest a strong inhibitory effect of perazine on human CYP1A2 activity with predicted Ki value similar to those of the known for CYP1A2 inhibitors, such as furafylline and fluvoxamine. Fluvoxamine 194-205 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 77-83 12419542-5 2002 The selective serotonin reuptake inhibitor, fluvoxamine, prolonged 5-HT clearance in both CA3 and DRN. Fluvoxamine 44-55 carbonic anhydrase 3 Rattus norvegicus 90-93 12352274-2 2002 In this study, pharmacokinetic interactions and clinical effects of adding the CYP1A2 inhibitor fluvoxamine to steady-state olanzapine was examined in patients suffering from schizophrenia. Fluvoxamine 96-107 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 79-85 12523495-8 2002 The obtained results suggest a strong inhibitory effect of perazine on human CYP1A2 activity with predicted Ki value similar to those of the known for CYP1A2 inhibitors, such as furafylline and fluvoxamine. Fluvoxamine 194-205 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 151-157 11985826-5 2002 In conclusion, like fluvoxamine and clomipramine, selective, non-peptidergic NK(1) receptor antagonists block marble-burying in mice. Fluvoxamine 20-31 tachykinin receptor 1 Mus musculus 77-91 11955666-3 2002 Microsomal flavonoid metabolism was potently inhibited by the CYP1A2 inhibitors, fluvoxamine and -naphthoflavone. Fluvoxamine 81-92 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 62-68 11907488-0 2002 Low daily 10-mg and 20-mg doses of fluvoxamine inhibit the metabolism of both caffeine (cytochrome P4501A2) and omeprazole (cytochrome P4502C19). Fluvoxamine 35-46 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 88-106 11937097-10 2002 Local administration of 0.3 microM fluvoxamine resulted in comparable increases in extracellular 5-HT in both genotypes, whereas 1.0 microM fluvoxamine produced a twofold greater increase in 5-HT levels in 5-HT(1B) knockout as compared to wildtype mice. Fluvoxamine 140-151 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 206-213 11937097-12 2002 In addition, the different dose-response to fluvoxamine suggests that 5-HT(1B) knockout mice have possible adaptations of 5-HT transporters in order to compensate for the loss of the terminal 5-HT(1B) autoreceptor. Fluvoxamine 44-55 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 70-77 11907488-11 2002 At steady state, the 25 mg x 1 or x 2 fluvoxamine dose caused a pronounced inhibition of about 75% to 80% for both CYP1A2 and CYP2C19, whereas the inhibition after the lower 10 mg x 1 or x 2 dose was about 40% to 50%. Fluvoxamine 38-49 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 115-121 11907488-1 2002 OBJECTIVES: Fluvoxamine is metabolized by the polymorphic cytochrome P450 (CYP) 2D6 and the smoking-inducible CYP1A2. Fluvoxamine 12-23 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 58-83 11907488-11 2002 At steady state, the 25 mg x 1 or x 2 fluvoxamine dose caused a pronounced inhibition of about 75% to 80% for both CYP1A2 and CYP2C19, whereas the inhibition after the lower 10 mg x 1 or x 2 dose was about 40% to 50%. Fluvoxamine 38-49 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 126-133 11907488-1 2002 OBJECTIVES: Fluvoxamine is metabolized by the polymorphic cytochrome P450 (CYP) 2D6 and the smoking-inducible CYP1A2. Fluvoxamine 12-23 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 110-116 11907488-18 2002 A nontherapeutic oral daily dose of fluvoxamine is sufficient to provide a marked inhibition of both caffeine (CYP1A2) and omeprazole (CYP2C19) metabolism. Fluvoxamine 36-47 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 111-117 11907488-18 2002 A nontherapeutic oral daily dose of fluvoxamine is sufficient to provide a marked inhibition of both caffeine (CYP1A2) and omeprazole (CYP2C19) metabolism. Fluvoxamine 36-47 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 135-142 11907488-2 2002 Therapeutic doses of fluvoxamine inhibit both CYP1A2 and CYP2C19. Fluvoxamine 21-32 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 46-52 11907488-2 2002 Therapeutic doses of fluvoxamine inhibit both CYP1A2 and CYP2C19. Fluvoxamine 21-32 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 57-64 11803239-6 2002 These findings are compatible with those in earlier reports that cytochrome P450 1A2 plays a major role in fluvoxamine metabolism. Fluvoxamine 107-118 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 65-84 12057029-1 2002 The aim of the present study was to test a possible effect of the monoamine oxidase A (MAOA) and serotonin receptor 2A (5-HT-2A) gene variants on the antidepressant activity of fluvoxamine and paroxetine in a sample of major (n = 248) and bipolar (n = 195) depressives, with or without psychotic features. Fluvoxamine 177-188 monoamine oxidase A Homo sapiens 66-85 12057029-1 2002 The aim of the present study was to test a possible effect of the monoamine oxidase A (MAOA) and serotonin receptor 2A (5-HT-2A) gene variants on the antidepressant activity of fluvoxamine and paroxetine in a sample of major (n = 248) and bipolar (n = 195) depressives, with or without psychotic features. Fluvoxamine 177-188 monoamine oxidase A Homo sapiens 87-91 12057029-1 2002 The aim of the present study was to test a possible effect of the monoamine oxidase A (MAOA) and serotonin receptor 2A (5-HT-2A) gene variants on the antidepressant activity of fluvoxamine and paroxetine in a sample of major (n = 248) and bipolar (n = 195) depressives, with or without psychotic features. Fluvoxamine 177-188 5-hydroxytryptamine receptor 2A Homo sapiens 120-127 11817517-0 2002 Influence of the serotonin transporter gene-linked polymorphic region on the antidepressant response to fluvoxamine in Japanese depressed patients. Fluvoxamine 104-115 solute carrier family 6 member 4 Homo sapiens 17-38 12222750-0 2002 Pharmacokinetics of fluvoxamine in relation to CYP2C19 phenotype and genotype. Fluvoxamine 20-31 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 47-54 12222750-1 2002 OBJECTIVE: To evaluate the pharmacokinetics of fluvoxamine (FLV) in poor metabolizers (PMs) versus extensive metabolizers (EMs) of cytochrome P450 (CYP)2C19. Fluvoxamine 47-58 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 131-156 12222750-1 2002 OBJECTIVE: To evaluate the pharmacokinetics of fluvoxamine (FLV) in poor metabolizers (PMs) versus extensive metabolizers (EMs) of cytochrome P450 (CYP)2C19. Fluvoxamine 60-63 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 131-156 11876575-6 2002 Fluvoxamine is a potent CYP1A2 and CYP2C19 inhibitor, and a moderate CYP2C9, CYP2D6, and CYP3A4 inhibitor. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 24-30 11876575-6 2002 Fluvoxamine is a potent CYP1A2 and CYP2C19 inhibitor, and a moderate CYP2C9, CYP2D6, and CYP3A4 inhibitor. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 35-42 11876575-6 2002 Fluvoxamine is a potent CYP1A2 and CYP2C19 inhibitor, and a moderate CYP2C9, CYP2D6, and CYP3A4 inhibitor. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 11876575-6 2002 Fluvoxamine is a potent CYP1A2 and CYP2C19 inhibitor, and a moderate CYP2C9, CYP2D6, and CYP3A4 inhibitor. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 77-83 11876575-6 2002 Fluvoxamine is a potent CYP1A2 and CYP2C19 inhibitor, and a moderate CYP2C9, CYP2D6, and CYP3A4 inhibitor. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 89-95 12038880-11 2002 Within the group of SSRIs, fluoxetine and paroxetine are potent inhibitors of CYP2D6, while fluvoxamine predominantly affects CYP1A2 and CYP2C19 activity. Fluvoxamine 92-103 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 126-132 12038880-11 2002 Within the group of SSRIs, fluoxetine and paroxetine are potent inhibitors of CYP2D6, while fluvoxamine predominantly affects CYP1A2 and CYP2C19 activity. Fluvoxamine 92-103 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 137-144 12422060-0 2002 Association between -1438G/A promoter polymorphism in the 5-HT(2A) receptor gene and fluvoxamine response in Japanese patients with major depressive disorder. Fluvoxamine 85-96 5-hydroxytryptamine receptor 2A Homo sapiens 58-75 11728615-3 2001 Plasma 5-hydroxyindoleacetic acid (p5-HIAA) levels after fluvoxamine administration were significantly higher in patients with nausea (6.6+/-3.4 ng/ml) than in those without nausea (3.5+/-2.7 ng/ml). Fluvoxamine 57-68 solute carrier family 10 member 5 Homo sapiens 35-42 11673783-0 2001 The effects of the selective serotonin reuptake-inhibitor fluvoxamine on body weight in Zucker rats are mediated by corticotropin-releasing hormone. Fluvoxamine 58-69 corticotropin releasing hormone Rattus norvegicus 116-147 11719727-1 2001 OBJECTIVE: Several reports indicate that fluvoxamine decreases the clearance of cytochrome P4501A2 (CYP1A2) substrates. Fluvoxamine 41-52 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 80-98 11719727-1 2001 OBJECTIVE: Several reports indicate that fluvoxamine decreases the clearance of cytochrome P4501A2 (CYP1A2) substrates. Fluvoxamine 41-52 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 100-106 11719727-2 2001 This study compared in vitro and in vivo inhibition potencies of fluvoxamine toward CYP1A2 with an approach based on inhibition constants (K(i)) determined in vitro and in vivo. Fluvoxamine 65-76 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 84-90 11719727-4 2001 Fluvoxamine in vivo inhibition constants (K(i)iv) for CYP1A2 were obtained from an investigation of single-dose theophylline (250 mg) disposition in 9 healthy volunteers receiving steady-state (9 days) fluvoxamine at 3 doses (0, 25, or 75 mg/d) in a randomized crossover design. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 54-60 11704898-7 2001 Inhibition of CYP1A2 by fluvoxamine yields increased concentrations; however, clinically relevant CYP2D6 inhibition was observed only in combination with additional disposition factors, such as female gender or old age. Fluvoxamine 24-35 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 14-20 11791895-0 2001 The major fluvoxamine metabolite in urine is formed by CYP2D6. Fluvoxamine 10-21 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 55-61 11791895-1 2001 OBJECTIVE: Previous studies have shown that fluvoxamine is metabolized by CYP1A2 and CYP2D6, but there is no information on the impact the various CYP enzymes have on the different metabolic pathways of fluvoxamine biotransformation. Fluvoxamine 44-55 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 74-80 11791895-1 2001 OBJECTIVE: Previous studies have shown that fluvoxamine is metabolized by CYP1A2 and CYP2D6, but there is no information on the impact the various CYP enzymes have on the different metabolic pathways of fluvoxamine biotransformation. Fluvoxamine 44-55 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 85-91 11791895-3 2001 METHODS: The major fluvoxamine metabolite, the 5-demethoxylated carboxylic acid metabolite, was analyzed in urine from 50 healthy volunteers after intake of a single oral dose of 50 mg fluvoxamine, and the formation clearance for the metabolite (CLm) was calculated. Fluvoxamine 19-30 laminin subunit beta 1 Homo sapiens 246-249 11791895-7 2001 RESULTS: Oral clearance of fluvoxamine was significantly higher in smokers, and significantly lower in CYP2D6 PMs than in non-smoking EMs. Fluvoxamine 27-38 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 103-109 11791895-11 2001 CLm decreased with increasing fluvoxamine dosage, but the decrease in oral clearance was even higher. Fluvoxamine 30-41 laminin subunit beta 1 Homo sapiens 0-3 11791895-12 2001 CONCLUSION: These results indicate that CYP2D6 catalyzes the major metabolic pathway of fluvoxamine, whereas CYP1A2 seems to catalyze other less important pathways. Fluvoxamine 88-99 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 40-46 11791895-12 2001 CONCLUSION: These results indicate that CYP2D6 catalyzes the major metabolic pathway of fluvoxamine, whereas CYP1A2 seems to catalyze other less important pathways. Fluvoxamine 88-99 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 109-115 11791895-13 2001 Both the CYP2D6 and the CYP1A2 pathways seem to be saturated in parallel with increasing fluvoxamine dosage. Fluvoxamine 89-100 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 9-15 11791895-13 2001 Both the CYP2D6 and the CYP1A2 pathways seem to be saturated in parallel with increasing fluvoxamine dosage. Fluvoxamine 89-100 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 24-30 11434513-5 2001 The first six phenotyping measures were used to estimate baseline CYP3A activity, then subjects received the moderate CYP3A inhibitor fluvoxamine 150 mg/day for the last 4 weeks (two phenotyping visits) of the study. Fluvoxamine 134-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 118-123 11599655-0 2001 Detailed characterization of experimentally derived human hepatic CYP1A1 activity and expression using differential inhibition of ethoxyresorufin O-deethylation by fluvoxamine. Fluvoxamine 164-175 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 66-72 11599655-1 2001 OBJECTIVE: To characterize the distribution of mathematically derived human hepatic CYP1A1 activity using differential inhibition of ethoxyresorufin O-deethylation (EROD) by fluvoxamine. Fluvoxamine 174-185 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 84-90 11599655-2 2001 METHODS: Quantitative CYP1A1- and CYP1A2-mediated EROD activities were determined in 42 human livers using differential inhibition of EROD by fluvoxamine. Fluvoxamine 142-153 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 22-28 11599655-2 2001 METHODS: Quantitative CYP1A1- and CYP1A2-mediated EROD activities were determined in 42 human livers using differential inhibition of EROD by fluvoxamine. Fluvoxamine 142-153 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 34-40 11477325-5 2001 This indicated that fluvoxamine inhibits the metabolism of olanzapine, probably because of inhibition of cytochrome P450 (CYP) 1A2, whereas sertraline is unlikely to interfere with the metabolism of olanzapine. Fluvoxamine 20-31 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 105-130 11526473-0 2001 Influence of tryptophan hydroxylase and serotonin transporter genes on fluvoxamine antidepressant activity. Fluvoxamine 71-82 solute carrier family 6 member 4 Homo sapiens 40-61 11519155-5 2001 Fluvoxamine, an SSRI, is a potent inhibitors for CYP1A2 and CYP2C19, moderate for CYP3A4 and weak for CYP 2D6. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 49-55 11519155-5 2001 Fluvoxamine, an SSRI, is a potent inhibitors for CYP1A2 and CYP2C19, moderate for CYP3A4 and weak for CYP 2D6. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 60-67 11519155-5 2001 Fluvoxamine, an SSRI, is a potent inhibitors for CYP1A2 and CYP2C19, moderate for CYP3A4 and weak for CYP 2D6. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 11519155-5 2001 Fluvoxamine, an SSRI, is a potent inhibitors for CYP1A2 and CYP2C19, moderate for CYP3A4 and weak for CYP 2D6. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 102-109 11270913-3 2001 Because fluvoxamine is an inhibitor of several cytochrome P450 (CYP) enzymes, the authors studied the biotransformation of melatonin and the effects of fluvoxamine on the metabolism of melatonin in vitro using human liver microsomes and recombinant human CYP isoenzymes. Fluvoxamine 8-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 11304754-0 2001 Effects of aging on serotonin transporter availability and its response to fluvoxamine in the living brain: PET study with [(11)C](+)McN5652 and [(11)C](-)McN5652 in conscious monkeys. Fluvoxamine 75-86 solute carrier family 6 member 4 Homo sapiens 20-41 11304754-5 2001 When the SERT blocker fluvoxamine (1 mg/kg) was administered intravenously 30 min after tracer injection, specific binding of SERT was displaced in both age groups. Fluvoxamine 22-33 solute carrier family 6 member 4 Homo sapiens 9-13 11304754-5 2001 When the SERT blocker fluvoxamine (1 mg/kg) was administered intravenously 30 min after tracer injection, specific binding of SERT was displaced in both age groups. Fluvoxamine 22-33 solute carrier family 6 member 4 Homo sapiens 126-130 11304754-9 2001 In addition, these observations suggest that the age-related impairment of SERT sensitivity for fluvoxamine might be related to the reduced efficacy of antidepressant therapy in elderly patients with depression. Fluvoxamine 96-107 solute carrier family 6 member 4 Homo sapiens 75-79 11270913-3 2001 Because fluvoxamine is an inhibitor of several cytochrome P450 (CYP) enzymes, the authors studied the biotransformation of melatonin and the effects of fluvoxamine on the metabolism of melatonin in vitro using human liver microsomes and recombinant human CYP isoenzymes. Fluvoxamine 8-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 11317475-7 2001 CONCLUSIONS: Our results strongly suggest that 6-hydroxylation, the main metabolic pathway of melatonin, is mediated mainly, but not exclusively, by CYP1A2, the high-affinity enzyme involved in melatonin metabolism, confirming the observation that a single oral dose of fluvoxamine increases nocturnal serum melatonin levels in healthy subjects. Fluvoxamine 270-281 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 149-155 11240973-1 2001 BACKGROUND AND OBJECTIVES: Fluvoxamine, a selective serotonin reuptake inhibitor, is known to inhibit several hepatic cytochrome P450 (CYP) isozymes, in particular CYP1A2. Fluvoxamine 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 11240973-1 2001 BACKGROUND AND OBJECTIVES: Fluvoxamine, a selective serotonin reuptake inhibitor, is known to inhibit several hepatic cytochrome P450 (CYP) isozymes, in particular CYP1A2. Fluvoxamine 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 11240973-1 2001 BACKGROUND AND OBJECTIVES: Fluvoxamine, a selective serotonin reuptake inhibitor, is known to inhibit several hepatic cytochrome P450 (CYP) isozymes, in particular CYP1A2. Fluvoxamine 27-38 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 164-170 11166521-5 2001 The hyperactivity elicited by fluvoxamine (20 mg/kg) plus mazindol (1 mg/kg) was significantly attenuated by the 5-HT(2A) receptor antagonist M100907 (2 mg/kg) and potentiated by the 5-HT(2B/2C) receptor antagonist SB 206553 (2 mg/kg). Fluvoxamine 30-41 5-hydroxytryptamine receptor 2A Rattus norvegicus 113-120 11166521-7 2001 The hyperactivity evoked by the combination of fluvoxamine and mazindol seems to be mediated in part by 5-HT(2A) receptors; whereas, 5-HT(2B/2C) receptors may serve to limit this effect. Fluvoxamine 47-58 5-hydroxytryptamine receptor 2A Rattus norvegicus 104-111 11206048-5 2001 The interaction may be a result of inhibition of both CYP2C9 and 2C19 by fluvoxamine. Fluvoxamine 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 11038165-8 2000 Fluvoxamine and anti-CYP2C antibody inhibited 3",4"-diHPPH formation from 10 microM 4"-HPPH in a human liver sample that contained relatively high levels of CYP2C, whereas ketoconazole and anti-CYP3A antibody showed inhibitory effects on the activities in liver microsomal samples in which CYP3A4 levels were relatively high. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 290-296 11791889-12 2001 These results indicate that CYP 3A4 inhibition may not be clinically significant compared to CYP 1A2, as previous studies show a dramatic increase in CLZ plasma concentrations with fluvoxamine (CYP 1A2 inhibitor). Fluvoxamine 181-192 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 194-201 11038165-8 2000 Fluvoxamine and anti-CYP2C antibody inhibited 3",4"-diHPPH formation from 10 microM 4"-HPPH in a human liver sample that contained relatively high levels of CYP2C, whereas ketoconazole and anti-CYP3A antibody showed inhibitory effects on the activities in liver microsomal samples in which CYP3A4 levels were relatively high. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 157-162 11180037-0 2001 Fluvoxamine inhibits the CYP2C9 catalyzed biotransformation of tolbutamide. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11180037-1 2001 OBJECTIVE: Our objective was to examine the interaction between fluvoxamine and tolbutamide to confirm that fluvoxamine inhibits CYP2C9. Fluvoxamine 108-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 11180037-13 2001 CONCLUSION: Fluvoxamine is a moderate inhibitor of CYP2C9 in vivo. Fluvoxamine 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 24921690-9 2001 Potent inhibitors of CYP 1A2 such as fluvoxamine can precipitate caffeine toxicity. Fluvoxamine 37-48 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 21-28 11074049-6 2000 Golgi analysis showed that a single injection of fluvoxamine produced a significant increase in dendritic spine density in stratum radiatum of CA1 and in the dentate gyrus. Fluvoxamine 49-60 carbonic anhydrase 1 Rattus norvegicus 143-146 11038165-8 2000 Fluvoxamine and anti-CYP2C antibody inhibited 3",4"-diHPPH formation from 10 microM 4"-HPPH in a human liver sample that contained relatively high levels of CYP2C, whereas ketoconazole and anti-CYP3A antibody showed inhibitory effects on the activities in liver microsomal samples in which CYP3A4 levels were relatively high. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 194-199 11098879-5 2000 One controlled study of the selective SRI fluvoxamine found it to be significantly better than placebo for reducing repetitive phenomena and aggression in adults with autistic disorder. Fluvoxamine 42-53 sorcin Homo sapiens 38-41 11085201-3 2000 Relative to other SSRIs, fluvoxamine is a weak inhibitor of cytochrome P450 (CYP) 2D6, a moderate inhibitor of CYP2C19 and CYP3A4 and a potent inhibitor of CYP1A2. Fluvoxamine 25-36 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 60-85 11085201-3 2000 Relative to other SSRIs, fluvoxamine is a weak inhibitor of cytochrome P450 (CYP) 2D6, a moderate inhibitor of CYP2C19 and CYP3A4 and a potent inhibitor of CYP1A2. Fluvoxamine 25-36 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 111-118 11085201-3 2000 Relative to other SSRIs, fluvoxamine is a weak inhibitor of cytochrome P450 (CYP) 2D6, a moderate inhibitor of CYP2C19 and CYP3A4 and a potent inhibitor of CYP1A2. Fluvoxamine 25-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 123-129 11085201-3 2000 Relative to other SSRIs, fluvoxamine is a weak inhibitor of cytochrome P450 (CYP) 2D6, a moderate inhibitor of CYP2C19 and CYP3A4 and a potent inhibitor of CYP1A2. Fluvoxamine 25-36 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 156-162 11062862-9 2000 This indicates that (+/-)-pindolol has the ability to block the somatodendritic 5-HT1A autoreceptors, thereby weakening the fluvoxamine-induced indirect action of the autoreceptors in the raphe. Fluvoxamine 124-135 5-hydroxytryptamine receptor 1A Rattus norvegicus 80-86 10917404-16 2000 An interaction with fluvoxamine may be of importance in poor metabolizers for CYP2D6. Fluvoxamine 20-31 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 78-84 10982203-2 2000 Previous reports have shown that fluvoxamine can increase plasma clozapine concentrations by inhibition of cytochrome P450 (CYP) 1A2. Fluvoxamine 33-44 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 107-132 10954064-1 2000 The cytochrome enzyme P450 2D6 (CYP2D6) is thought to play a role in the human metabolism of fluvoxamine. Fluvoxamine 93-104 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 4-30 10954064-1 2000 The cytochrome enzyme P450 2D6 (CYP2D6) is thought to play a role in the human metabolism of fluvoxamine. Fluvoxamine 93-104 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-38 10877005-0 2000 Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 10877005-0 2000 Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 114-120 10877005-0 2000 Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 125-132 10877005-6 2000 FLU inhibits CYP1A2 potently, and to some extent also CYP2C19, whereas CIT is without such an effect. Fluvoxamine 0-3 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 13-19 10877005-6 2000 FLU inhibits CYP1A2 potently, and to some extent also CYP2C19, whereas CIT is without such an effect. Fluvoxamine 0-3 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 54-61 10805611-4 2000 RESULTS: On the basis of comparable plasma levels of clozapine and N-desmethylclozapine, the coadministration of fluvoxamine 1) attenuated and delayed the clozapine-induced increase in TNF-alpha plasma levels, 2) enhanced and accelerated the clozapine-induced increase in leptin plasma levels without significant effect on clozapine-induced weight gain, and 3) decreased granulocyte counts. Fluvoxamine 113-124 tumor necrosis factor Homo sapiens 185-194 10706994-1 2000 In the course of investigating the mechanisms underlying the beneficial effect of fluvoxamine augmentation on negative symptoms of schizophrenia, the authors found a reduction in human platelet monoamine oxidase-B activity after 5 weeks of treatment. Fluvoxamine 82-93 monoamine oxidase B Homo sapiens 194-213 10774624-6 2000 Fluvoxamine is metabolized to inactive metabolites by CYP1A2 and CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 54-60 10774624-6 2000 Fluvoxamine is metabolized to inactive metabolites by CYP1A2 and CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 65-71 10706994-3 2000 The current study examined the effect of long-term administration, up to 6 weeks, of fluvoxamine, desipramine or saline on MAO-A and MAO-B activities in rat striatum, frontal cortex and liver. Fluvoxamine 85-96 monoamine oxidase A Rattus norvegicus 123-128 10706994-3 2000 The current study examined the effect of long-term administration, up to 6 weeks, of fluvoxamine, desipramine or saline on MAO-A and MAO-B activities in rat striatum, frontal cortex and liver. Fluvoxamine 85-96 monoamine oxidase B Rattus norvegicus 133-138 10653206-0 2000 Fluvoxamine-Clozapine drug interaction: inhibition in vitro of five cytochrome P450 isoforms involved in clozapine metabolism. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 11249525-5 2000 Differences in the metabolic balance between hepatic P450 (especially CYP 1A2) and MAO-A inactivation lead to potential drug interactions for all TELs with the oral contraceptive pill (OCP), fluvoxamine and the quinilone antibiotics (with increased triptan levels). Fluvoxamine 191-202 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 70-77 11249525-5 2000 Differences in the metabolic balance between hepatic P450 (especially CYP 1A2) and MAO-A inactivation lead to potential drug interactions for all TELs with the oral contraceptive pill (OCP), fluvoxamine and the quinilone antibiotics (with increased triptan levels). Fluvoxamine 191-202 monoamine oxidase A Homo sapiens 83-88 10653206-5 2000 Fluvoxamine also inhibited in a concentration-dependent manner the activity of all five cytochrome P450 (CYP) isoforms previously determined to be capable of catalyzing the demethylation of clozapine. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 10653206-5 2000 Fluvoxamine also inhibited in a concentration-dependent manner the activity of all five cytochrome P450 (CYP) isoforms previously determined to be capable of catalyzing the demethylation of clozapine. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 10653206-6 2000 Fluvoxamine inhibited CYP1A2 and 2C19 with the highest affinities (Ki values of 0.041 and 0.087 microM, respectively). Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 22-28 11015030-8 2000 Chronic treatment with fluvoxamine, desipramine and lithium carbonate is apparently associated with a modulation in PBR expression in the testes, adrenals, kidneys, liver and brain, and in CBR expression in brain. Fluvoxamine 23-34 translocator protein Rattus norvegicus 116-119 10627170-0 1999 CYP2C9 is a principal low-affinity phenacetin O-deethylase: fluvoxamine is not a specific CYP1A2 inhibitor. Fluvoxamine 60-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11015030-3 2000 We investigated the effect of 21 days administration, followed by 7 days withdrawal, of fluvoxamine (10 mg/kg), desipramine (10 mg/kg) and lithium carbonate (25 mg/kg) on PBR and CBR binding characteristics in male Sprague-Dawley rats. Fluvoxamine 88-99 translocator protein Rattus norvegicus 171-174 10674711-9 2000 Paroxetine and, to a lesser degree, fluoxetine and norfluoxetine are potent inhibitors of CYP2D6 and fluvoxamine of CYP1A2 and CYP2C19. Fluvoxamine 101-112 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 116-122 10674711-9 2000 Paroxetine and, to a lesser degree, fluoxetine and norfluoxetine are potent inhibitors of CYP2D6 and fluvoxamine of CYP1A2 and CYP2C19. Fluvoxamine 101-112 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 127-134 10456487-3 1999 The aim of this study was to establish whether the potent CYP1A2 inhibitor fluvoxamine in clinically relevant doses could inhibit tacrine metabolism. Fluvoxamine 75-86 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 58-64 10550852-5 1999 Of the SSRIs, fluvoxamine is one of the most potent inhibitors of the isoenzyme CYP 1A2. Fluvoxamine 14-25 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 80-87 10587283-6 1999 In conclusion, fluvoxamine markedly interferes with the metabolism of THD, probably at the CYP2C19 and/or CYP1A2 enzyme level. Fluvoxamine 15-26 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 91-98 10587283-6 1999 In conclusion, fluvoxamine markedly interferes with the metabolism of THD, probably at the CYP2C19 and/or CYP1A2 enzyme level. Fluvoxamine 15-26 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 106-112 10023505-7 1999 In fact, prolactin response to d-fenfluramine was significantly diminished after treatment with fluvoxamine but not fluoxetine. Fluvoxamine 96-107 prolactin Homo sapiens 9-18 10371024-0 1999 Serum beta-endorphin level in patients with depression on fluvoxamine. Fluvoxamine 58-69 proopiomelanocortin Homo sapiens 6-20 10371024-1 1999 The main interest of the present study was to determine possible alternations in beta-endorphin serum levels in healthy volunteers and in patients with depression, as well as changes in beta-endorphin serum levels caused by fluvoxamine treatment. Fluvoxamine 224-235 proopiomelanocortin Homo sapiens 186-200 10371024-5 1999 A 4-week treatment of fluvoxamine (200 mg/day) caused a statistically significant increase in beta-endorphin serum levels in all patients (nonendogenous depression 132.10 +/- 2.38 pg/ml and endogenous depression 50.09 +/- 2.45 pg/ml) in comparison to values found before the onset of the therapy. Fluvoxamine 22-33 proopiomelanocortin Homo sapiens 94-108 10340631-11 1999 Inhibition of fluvoxamine induced potentiation of 5-HT signal in the presence of MK-801 suggests that MK-801 and fluvoxamine may interact at the level of the 5-HT transporter. Fluvoxamine 14-25 solute carrier family 6 member 4 Homo sapiens 158-174 10340631-11 1999 Inhibition of fluvoxamine induced potentiation of 5-HT signal in the presence of MK-801 suggests that MK-801 and fluvoxamine may interact at the level of the 5-HT transporter. Fluvoxamine 113-124 solute carrier family 6 member 4 Homo sapiens 158-174 10471171-8 1999 Fluvoxamine is a potent inhibitor of CYP1A2, a moderate inhibitor of CYP3A and a mild inhibitor of CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-43 10471171-8 1999 Fluvoxamine is a potent inhibitor of CYP1A2, a moderate inhibitor of CYP3A and a mild inhibitor of CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-74 10471171-8 1999 Fluvoxamine is a potent inhibitor of CYP1A2, a moderate inhibitor of CYP3A and a mild inhibitor of CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 99-105 10192828-15 1999 Fluvoxamine (FX), at a concentration of 20 microM, decreased NDV production by 46% consistent with the capacity of FX to inhibit CYP3A, 2C9, and 2C19. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-139 10192828-15 1999 Fluvoxamine (FX), at a concentration of 20 microM, decreased NDV production by 46% consistent with the capacity of FX to inhibit CYP3A, 2C9, and 2C19. Fluvoxamine 13-15 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 129-139 10211917-0 1999 CYP2D6 status of extensive metabolizers after multiple-dose fluoxetine, fluvoxamine, paroxetine, or sertraline. Fluvoxamine 72-83 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 10051063-7 1999 Serotonin-specific reuptake inhibitor (SSRI) comedication (fluoxetine, two patients; citalopram, two patients; paroxetine, one patient; fluvoxamine, one patient) was significantly associated with 4.6-fold higher concentrations of parent compound, in keeping with an inhibitory action on CYP2D6 enzyme. Fluvoxamine 136-147 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 287-293 10192755-1 1999 OBJECTIVE: Evidence exists to suggest that fluvoxamine is metabolized by CYP1A2. Fluvoxamine 43-54 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 73-79 10192755-2 1999 The present study was undertaken in order to further elucidate the role of CYPIA2 in fluvoxamine disposition. Fluvoxamine 85-96 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 75-81 10192756-10 1999 This is consistent with in vivo data indicating that amongst the SSRIs, fluvoxamine has the greatest potential for inhibiting CYP2C9-mediated drug metabolism. Fluvoxamine 72-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 9871099-3 1998 While binding of [123I]nor-beta-CIT in the hypothalamus was blocked significantly by fluvoxamine (a selective serotonin transporter blocker) but not by GBR12,909 (a selective dopamine transporter blocker), the opposite was observed in the striatum. Fluvoxamine 85-96 citron rho-interacting serine/threonine kinase Rattus norvegicus 32-35 9871099-3 1998 While binding of [123I]nor-beta-CIT in the hypothalamus was blocked significantly by fluvoxamine (a selective serotonin transporter blocker) but not by GBR12,909 (a selective dopamine transporter blocker), the opposite was observed in the striatum. Fluvoxamine 85-96 solute carrier family 6 member 4 Rattus norvegicus 110-131 9888626-4 1998 Pre-exposure with the 5-HT2C receptor agonist MK 212 [6-chloro-2(1-piperazinyl)pyrazine] partially prevented the fluvoxamine-induced CTA, pre-exposure with the 5-HT2A/2C receptor agonist DOI [1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane HCl] did not prevent the CTA induced by fluvoxamine. Fluvoxamine 113-124 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 22-37 9868741-7 1998 The formation of 1,7-dimethylxanthine was virtually abolished by 10 microM of fluvoxamine, indicating that the N3-demethylation of caffeine is almost exclusively catalysed by CYP1A2. Fluvoxamine 78-89 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 175-181 9868741-1 1998 The selective serotonin re-uptake inhibitor, fluvoxamine, is a very potent inhibitor of CYP1A2, and accordingly causes pharmacokinetic interactions with drugs metabolised by CYP1A2, such as caffeine, theophylline, imipramine, tacrine and clozapine. Fluvoxamine 45-56 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 88-94 9868741-1 1998 The selective serotonin re-uptake inhibitor, fluvoxamine, is a very potent inhibitor of CYP1A2, and accordingly causes pharmacokinetic interactions with drugs metabolised by CYP1A2, such as caffeine, theophylline, imipramine, tacrine and clozapine. Fluvoxamine 45-56 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 174-180 9888626-4 1998 Pre-exposure with the 5-HT2C receptor agonist MK 212 [6-chloro-2(1-piperazinyl)pyrazine] partially prevented the fluvoxamine-induced CTA, pre-exposure with the 5-HT2A/2C receptor agonist DOI [1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane HCl] did not prevent the CTA induced by fluvoxamine. Fluvoxamine 278-289 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 22-37 9888626-9 1998 We conclude that 5-HT1A receptors are involved in the stimulus properties of both fluvoxamine and fluoxetine, that 5-HT2C receptors are involved in fluvoxamine and especially fluoxetine, and, based primarily on the cross-comparison tests, that the two SSRIs have somewhat different stimulus properties. Fluvoxamine 82-93 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 17-23 9888626-9 1998 We conclude that 5-HT1A receptors are involved in the stimulus properties of both fluvoxamine and fluoxetine, that 5-HT2C receptors are involved in fluvoxamine and especially fluoxetine, and, based primarily on the cross-comparison tests, that the two SSRIs have somewhat different stimulus properties. Fluvoxamine 148-159 5-hydroxytryptamine (serotonin) receptor 2C Mus musculus 115-121 9923581-9 1998 The mechanism of this interaction is probably inhibition of the CYP3A4-mediated first-pass metabolism of buspirone by fluvoxamine. Fluvoxamine 118-129 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-70 9757149-0 1998 Effect of fluvoxamine therapy on the activities of CYP1A2, CYP2D6, and CYP3A as determined by phenotyping. Fluvoxamine 10-21 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 51-57 9923577-0 1998 Fluvoxamine inhibits the CYP2C19-catalysed metabolism of proguanil in vitro. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 25-32 9923577-1 1998 OBJECTIVE: The potent CYP1A2 inhibitor fluvoxamine has recently been shown also to be an effective inhibitor of the CYP2C19-mediated metabolism of the antimalarial drug proguanil in vivo. Fluvoxamine 39-50 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 22-28 9923577-1 1998 OBJECTIVE: The potent CYP1A2 inhibitor fluvoxamine has recently been shown also to be an effective inhibitor of the CYP2C19-mediated metabolism of the antimalarial drug proguanil in vivo. Fluvoxamine 39-50 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 116-123 9923577-11 1998 CONCLUSIONS: Fluvoxamine is a fairly potent inhibitor of CYP2C19 and it has the potential for causing drug-drug interactions with substrates for CYP2C19 such as imipramine, clomipramine, amitriptyline and diazepam. Fluvoxamine 13-24 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 57-64 9923577-11 1998 CONCLUSIONS: Fluvoxamine is a fairly potent inhibitor of CYP2C19 and it has the potential for causing drug-drug interactions with substrates for CYP2C19 such as imipramine, clomipramine, amitriptyline and diazepam. Fluvoxamine 13-24 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 145-152 9857976-0 1998 Polymorphism within the promoter of the serotonin transporter gene and antidepressant efficacy of fluvoxamine. Fluvoxamine 98-109 solute carrier family 6 member 4 Homo sapiens 40-61 9857976-4 1998 We tested the hypothesis that allelic variation of the 5-HTT promoter could be related to the antidepressant response to fluvoxamine and/or augmentation with pindolol (a serotonin autoreceptors antagonist) which has been suggested as an augmentation therapy for nonresponders. Fluvoxamine 121-132 solute carrier family 6 member 4 Homo sapiens 55-60 9857976-9 1998 Both homozygotes for the long variant (l/l) of the 5-HTT promoter and heterozygotes (l/s) showed a better response to fluvoxamine than homozygotes for the short variant (s/s). Fluvoxamine 118-129 solute carrier family 6 member 4 Homo sapiens 51-56 9857976-11 1998 Fluvoxamine efficacy in delusional depression seems to be related to allelic variation within the promoter of the 5-HTT gene. Fluvoxamine 0-11 solute carrier family 6 member 4 Homo sapiens 114-119 9757149-0 1998 Effect of fluvoxamine therapy on the activities of CYP1A2, CYP2D6, and CYP3A as determined by phenotyping. Fluvoxamine 10-21 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 59-65 9757149-0 1998 Effect of fluvoxamine therapy on the activities of CYP1A2, CYP2D6, and CYP3A as determined by phenotyping. Fluvoxamine 10-21 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 71-76 9757149-1 1998 OBJECTIVE: To determine the effect of 150 mg/day fluvoxamine on the activities of CYP1A2, CYP2D6, CYP3A, N-acetyltransferase-2 (NAT2), and xanthine oxidase (XO) by phenotyping with caffeine, dextromethorphan, and midazolam. Fluvoxamine 49-60 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 82-88 9757149-1 1998 OBJECTIVE: To determine the effect of 150 mg/day fluvoxamine on the activities of CYP1A2, CYP2D6, CYP3A, N-acetyltransferase-2 (NAT2), and xanthine oxidase (XO) by phenotyping with caffeine, dextromethorphan, and midazolam. Fluvoxamine 49-60 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 90-96 9757149-1 1998 OBJECTIVE: To determine the effect of 150 mg/day fluvoxamine on the activities of CYP1A2, CYP2D6, CYP3A, N-acetyltransferase-2 (NAT2), and xanthine oxidase (XO) by phenotyping with caffeine, dextromethorphan, and midazolam. Fluvoxamine 49-60 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 98-103 9757149-1 1998 OBJECTIVE: To determine the effect of 150 mg/day fluvoxamine on the activities of CYP1A2, CYP2D6, CYP3A, N-acetyltransferase-2 (NAT2), and xanthine oxidase (XO) by phenotyping with caffeine, dextromethorphan, and midazolam. Fluvoxamine 49-60 N-acetyltransferase 2 Homo sapiens 105-126 9757149-1 1998 OBJECTIVE: To determine the effect of 150 mg/day fluvoxamine on the activities of CYP1A2, CYP2D6, CYP3A, N-acetyltransferase-2 (NAT2), and xanthine oxidase (XO) by phenotyping with caffeine, dextromethorphan, and midazolam. Fluvoxamine 49-60 N-acetyltransferase 2 Homo sapiens 128-132 9757149-8 1998 For CYP1A2, CYP2D6, and CYP3A phenotypes, significant differences existed between baseline and fluvoxamine therapy. Fluvoxamine 95-106 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 4-10 9757149-8 1998 For CYP1A2, CYP2D6, and CYP3A phenotypes, significant differences existed between baseline and fluvoxamine therapy. Fluvoxamine 95-106 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 12-18 9757149-8 1998 For CYP1A2, CYP2D6, and CYP3A phenotypes, significant differences existed between baseline and fluvoxamine therapy. Fluvoxamine 95-106 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 9757149-9 1998 For CYP1A2, the mean urinary metabolite ratio (+/-SD) was 7.53 +/- 7.44 at baseline and 4.30 +/- 2.82 with fluvoxamine ( P = .012). Fluvoxamine 107-118 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 4-10 9757149-12 1998 For CYP1A2, CYP2D6, and CYP3A, fluvoxamine therapy changed the phenotyping measures by a median of -44.4%, 123.5%, and -34.4%, respectively. Fluvoxamine 31-42 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 4-10 9757149-12 1998 For CYP1A2, CYP2D6, and CYP3A, fluvoxamine therapy changed the phenotyping measures by a median of -44.4%, 123.5%, and -34.4%, respectively. Fluvoxamine 31-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 24-29 9757149-13 1998 CONCLUSIONS: We concluded that fluvoxamine may cause significant inhibition of CYP1A2, CYP2D6, and CYP3A activity. Fluvoxamine 31-42 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 79-85 9757149-13 1998 CONCLUSIONS: We concluded that fluvoxamine may cause significant inhibition of CYP1A2, CYP2D6, and CYP3A activity. Fluvoxamine 31-42 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 87-93 9757149-13 1998 CONCLUSIONS: We concluded that fluvoxamine may cause significant inhibition of CYP1A2, CYP2D6, and CYP3A activity. Fluvoxamine 31-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-104 9469504-8 1998 Interestingly, both nefazodone and fluvoxamine are known to potently inhibit the CYP3A3/4 isoenzymes. Fluvoxamine 35-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 81-87 9817620-4 1998 Accordingly fluvoxamine has interactions with other drugs eliminated by CYP1A2 including caffeine, clozapine, olanzapine, theophylline, propranolol and tacrine. Fluvoxamine 12-23 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 72-78 9817620-10 1998 CYP2D6 only makes up about 2-5% of the total P450 in the human liver, but anyway is the major enzyme catalyzing more than 30 clinically used drugs including all of the tricyclic antidepressants, several neuroleptics, opiates, betablockers, antiarrhythmics and among the SSRIs N-desmethylcitalopram, fluvoxamine, fluoxetine and paroxetine but not sertraline. Fluvoxamine 299-310 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 9617978-0 1998 Determinants of interindividual variability and extent of CYP2D6 and CYP1A2 inhibition by paroxetine and fluvoxamine in vivo. Fluvoxamine 105-116 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 58-64 9617978-0 1998 Determinants of interindividual variability and extent of CYP2D6 and CYP1A2 inhibition by paroxetine and fluvoxamine in vivo. Fluvoxamine 105-116 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 69-75 9617978-3 1998 The antidepressants paroxetine and fluvoxamine are potent in vitro inhibitors of CYP2D6 and CYP1A2 isozymes, respectively. Fluvoxamine 35-46 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 81-87 9617978-3 1998 The antidepressants paroxetine and fluvoxamine are potent in vitro inhibitors of CYP2D6 and CYP1A2 isozymes, respectively. Fluvoxamine 35-46 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 92-98 9617978-6 1998 The aim of this study was to assess the determinants of interindividual variability and extent of CYP2D6 and CYP1A2 inhibition during paroxetine and fluvoxamine treatment. Fluvoxamine 149-160 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 98-104 9617978-6 1998 The aim of this study was to assess the determinants of interindividual variability and extent of CYP2D6 and CYP1A2 inhibition during paroxetine and fluvoxamine treatment. Fluvoxamine 149-160 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 109-115 9617978-16 1998 The extent of inhibition of CYP2D6 and CYP1A2 by paroxetine and fluvoxamine, respectively, displayed a positive correlation with baseline enzyme activity (p < 0.05). Fluvoxamine 64-75 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 28-34 9617978-16 1998 The extent of inhibition of CYP2D6 and CYP1A2 by paroxetine and fluvoxamine, respectively, displayed a positive correlation with baseline enzyme activity (p < 0.05). Fluvoxamine 64-75 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 39-45 9617978-18 1998 These data indicate that paroxetine and fluvoxamine treatment with minimum clinically effective doses significantly inhibit CYP2D6 and CYP1A2, respectively. Fluvoxamine 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 124-130 9617978-18 1998 These data indicate that paroxetine and fluvoxamine treatment with minimum clinically effective doses significantly inhibit CYP2D6 and CYP1A2, respectively. Fluvoxamine 40-51 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 135-141 9617978-19 1998 The extent of inhibition of CYP2D6 by paroxetine and of CYP1A2 by fluvoxamine is dependent in part on the baseline enzyme activity. Fluvoxamine 66-77 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 28-34 9617978-19 1998 The extent of inhibition of CYP2D6 by paroxetine and of CYP1A2 by fluvoxamine is dependent in part on the baseline enzyme activity. Fluvoxamine 66-77 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 56-62 9617978-21 1998 Most patients treated with fluvoxamine (50-100 mg/day) will reach population minimums for CYP1A2 activity. Fluvoxamine 27-38 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 90-96 9669506-2 1998 The selective 5-HT reuptake inhibitors paroxetine, indalpine and fluvoxamine displayed a high affinity for the 5-HT transporter, whereas the norepinephrine reuptake inhibitor desipramine had a high affinity for the norepinephrine transporter. Fluvoxamine 65-76 solute carrier family 6 member 4 Rattus norvegicus 111-127 9669506-2 1998 The selective 5-HT reuptake inhibitors paroxetine, indalpine and fluvoxamine displayed a high affinity for the 5-HT transporter, whereas the norepinephrine reuptake inhibitor desipramine had a high affinity for the norepinephrine transporter. Fluvoxamine 65-76 solute carrier family 6 member 2 Rattus norvegicus 215-241 9817620-3 1998 Thus fluvoxamine, but not citalopram, fluoxetine, paroxetine and sertraline is a potent inhbitor of CYP1A2. Fluvoxamine 5-16 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 100-106 9832352-10 1998 The results of this pilot study should be regarded as a hint at the possible therapeutic benefits of lower fluvoxamine serum concentrations by means of lower fluvoxamine dosages. Fluvoxamine 107-118 histidine triad nucleotide binding protein 1 Homo sapiens 56-60 9832352-10 1998 The results of this pilot study should be regarded as a hint at the possible therapeutic benefits of lower fluvoxamine serum concentrations by means of lower fluvoxamine dosages. Fluvoxamine 158-169 histidine triad nucleotide binding protein 1 Homo sapiens 56-60 9716316-0 1998 Patterns of c-fos expression induced by fluvoxamine are different after acute vs. chronic oral administration. Fluvoxamine 40-51 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 12-17 9716316-2 1998 We used the expression of c-fos, after both acute and chronic oral administration of fluvoxamine in the rat, to study its immediate and long-term effects, in relation to the distribution of Galanin (GAL) and Vasoactive Intestinal Polypeptide (VIP). Fluvoxamine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 26-31 9716316-5 1998 It is concluded that activation of 5-HT3-receptors in the caudal brainstem or gastro-intestinal afferents of the vagal nerve may play a role in the observed pattern of Fos-IR after fluvoxamine administration. Fluvoxamine 181-192 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 168-171 10022747-0 1998 Use of heterologously expressed human cytochrome P450 1A2 to predict tacrine-fluvoxamine drug interaction in man. Fluvoxamine 77-88 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 38-57 10022747-6 1998 The Ki of fluvoxamine on 1-hydroxytacrine formation rate observed with rH-CYP1A2 was similar to that observed with human liver microsome (0.35+/-0.05 versus 0.20+/-0.20 microM, respectively). Fluvoxamine 10-21 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 74-80 10022747-7 1998 Using the Km, Vmax and Ki determined with rH-CYP1A2, we calculated that fluvoxamine produced an inhibition of 1-, 2- and 4-hydroxytacrine formation rate of 91, 87 and 88%, respectively, in the range of tacrine and fluvoxamine concentrations observed in man. Fluvoxamine 72-83 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 45-51 9435993-14 1997 Fluvoxamine, as a potent inhibitor of CYP1A2, can inhibit the metabolism of clozapine, resulting in higher plasma concentrations. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 38-44 9384467-6 1997 Thus, assuming comparable molar concentrations at the site of inhibition, fluvoxamine can be expected to have the highest probability of interfering with the metabolism of CYP2C9 substrates. Fluvoxamine 74-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 9418315-8 1997 The positive response to buspirone (5-HT1A) augmentation of fluvoxamine (SSRI) suggested that disturbed central serotonergic neurotransmission might play an important role in the pathogenesis of kleptomania. Fluvoxamine 60-71 5-hydroxytryptamine receptor 1A Homo sapiens 36-42 9442550-5 1997 With regard to interactions of SSRIs and clozapine, fluvoxamine, a potent inhibitor of cytochrome P450 1A2, gives rise to higher clozapine levels at an earlier time, compared to other SSRIs (paroxetine, fluoxetine and sertraline), which are potent cytochrome P450 2D6 inhibitors. Fluvoxamine 52-63 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 87-106 9353355-5 1997 Omeprazole 5-hydroxylation by liver microsomes of a human sample that contained relatively high levels of CYP3A4 and low levels of CYP2C19 were inhibited very significantly by ketoconazole and anti-CYP3A4 antibodies, although a human sample having high in CYP2C19 and low in CYP3A4 was found to be sensitive toward fluvoxamine and anti-CYP2C9 antibodies. Fluvoxamine 315-326 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 106-112 9353355-5 1997 Omeprazole 5-hydroxylation by liver microsomes of a human sample that contained relatively high levels of CYP3A4 and low levels of CYP2C19 were inhibited very significantly by ketoconazole and anti-CYP3A4 antibodies, although a human sample having high in CYP2C19 and low in CYP3A4 was found to be sensitive toward fluvoxamine and anti-CYP2C9 antibodies. Fluvoxamine 315-326 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 131-138 9353355-5 1997 Omeprazole 5-hydroxylation by liver microsomes of a human sample that contained relatively high levels of CYP3A4 and low levels of CYP2C19 were inhibited very significantly by ketoconazole and anti-CYP3A4 antibodies, although a human sample having high in CYP2C19 and low in CYP3A4 was found to be sensitive toward fluvoxamine and anti-CYP2C9 antibodies. Fluvoxamine 315-326 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 198-204 9353355-5 1997 Omeprazole 5-hydroxylation by liver microsomes of a human sample that contained relatively high levels of CYP3A4 and low levels of CYP2C19 were inhibited very significantly by ketoconazole and anti-CYP3A4 antibodies, although a human sample having high in CYP2C19 and low in CYP3A4 was found to be sensitive toward fluvoxamine and anti-CYP2C9 antibodies. Fluvoxamine 315-326 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 198-204 9442550-5 1997 With regard to interactions of SSRIs and clozapine, fluvoxamine, a potent inhibitor of cytochrome P450 1A2, gives rise to higher clozapine levels at an earlier time, compared to other SSRIs (paroxetine, fluoxetine and sertraline), which are potent cytochrome P450 2D6 inhibitors. Fluvoxamine 52-63 cytochrome P450 2D6 Homo sapiens 248-267 9333103-0 1997 Fluvoxamine inhibits the CYP2C19-catalyzed bioactivation of chloroguanide. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 25-32 9333103-1 1997 OBJECTIVE: To investigate the interaction between fluvoxamine and chloroguanide (INN, proguanil) to confirm that fluvoxamine inhibits CYP2C19. Fluvoxamine 113-124 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 134-141 9333103-13 1997 CONCLUSION: Fluvoxamine is an effective inhibitor of CYP2C19. Fluvoxamine 12-23 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 53-60 9335078-0 1997 Effect of fluvoxamine on platelet 5-HT2A receptors as studied by [3H]lysergic acid diethylamide ([3H]LSD) binding in healthy volunteers. Fluvoxamine 10-21 5-hydroxytryptamine receptor 2A Homo sapiens 34-40 9335078-6 1997 The study demonstrates that fluvoxamine affects platelet 5-HT2A receptor status irrespective of underlying psychiatric disease, and that this effect is evident already after 1 week at a subtherapeutic fluvoxamine dose. Fluvoxamine 28-39 5-hydroxytryptamine receptor 2A Homo sapiens 57-72 9232209-0 1997 Plasma prolactin response to d-fenfluramine in obsessive-compulsive patients before and after fluvoxamine treatment. Fluvoxamine 94-105 prolactin Homo sapiens 7-16 9232209-5 1997 After 10-week fluvoxamine treatment, the PRL response to the serotonergic agent normalized. Fluvoxamine 14-25 prolactin Homo sapiens 41-44 9223030-2 1997 The present study was designed to investigate whether ACTH 4-9 interferes with the effects of antidepressants: fluoxetine (FLU), fluvoxamine (FOX), selegiline (SEL) or dopamine agonists: piribedil (PRB) or quinpirol (QPR) in forced swimming test and in open field in rats. Fluvoxamine 129-140 proopiomelanocortin Homo sapiens 54-58 9241009-0 1997 The effect of fluvoxamine on serum prolactin and serum sodium concentrations: relation to platelet 5-HT2A receptor status. Fluvoxamine 14-25 prolactin Homo sapiens 35-44 9241009-6 1997 Two subjects had substantial increases in serum prolactin levels (up to 35 microg/L) during fluvoxamine treatment, and these two subjects had higher Bmax for platelet [3H]LSD binding before fluvoxamine treatment than the six other subjects (32.7 vs. 23.1 fmol/mg protein). Fluvoxamine 92-103 prolactin Homo sapiens 48-57 9241009-9 1997 The results indicate that fluvoxamine affects serum prolactin as well as serum sodium concentrations and lend indirect support to the suggestion that 5-HT2A receptors might be involved in the mediation of these effects. Fluvoxamine 26-37 prolactin Homo sapiens 52-61 9241009-9 1997 The results indicate that fluvoxamine affects serum prolactin as well as serum sodium concentrations and lend indirect support to the suggestion that 5-HT2A receptors might be involved in the mediation of these effects. Fluvoxamine 26-37 5-hydroxytryptamine receptor 2A Homo sapiens 150-156 9209244-2 1997 Fluvoxamine, a potent CYP1A2 inhibitor, may be coadministered with tacrine. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 22-28 9330023-5 1997 RESULTS: PRL response in OCD patients was blunted under the drug-free condition; correlated inversely with pretreatment ratings of obsessive-compulsive and depressive symptomatology; and correlated inversely with the improvement in obsessive-compulsive score observed after fluvoxamine treatment. Fluvoxamine 274-285 prolactin Homo sapiens 9-12 9147017-2 1997 Fluoxetine and fluvoxamine reinforced the response to norepinephrine of isolated rat vas deferens incubated in Krebs-Henseleit solution. Fluvoxamine 15-26 arginine vasopressin Rattus norvegicus 85-88 9090339-1 1997 OBJECTIVE: The purpose of this study was to investigate the pharmacokinetics of fluvoxamine in the human brain by using fluorine-19 magnetic resonance spectroscopy (19F MRS) and to assess the relationships among fluvoxamine brain levels, fluvoxamine plasma levels, and clinical efficacy. Fluvoxamine 80-91 MROS Homo sapiens 169-172 10950475-3 1997 This suggests that cytochrome P450IID6 (CYP2D6), an enzyme that is strongly inhibited by FLX, preferentially metabolizes (R)-MTD, whereas CYP1A2, which is strongly inhibited by FLV, metabolizes both enantiomers. Fluvoxamine 177-180 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 40-46 10950475-3 1997 This suggests that cytochrome P450IID6 (CYP2D6), an enzyme that is strongly inhibited by FLX, preferentially metabolizes (R)-MTD, whereas CYP1A2, which is strongly inhibited by FLV, metabolizes both enantiomers. Fluvoxamine 177-180 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 138-144 9085405-0 1997 Effect of the selective serotonin reuptake inhibitor fluvoxamine on CCK-4 induced panic attacks. Fluvoxamine 53-64 protein tyrosine kinase 7 (inactive) Homo sapiens 68-73 9105404-0 1997 Distinction of CYP1A1 and CYP1A2 activity by selective inhibition using fluvoxamine and isosafrole. Fluvoxamine 72-83 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 15-21 9105404-0 1997 Distinction of CYP1A1 and CYP1A2 activity by selective inhibition using fluvoxamine and isosafrole. Fluvoxamine 72-83 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 26-32 9105404-5 1997 Isosafrole and fluvoxamine were found to inhibit CYP1A2 selectively, with Ki values of 14 and 800 times, respectively, lower than those for CY1A1. Fluvoxamine 15-26 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 49-55 9184622-12 1997 Fluvoxamine has prominent affinity for the CYP12 isozyme, lesser affinity for the CYP3A4 and CYP2C isozymes, and minimal affinity for CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 8 subfamily B member 1 Homo sapiens 43-48 9029748-7 1997 Fluvoxamine is a potent inhibitor of CYP1A2, and period B was included as a positive control. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-43 9174682-0 1997 Relationship between fluvoxamine pharmacokinetics and CYP2D6/CYP2C19 phenotype polymorphisms. Fluvoxamine 21-32 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 54-60 9174682-0 1997 Relationship between fluvoxamine pharmacokinetics and CYP2D6/CYP2C19 phenotype polymorphisms. Fluvoxamine 21-32 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 61-68 9174682-1 1997 OBJECTIVE: The purpose of this study was to investigate whether the disposition of fluvoxamine is associated with the CYP2D6 and CYP2C19 phenotype polymorphisms. Fluvoxamine 83-94 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 118-124 9174682-1 1997 OBJECTIVE: The purpose of this study was to investigate whether the disposition of fluvoxamine is associated with the CYP2D6 and CYP2C19 phenotype polymorphisms. Fluvoxamine 83-94 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 129-136 9174682-5 1997 CONCLUSION: The results are consistent with a possible minor to moderate role of CYP2D6, but not CYP2C19, in fluvoxamine metabolism. Fluvoxamine 109-120 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 81-87 9184622-12 1997 Fluvoxamine has prominent affinity for the CYP12 isozyme, lesser affinity for the CYP3A4 and CYP2C isozymes, and minimal affinity for CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 82-88 9184622-12 1997 Fluvoxamine has prominent affinity for the CYP12 isozyme, lesser affinity for the CYP3A4 and CYP2C isozymes, and minimal affinity for CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 93-98 9184622-12 1997 Fluvoxamine has prominent affinity for the CYP12 isozyme, lesser affinity for the CYP3A4 and CYP2C isozymes, and minimal affinity for CYP2D6. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 134-140 8968657-11 1996 CYP2D6 is inhibited by SSRIs, in order of decreasing potency paroxetine, norfluoxetine, fluoxetine, sertraline, citalopram and fluvoxamine. Fluvoxamine 127-138 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 8968657-19 1996 Fluvoxamine inhibits with decreasing potency, CYP1A2, CYP2C19, CYP2D6 and CYP1A1, but it is also an inhibitor of CYP3A. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 46-52 8968657-19 1996 Fluvoxamine inhibits with decreasing potency, CYP1A2, CYP2C19, CYP2D6 and CYP1A1, but it is also an inhibitor of CYP3A. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 54-61 8968657-19 1996 Fluvoxamine inhibits with decreasing potency, CYP1A2, CYP2C19, CYP2D6 and CYP1A1, but it is also an inhibitor of CYP3A. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 63-69 8968657-19 1996 Fluvoxamine inhibits with decreasing potency, CYP1A2, CYP2C19, CYP2D6 and CYP1A1, but it is also an inhibitor of CYP3A. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 74-80 8968657-19 1996 Fluvoxamine inhibits with decreasing potency, CYP1A2, CYP2C19, CYP2D6 and CYP1A1, but it is also an inhibitor of CYP3A. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 113-118 9032002-8 1996 Fluvoxamine is a substantial inhibitor of CYP1A2 and CYP2C19, and a moderate inhibitor of CYP3A3/4. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 42-48 9032002-8 1996 Fluvoxamine is a substantial inhibitor of CYP1A2 and CYP2C19, and a moderate inhibitor of CYP3A3/4. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 53-60 9032002-8 1996 Fluvoxamine is a substantial inhibitor of CYP1A2 and CYP2C19, and a moderate inhibitor of CYP3A3/4. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 8986010-6 1996 Among six selective serotonin reuptake inhibitor (SSRI) antidepressants, fluvoxamine was a potent inhibitor of 1A2 (mean Ki1 = 0.24 microM). Fluvoxamine 73-84 TNF receptor superfamily member 8 Homo sapiens 121-124 8986013-5 1996 According to a previous in vitro study, this pharmacokinetic interaction occurs on the level of CYP2C19, but also of CYP2D6 and CYP3A4 which, in contrast to CYP1A2, contribute to the N-demethylation of citalopram and which are stereoselectively inhibited by fluvoxamine. Fluvoxamine 258-269 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 128-134 8986013-5 1996 According to a previous in vitro study, this pharmacokinetic interaction occurs on the level of CYP2C19, but also of CYP2D6 and CYP3A4 which, in contrast to CYP1A2, contribute to the N-demethylation of citalopram and which are stereoselectively inhibited by fluvoxamine. Fluvoxamine 258-269 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 96-103 8986013-5 1996 According to a previous in vitro study, this pharmacokinetic interaction occurs on the level of CYP2C19, but also of CYP2D6 and CYP3A4 which, in contrast to CYP1A2, contribute to the N-demethylation of citalopram and which are stereoselectively inhibited by fluvoxamine. Fluvoxamine 258-269 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 117-123 8838442-6 1996 The average apparent Ki values determined using CYP2D6-dependent dextromethorphan O-demethylation were: 33, 52 and 22 microM for rac-venlafaxine, R(+)-venlafaxine and S(-)-venlafaxine, respectively, vs 0.065 to 1.8 microM for paroxetine, fluoxetine, norfluoxetine, fluvoxamine and sertraline. Fluvoxamine 265-276 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 48-54 8885123-1 1996 All available antidepressants with the exception of fluvoxamine and nefazodone either are metabolized by cytochrome P450 2D6 (CYP2D6) and/or inhibit this isozyme. Fluvoxamine 52-63 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 105-124 8885123-1 1996 All available antidepressants with the exception of fluvoxamine and nefazodone either are metabolized by cytochrome P450 2D6 (CYP2D6) and/or inhibit this isozyme. Fluvoxamine 52-63 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 126-132 8823236-0 1996 Disposition of fluvoxamine in humans is determined by the polymorphic CYP2D6 and also by the CYP1A2 activity. Fluvoxamine 15-26 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 70-76 8823236-0 1996 Disposition of fluvoxamine in humans is determined by the polymorphic CYP2D6 and also by the CYP1A2 activity. Fluvoxamine 15-26 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 93-99 8823236-2 1996 This study investigated the relationship between fluvoxamine disposition and the polymorphic CYP2D6 and the polycyclic aromatic hydrocarbon (as contained in cigarette smoke) inducible CYP1A2. Fluvoxamine 49-60 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 93-99 8823236-2 1996 This study investigated the relationship between fluvoxamine disposition and the polymorphic CYP2D6 and the polycyclic aromatic hydrocarbon (as contained in cigarette smoke) inducible CYP1A2. Fluvoxamine 49-60 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 184-190 8823236-7 1996 The oral clearance of fluvoxamine correlated to the CYP1A2 index in the 14 subjects (rs = 0.58; p < 0.05; Spearman rank correlation). Fluvoxamine 22-33 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 52-58 8823236-8 1996 CONCLUSION: The disposition of fluvoxamine in humans is associated with the polymorphic CYP2D6 activity, but CYP1A2 also seems to be involved. Fluvoxamine 31-42 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 88-94 8823236-8 1996 CONCLUSION: The disposition of fluvoxamine in humans is associated with the polymorphic CYP2D6 activity, but CYP1A2 also seems to be involved. Fluvoxamine 31-42 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 109-115 8944409-8 1996 The increased buspirone concentrations are likely to be due to inhibition of first pass liver metabolism by fluvoxamine acting on the cytochrome P-450 system. Fluvoxamine 108-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 8944409-10 1996 The blunting of GH and psychological responses suggest that 5-HT1A receptor function is reduced by chronic fluvoxamine treatment. Fluvoxamine 107-118 5-hydroxytryptamine receptor 1A Homo sapiens 60-75 8904628-1 1996 Studies were performed in eight healthy extensive metabolizers of mephenytoin and debrisoquine to determine the effect of fluvoxamine on the activities of S-mephenytoin 4"-hydroxylase (CYP2C19) and metoprolol alpha-hydroxylase (CYP2D6). Fluvoxamine 122-133 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 155-183 8904628-1 1996 Studies were performed in eight healthy extensive metabolizers of mephenytoin and debrisoquine to determine the effect of fluvoxamine on the activities of S-mephenytoin 4"-hydroxylase (CYP2C19) and metoprolol alpha-hydroxylase (CYP2D6). Fluvoxamine 122-133 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 185-192 8904628-6 1996 These results indicated fluvoxamine has a modest inhibitory effect on the activity of CYP2C19, but no effect on that of CYP2D6 in vivo. Fluvoxamine 24-35 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 86-93 8610817-5 1996 Cytochrome P450 1A2 is inhibited by fluvoxamine and is implicated in drug interactions with theophylline, clozapine, and others. Fluvoxamine 36-47 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 0-19 8732441-3 1996 Thus, among the SSRIs, fluvoxamine is the only very potent inhibitor of cytochrome P4501A2 (CYP1A2). Fluvoxamine 23-34 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 72-90 8732441-3 1996 Thus, among the SSRIs, fluvoxamine is the only very potent inhibitor of cytochrome P4501A2 (CYP1A2). Fluvoxamine 23-34 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 92-98 8587855-3 1995 This review summarizes information to date regarding the inhibitory potency of fluoxetine, fluvoxamine, paroxetine, sertraline, nefazodone, and venlafaxine on five isoenzyme systems: CYP2D6, CYP3A3/4, CYP1A2, CYP2C9, and CYP2C19. Fluvoxamine 91-102 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 183-189 8880055-13 1996 CONCLUSION: This investigation confirms that paroxetine and fluoxetine are potent inhibitors of CYP2D6, that fluvoxamine and fluoxetine are moderate inhibitors of CYP2C19 and that fluvoxamine is a potent inhibitor of CYP1A2 in humans in vivo. Fluvoxamine 109-120 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 163-170 8880055-13 1996 CONCLUSION: This investigation confirms that paroxetine and fluoxetine are potent inhibitors of CYP2D6, that fluvoxamine and fluoxetine are moderate inhibitors of CYP2C19 and that fluvoxamine is a potent inhibitor of CYP1A2 in humans in vivo. Fluvoxamine 180-191 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 96-102 8880055-13 1996 CONCLUSION: This investigation confirms that paroxetine and fluoxetine are potent inhibitors of CYP2D6, that fluvoxamine and fluoxetine are moderate inhibitors of CYP2C19 and that fluvoxamine is a potent inhibitor of CYP1A2 in humans in vivo. Fluvoxamine 180-191 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 217-223 8771220-8 1995 Baseline beta-EP values were normal in the bulimic patients but had declined by month 4 of fluvoxamine therapy. Fluvoxamine 91-102 proopiomelanocortin Homo sapiens 9-16 8880055-0 1996 Dose-dependent inhibition of CYP1A2, CYP2C19 and CYP2D6 by citalopram, fluoxetine, fluvoxamine and paroxetine. Fluvoxamine 83-94 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 29-35 8880055-0 1996 Dose-dependent inhibition of CYP1A2, CYP2C19 and CYP2D6 by citalopram, fluoxetine, fluvoxamine and paroxetine. Fluvoxamine 83-94 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 37-44 8880055-0 1996 Dose-dependent inhibition of CYP1A2, CYP2C19 and CYP2D6 by citalopram, fluoxetine, fluvoxamine and paroxetine. Fluvoxamine 83-94 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 49-55 8880055-1 1996 OBJECTIVE: The purpose of this pharmacokinetic study was to investigate the dose-dependent inhibition of model substrates for CYP2D6, CYP2C19 and CYP1A2 by four marketed selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine and paroxetine. Fluvoxamine 243-254 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 126-132 8880055-1 1996 OBJECTIVE: The purpose of this pharmacokinetic study was to investigate the dose-dependent inhibition of model substrates for CYP2D6, CYP2C19 and CYP1A2 by four marketed selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine and paroxetine. Fluvoxamine 243-254 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 134-141 8880055-1 1996 OBJECTIVE: The purpose of this pharmacokinetic study was to investigate the dose-dependent inhibition of model substrates for CYP2D6, CYP2C19 and CYP1A2 by four marketed selective serotonin reuptake inhibitors (SSRIs): citalopram, fluoxetine, fluvoxamine and paroxetine. Fluvoxamine 243-254 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 146-152 7770611-1 1995 This study investigated the alterations of the 5-HT1A and 5-HT1B autoreceptor function following chronic treatment with fluvoxamine using osmotic minipumps. Fluvoxamine 120-131 5-hydroxytryptamine receptor 1A Rattus norvegicus 47-64 7586931-1 1995 OBJECTIVES: Although fluvoxamine inhibits the biotransformation of drugs known to be metabolized by CYP1A2, there are no data available with regard to the importance of CYP1A2 for the metabolism of fluvoxamine itself. Fluvoxamine 21-32 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 100-106 7586931-2 1995 Because smoking induces the metabolism of drugs catalyzed by CYP1A2, this study investigated the pharmacokinetics of fluvoxamine in smokers and nonsmokers. Fluvoxamine 117-128 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 61-67 7586931-8 1995 This finding is consistent with a possible role of CYP1A2 in fluvoxamine metabolism. Fluvoxamine 61-72 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 51-57 7479582-7 1995 Propylene glycol inhibited the activity of CYP2E1 as indicated by 84% reduction in the clearance of 3 mg/kg dose of chlorzoxazone, whereas fluvoxamine inhibited the activity of CYP1A2 as indicated by 40% reduction in the clearance of a 10 mg/kg dose of caffeine. Fluvoxamine 139-150 cytochrome P450, family 1, subfamily a, polypeptide 2 Mus musculus 177-183 7494384-0 1995 5-HT1A-receptor subtype mediates the effect of fluvoxamine, a selective serotonin reuptake inhibitor, on marble-burying behavior in mice. Fluvoxamine 47-58 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 0-15 7494384-10 1995 However, the 5-HT1A antagonist NAN-190 (1-(2-methoxyphenyl)-4-[4-(2-phthalimido)butyl] piperazine) inhibited the suppressive effect of fluvoxamine on the marble-burying. Fluvoxamine 135-146 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 13-19 7494384-11 1995 From these results, the 5-HT1A-receptor subtype may be involved in the suppressive effect of fluvoxamine on the marble-burying, but the 5-HT2-receptor subtype is not involved in this effect. Fluvoxamine 93-104 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 24-39 7494385-7 1995 On the other hand, the 5-HT1A antagonist NAN-190 (1-(2-methoxyphenyl)-4-[4-(2-phthalimido)butyl] piperazine) potentiated the effect of fluvoxamine on forced-swimming. Fluvoxamine 135-146 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 23-29 7494385-8 1995 It is expected, however, that a 5-HT1A antagonist should antagonize the effect of fluvoxamine when 5-HT1A mediates the suppressive effect of fluvoxamine on the immobility time in forced-swimming. Fluvoxamine 82-93 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 32-38 7494385-8 1995 It is expected, however, that a 5-HT1A antagonist should antagonize the effect of fluvoxamine when 5-HT1A mediates the suppressive effect of fluvoxamine on the immobility time in forced-swimming. Fluvoxamine 141-152 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 32-38 7494385-8 1995 It is expected, however, that a 5-HT1A antagonist should antagonize the effect of fluvoxamine when 5-HT1A mediates the suppressive effect of fluvoxamine on the immobility time in forced-swimming. Fluvoxamine 141-152 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 99-105 7617560-0 1995 Kinetics and inhibition by fluvoxamine of phenacetin O-deethylation in V79 cells expressing human CYP1A2. Fluvoxamine 27-38 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 98-104 7622807-2 1995 From a clinical point of view, it is of relevance that potency to inhibit the cytochrome P450 isozyme CYP2D6 gradually decreases from paroxetine, fluoxetine, norfluoxetine, desmethylcitalopram, fluvoxamine, and sertraline down to citalopram, explaining to a great extent differences in pharmacokinetic interactions between the SSRIs and tricyclic antidepressants, which are metabolized by this enzyme. Fluvoxamine 194-205 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 102-108 7622807-3 1995 Fluvoxamine interacts with these drugs by a mechanism involving inhibition of CYP1A2, CYP3A4, and CYP2C19. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 78-84 7622807-3 1995 Fluvoxamine interacts with these drugs by a mechanism involving inhibition of CYP1A2, CYP3A4, and CYP2C19. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 86-92 7622807-3 1995 Fluvoxamine interacts with these drugs by a mechanism involving inhibition of CYP1A2, CYP3A4, and CYP2C19. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 98-105 8685072-12 1995 This study suggests, that fluoxetine and fluvoxamine differ in their interaction with the metabolism of some other basic psychotropic drugs, by a mechanism which implies CYP2D6 and CYPmeph and possibly other isoformes of cytochrome P-450. Fluvoxamine 41-52 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 170-176 8685072-12 1995 This study suggests, that fluoxetine and fluvoxamine differ in their interaction with the metabolism of some other basic psychotropic drugs, by a mechanism which implies CYP2D6 and CYPmeph and possibly other isoformes of cytochrome P-450. Fluvoxamine 41-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 7742153-6 1995 Norfluoxetine, sertraline and fluvoxamine inhibited CYP1A1 (7-ethoxyresorufin O-deethylase) in microsomes from human placenta (IC50 values 29 microM, 35 microM and 80 microM, respectively). Fluvoxamine 30-41 cytochrome P450 family 1 subfamily A member 1 Homo sapiens 52-58 7742153-7 1995 Fluvoxamine was a potent inhibitor of CYP1A2-mediated 7-ethoxyresorufin O-deethylase activity (IC50 = 0.3 microM) in human liver. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 38-44 7742153-13 1995 Ethanol and fluvoxamine both inhibited 8-hydroxylation by about 45% and, in combination, the compounds decreased the formation of 1,3-dimethyluric acid by 90%, indicating that CYP1A2 and CYP2E1 are equally important isoforms for the 8-hydroxylation of theophylline. Fluvoxamine 12-23 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 176-182 7742153-13 1995 Ethanol and fluvoxamine both inhibited 8-hydroxylation by about 45% and, in combination, the compounds decreased the formation of 1,3-dimethyluric acid by 90%, indicating that CYP1A2 and CYP2E1 are equally important isoforms for the 8-hydroxylation of theophylline. Fluvoxamine 12-23 cytochrome P450 family 2 subfamily E member 1 Homo sapiens 187-193 7742153-15 1995 It is concluded that pharmacokinetic interaction between fluvoxamine and theophylline is due to potent inhibition of CYP1A2. Fluvoxamine 57-68 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 117-123 8775761-2 1995 Fluvoxamine administered for 5 days significantly enhanced the 32P incorporation stimulated by cAMP into MAP2, while it failed to produce this effect after 12 and 21 days. Fluvoxamine 0-11 cathelicidin antimicrobial peptide Rattus norvegicus 95-99 8846617-10 1995 The specific cytochrome P450 (CYP) isoenzymes involved in the metabolism of fluvoxamine are unknown. Fluvoxamine 76-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 8846617-10 1995 The specific cytochrome P450 (CYP) isoenzymes involved in the metabolism of fluvoxamine are unknown. Fluvoxamine 76-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 8846617-11 1995 CYP2D6, which is crucially involved in the metabolism of paroxetine and fluoxetine, appears to play a clinically insignificant role in the metabolism of fluvoxamine. Fluvoxamine 153-164 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 8846617-17 1995 Fluvoxamine inhibits oxidative drug metabolising enzymes (particularly CYP1A2, and less potently and much less potently CYP3A4 and CYP2D6, respectively) and has the potential for clinically significant drug interactions. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 71-77 8846617-17 1995 Fluvoxamine inhibits oxidative drug metabolising enzymes (particularly CYP1A2, and less potently and much less potently CYP3A4 and CYP2D6, respectively) and has the potential for clinically significant drug interactions. Fluvoxamine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 120-126 8846617-17 1995 Fluvoxamine inhibits oxidative drug metabolising enzymes (particularly CYP1A2, and less potently and much less potently CYP3A4 and CYP2D6, respectively) and has the potential for clinically significant drug interactions. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 131-137 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluvoxamine 335-346 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluvoxamine 335-346 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluvoxamine 335-346 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 165-171 8846618-11 1995 Consistent with its minimal in vitro effect on CYP2D6, fluvoxamine shows minimal in vivo pharmacokinetic interaction with desipramine, but does interact with imipramine (approximately 3- to 4-fold increase in AUC) through inhibition of CYP3A3/4, CYP1A2, and CYP2C19. Fluvoxamine 55-66 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 246-252 8846618-11 1995 Consistent with its minimal in vitro effect on CYP2D6, fluvoxamine shows minimal in vivo pharmacokinetic interaction with desipramine, but does interact with imipramine (approximately 3- to 4-fold increase in AUC) through inhibition of CYP3A3/4, CYP1A2, and CYP2C19. Fluvoxamine 55-66 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 258-265 8846619-4 1995 Fluvoxamine is a potent inhibitor of CYP1A2, and there is potential for interaction with drugs that are metabolised by this isoenzyme. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-43 8846619-5 1995 This property of fluvoxamine may be usefully applied to identifying agents that are substrates of CYP1A2, and it has a possible therapeutic application in the prevention of CYP1A2-mediated toxic metabolite formation. Fluvoxamine 17-28 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 98-104 8846619-5 1995 This property of fluvoxamine may be usefully applied to identifying agents that are substrates of CYP1A2, and it has a possible therapeutic application in the prevention of CYP1A2-mediated toxic metabolite formation. Fluvoxamine 17-28 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 173-179 8846620-11 1995 Those agents metabolised by cytochrome P450 1A2 isoenzyme appear most likely to be involved in drug-drug interactions with fluvoxamine. Fluvoxamine 123-134 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 28-47 8846622-8 1995 A number of drugs have been shown to inhibit CYP2C19 in vivo, including fluvoxamine and fluoxetine. Fluvoxamine 72-83 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 45-52 8775761-2 1995 Fluvoxamine administered for 5 days significantly enhanced the 32P incorporation stimulated by cAMP into MAP2, while it failed to produce this effect after 12 and 21 days. Fluvoxamine 0-11 microtubule-associated protein 2 Rattus norvegicus 105-109 8466541-0 1993 Fluvoxamine is a potent inhibitor of cytochrome P4501A2. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-55 8567194-5 1995 As shown in another simple table, the author compared the hepatic enzymatic inhibitions of four selective serotonin reuptake inhibitors and pointed out the inhibition potentials of fluvoxamine at CYP1A2, fluoxetine and paroxetine at CYP2D6, and fluoxetine and fluvoxamine at CYP3A4 if these two SSRIs have higher serum concentrations. Fluvoxamine 181-192 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 196-202 8567194-5 1995 As shown in another simple table, the author compared the hepatic enzymatic inhibitions of four selective serotonin reuptake inhibitors and pointed out the inhibition potentials of fluvoxamine at CYP1A2, fluoxetine and paroxetine at CYP2D6, and fluoxetine and fluvoxamine at CYP3A4 if these two SSRIs have higher serum concentrations. Fluvoxamine 181-192 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 233-239 8567194-5 1995 As shown in another simple table, the author compared the hepatic enzymatic inhibitions of four selective serotonin reuptake inhibitors and pointed out the inhibition potentials of fluvoxamine at CYP1A2, fluoxetine and paroxetine at CYP2D6, and fluoxetine and fluvoxamine at CYP3A4 if these two SSRIs have higher serum concentrations. Fluvoxamine 181-192 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 275-281 7974626-12 1994 The pathways are not known with certainty, but CYP1A2 may be of major importance for the metabolism of clozapine, since fluvoxamine is a potent inhibitor of this enzyme. Fluvoxamine 120-131 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 47-53 7928004-4 1994 The uptake of [3H]5-HT was temperature, sodium and chloride dependent and was potently inhibited by the antidepressants clomipramine, imipramine, fluoxetine and fluvoxamine, which are specific for the 5-HT transporter. Fluvoxamine 161-172 solute carrier family 6 member 4 Homo sapiens 201-217 7938563-6 1994 At endpoint both the decrease in HAMD-scores and the percentage responders (CGI 1-2) differed significantly comparing fluvoxamine with placebo and tended to differ significantly comparing with imipramine in the severely depressed patients. Fluvoxamine 118-129 mitochondrial transcription termination factor 3 Homo sapiens 76-83 8466541-2 1993 The present study demonstrates that fluvoxamine is a very potent inhibitor of the high-affinity O-deethylation of phenacetin, which is catalysed by cytochrome P4501A2 (CYP1A2), in microsomes from three human livers. Fluvoxamine 36-47 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 148-166 8466541-2 1993 The present study demonstrates that fluvoxamine is a very potent inhibitor of the high-affinity O-deethylation of phenacetin, which is catalysed by cytochrome P4501A2 (CYP1A2), in microsomes from three human livers. Fluvoxamine 36-47 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 168-174 8466541-5 1993 Our findings explain the mechanism of the pharmacokinetic interactions between fluvoxamine and drugs that are metabolized by CYP1A2, e.g. theophylline and imipramine. Fluvoxamine 79-90 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 125-131 33806634-6 2021 DDI model performance was evaluated by prediction of clinical studies with rifampicin (CYP2B6 and CYP2C19 inducer), fluvoxamine (CYP2C19 inhibitor) and voriconazole (CYP2B6 and CYP2C19 inhibitor). Fluvoxamine 116-127 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 129-136 33806634-6 2021 DDI model performance was evaluated by prediction of clinical studies with rifampicin (CYP2B6 and CYP2C19 inducer), fluvoxamine (CYP2C19 inhibitor) and voriconazole (CYP2B6 and CYP2C19 inhibitor). Fluvoxamine 116-127 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 129-136 1389950-10 1992 Three other SSRIs, citalopram, desmethylcitalopram and fluvoxamine, were less potent inhibitors of CYP2D6, with apparent Ki-values of 19, 1.3 and 3.9 microM, respectively. Fluvoxamine 55-66 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 99-105 1389951-4 1992 Fluvoxamine (8.2 microM) and citalopram (5.1 microM) also inhibited CYP2D6 activity. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 68-74 1933139-22 1991 Blockers of neuronal 5-HT uptake (citalopram 0.1 or 1 microM, fluvoxamine 1 microM) usually reduced the MSR and, to a lesser extent, the PSR. Fluvoxamine 62-73 5-methyltetrahydrofolate-homocysteine methyltransferase reductase Rattus norvegicus 104-107 34975483-3 2021 Here, I investigated the effect of an antidepressant drug fluvoxamine on membrane trafficking of the SARS-CoV-2 spike protein and its cell host receptor ACE2 in HEK293T cells. Fluvoxamine 58-69 angiotensin converting enzyme 2 Homo sapiens 153-157 34555857-0 2022 Anti-SARS-CoV-2 Action of Fluvoxamine may be Mediated by Endothelial Nitric Oxide Synthase. Fluvoxamine 26-37 nitric oxide synthase 3 Homo sapiens 57-90 34975483-0 2021 Low-Dose Fluvoxamine Modulates Endocytic Trafficking of SARS-CoV-2 Spike Protein: A Potential Mechanism for Anti-COVID-19 Protection by Antidepressants. Fluvoxamine 9-20 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 67-72 34975483-4 2021 A sub-therapeutic concentration (80 nM) of fluvoxamine rapidly upregulated fluid-phase endocytosis, resulting in enhanced accumulation of the spike-ACE2 complex in enlarged early endosomes. Fluvoxamine 43-54 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 142-147 34975483-3 2021 Here, I investigated the effect of an antidepressant drug fluvoxamine on membrane trafficking of the SARS-CoV-2 spike protein and its cell host receptor ACE2 in HEK293T cells. Fluvoxamine 58-69 surface glycoprotein Severe acute respiratory syndrome coronavirus 2 112-117 34975483-4 2021 A sub-therapeutic concentration (80 nM) of fluvoxamine rapidly upregulated fluid-phase endocytosis, resulting in enhanced accumulation of the spike-ACE2 complex in enlarged early endosomes. Fluvoxamine 43-54 angiotensin converting enzyme 2 Homo sapiens 148-152 34564289-7 2021 Fluvoxamine is one of the most potent inhibitors of CYP1A2 and can lead to an increase in clozapine levels. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 52-58 34605737-4 2021 For CYP2D6.39, the kcat values for fluvoxamine, fluoxetine, and milnacipran, but not for paroxetine and fluphenazine, gradually increased with increasing concentrations, indicating activation of the catalyzed reaction.3. Fluvoxamine 35-46 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 4-10 34881402-8 2022 Meanwhile, preadministration of fluvoxamine significantly extended the time needed for the elimination of phenacetin, possibly due to the inhibition of CYP1A2. Fluvoxamine 32-43 cytochrome P450, family 1, subfamily a, polypeptide 2 Rattus norvegicus 152-158 34881402-9 2022 This suggests that the intake of an excess amount of atemoya juice is necessary to cause a change in the pharmacokinetics of phenacetin when the IC50 values for CYP1A2 inhibition by atemoya and fluvoxamine are taken into account. Fluvoxamine 194-205 cytochrome P450, family 1, subfamily a, polypeptide 2 Rattus norvegicus 161-167 34630662-6 2021 MI rats were administered different treatments, including normal saline, YQHX and fluvoxamine, an agonist of the Sigma-1 receptor. Fluvoxamine 82-93 sigma non-opioid intracellular receptor 1 Rattus norvegicus 113-129 34697991-3 2021 Fluvoxamine can bind to S1R and reduce the serotonin uptake of neurons and platelets. Fluvoxamine 0-11 sigma non-opioid intracellular receptor 1 Rattus norvegicus 24-27 34392133-7 2021 Not the selective norepinephrine reuptake inhibitor reboxetine but the selective serotonin reuptake inhibitors fluvoxamine and sertraline upregulated the PGD2-induced osteoprotegerin release, which was further amplified by morphine. Fluvoxamine 111-122 tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin) Mus musculus 167-182 34117140-0 2021 Effect of Genetic Polymorphism of the CYP2D6 Gene on the Efficacy and Safety of Fluvoxamine in Major Depressive Disorder. Fluvoxamine 80-91 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 38-44 34234701-0 2021 Interest of Fluvoxamine as an Add-On to Clozapine in Children With Severe Psychiatric Disorder According to CYP Polymorphisms: Experience From a Case Series. Fluvoxamine 12-23 peptidylprolyl isomerase G Homo sapiens 108-111 34234701-5 2021 Clozapine plasma levels may be increased with CYP inhibitors such as fluvoxamine. Fluvoxamine 69-80 peptidylprolyl isomerase G Homo sapiens 46-49 34117140-13 2021 CONCLUSIONS: The effect of genetic polymorphism of the CYP2D6 gene on the efficacy and safety profiles of fluvoxamine was demonstrated in a group of 96 patients with depressive disorders comorbid with alcohol use disorder. Fluvoxamine 106-117 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 55-61 34117140-1 2021 BACKGROUND: Previous studies have shown that cytochrome P450 2D6 (CYP2D6) is involved in the metabolism of fluvoxamine, the activity of which is highly dependent, inter alia, on the polymorphism of the gene encoding it. Fluvoxamine 107-118 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 45-64 34117140-1 2021 BACKGROUND: Previous studies have shown that cytochrome P450 2D6 (CYP2D6) is involved in the metabolism of fluvoxamine, the activity of which is highly dependent, inter alia, on the polymorphism of the gene encoding it. Fluvoxamine 107-118 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 66-72 34117140-2 2021 The objective of our study was to investigate the effect of 1846G>A polymorphism of the CYP2D6 gene on the efficacy and safety of fluvoxamine, using findings on CYP2D6 enzymatic activity and on CYP2D6 expression level in patients with depressive disorders comorbid with alcohol use disorder. Fluvoxamine 130-141 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 88-94 34117140-2 2021 The objective of our study was to investigate the effect of 1846G>A polymorphism of the CYP2D6 gene on the efficacy and safety of fluvoxamine, using findings on CYP2D6 enzymatic activity and on CYP2D6 expression level in patients with depressive disorders comorbid with alcohol use disorder. Fluvoxamine 130-141 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 161-167 34117140-2 2021 The objective of our study was to investigate the effect of 1846G>A polymorphism of the CYP2D6 gene on the efficacy and safety of fluvoxamine, using findings on CYP2D6 enzymatic activity and on CYP2D6 expression level in patients with depressive disorders comorbid with alcohol use disorder. Fluvoxamine 130-141 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 194-200 34117140-3 2021 STUDY QUESTION: Efficacy and safety of fluvoxamine depend on the polymorphism of CYP2D6 gene in patients with major depressive disorder. Fluvoxamine 39-50 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 81-87 35611779-6 2022 The pharmacokinetics of many antiseizure medications is not influenced by antipsychotics and anxiolytics while some newer antidepressants namely fluoxetine, fluvoxamine and viloxazine, may inhibit CYP enzymes leading to increased serum concentrations of some antiseizure medications including phenytoin and carbamazepine. Fluvoxamine 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 34130375-3 2021 Thus, in this current computational study we carried out molecular docking experiments to assess the bridging potentials of some commercial drugs such as chloroquine, hydroxychloroquine, lopinavir, ritonavir, nafamostat, camostat, famotidine, umifenovir, nitazoxanide, ivermectin, and fluvoxamine at the interface between human ACE2 and the coronavirus spike glycoprotein complex. Fluvoxamine 285-296 angiotensin converting enzyme 2 Homo sapiens 328-332 35258103-6 2022 Finally, pharmacoepidemiological-pharmacodynamic analysis revealed a significant correlation between the decrease in serum sodium levels and binding affinity for serotonin transporter (SERT; r=-0.84, P=0.02), suggesting that lower binding affinity of mirtazapine, fluvoxamine, and milnacipran against SERT is responsible for the above difference. Fluvoxamine 264-275 solute carrier family 6 member 4 Homo sapiens 162-183 35258103-6 2022 Finally, pharmacoepidemiological-pharmacodynamic analysis revealed a significant correlation between the decrease in serum sodium levels and binding affinity for serotonin transporter (SERT; r=-0.84, P=0.02), suggesting that lower binding affinity of mirtazapine, fluvoxamine, and milnacipran against SERT is responsible for the above difference. Fluvoxamine 264-275 solute carrier family 6 member 4 Homo sapiens 185-189 35362343-0 2022 Fluvoxamine used to treat COVID-19 resulting in theophylline toxicity from CYP 1A2 drug-drug interaction. Fluvoxamine 0-11 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 75-82 35383578-1 2022 BACKGROUND: Recent clinical trials have investigated the use of fluvoxamine in preventing clinical deterioration in nonhospitalized patients with acute COVID-19 infection via stimulation of sigma-1 receptors, which regulates cytokine production and functional inhibition of acid sphingomyelinase activity, which may prevent infection of epithelial cells with SARS-CoV-2. Fluvoxamine 64-75 sphingomyelin phosphodiesterase 1 Homo sapiens 274-295 35599943-6 2022 Results: Fluvoxamine and mirtazapine significantly protected against WIRS and pyloric ligation-induced gastric lesions, as evidenced by a dose-dependent decrease in ulcer index, myeloperoxidase (MPO) activity, lipid peroxidation, and an increase in prostaglandin E2, nitric oxide (NO), and reduced glutathione levels, as well as increased antioxidant enzyme activity. Fluvoxamine 9-20 myeloperoxidase Rattus norvegicus 178-193 35599943-6 2022 Results: Fluvoxamine and mirtazapine significantly protected against WIRS and pyloric ligation-induced gastric lesions, as evidenced by a dose-dependent decrease in ulcer index, myeloperoxidase (MPO) activity, lipid peroxidation, and an increase in prostaglandin E2, nitric oxide (NO), and reduced glutathione levels, as well as increased antioxidant enzyme activity. Fluvoxamine 9-20 myeloperoxidase Rattus norvegicus 195-198 35473584-2 2022 Monocrotaline (60 mg/kg) was administered to induce pulmonary arterial hypertension, and fluvoxamine was given for 21 consecutive days to activate S1R after one week of monocrotaline administration. Fluvoxamine 89-100 sigma non-opioid intracellular receptor 1 Rattus norvegicus 147-150 34997196-5 2022 In this article, we review the mechanisms of action (i.e., serotonin transporter, sigma-1 receptor, and acid sphingomyelinase) of fluvoxamine for COVID-19. Fluvoxamine 130-141 solute carrier family 6 member 4 Homo sapiens 59-80 34997196-5 2022 In this article, we review the mechanisms of action (i.e., serotonin transporter, sigma-1 receptor, and acid sphingomyelinase) of fluvoxamine for COVID-19. Fluvoxamine 130-141 sphingomyelin phosphodiesterase 1 Homo sapiens 104-125 35327555-11 2022 Clinical trials demonstrated that several Sig-1R agonists (pridopidine, ANAVEX3-71, fluvoxamine, dextrometorphan) and their combinations have a neuroprotective effect and slow down the progression of distinct NDDs. Fluvoxamine 84-95 sigma non-opioid intracellular receptor 1 Homo sapiens 42-48