PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 18033153-2 2007 Atomoxetine is a highly specific inhibitor of the presynaptic norepinephrine transporter, with minimal affinity for other transporters or other neurotransmitter receptors. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 62-88 18352838-2 2008 Atomoxetine is a selective norepinephrine transporter (NET) inhibitor currently indicated for treatment of attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Rattus norvegicus 27-53 18352838-2 2008 Atomoxetine is a selective norepinephrine transporter (NET) inhibitor currently indicated for treatment of attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Rattus norvegicus 55-58 17610534-0 2007 Atomoxetine pharmacokinetics in healthy Chinese subjects and effect of the CYP2D6*10 allele. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 75-81 17224704-0 2007 Combined overdose of atomoxetine and methylphenidate in a cytochrome P450 2D6 poor metabolizer. Atomoxetine Hydrochloride 21-32 cytochrome P450 2D6 Homo sapiens 58-77 17242628-1 2007 BACKGROUND: Atomoxetine, a selective norepinephrine reuptake inhibitor effective in the treatment of attention-deficit/hyperactivity disorder (ADHD), is metabolized through the cytochrome P-450 2D6 (CYP2D6) enzyme pathway, which is genetically polymorphic in humans. Atomoxetine Hydrochloride 12-23 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 177-197 17242628-1 2007 BACKGROUND: Atomoxetine, a selective norepinephrine reuptake inhibitor effective in the treatment of attention-deficit/hyperactivity disorder (ADHD), is metabolized through the cytochrome P-450 2D6 (CYP2D6) enzyme pathway, which is genetically polymorphic in humans. Atomoxetine Hydrochloride 12-23 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 199-205 17242628-2 2007 Variations in plasma atomoxetine exposures can occur because of genetic variation or as a consequence of coadministration with drugs that inhibit CYP2D6. Atomoxetine Hydrochloride 21-32 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 146-152 17242628-3 2007 METHOD: We examined the effects of CYP2D6 on the efficacy, safety, and tolerability of atomoxetine in children and adolescents using pooled data from atomoxetine clinical trials. Atomoxetine Hydrochloride 87-98 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 35-41 17242628-7 2007 CONCLUSIONS: These results suggest that CYP2D6 poor metabolizers taking atomoxetine in doses up to 1.8 mg/kg/day are likely to have greater efficacy, greater increases in cardiovascular tone, and some differences in tolerability compared with CYP2D6 extensive metabolizers taking similar doses. Atomoxetine Hydrochloride 72-83 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 40-46 17242628-7 2007 CONCLUSIONS: These results suggest that CYP2D6 poor metabolizers taking atomoxetine in doses up to 1.8 mg/kg/day are likely to have greater efficacy, greater increases in cardiovascular tone, and some differences in tolerability compared with CYP2D6 extensive metabolizers taking similar doses. Atomoxetine Hydrochloride 72-83 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 243-249 17822337-4 2007 Mean decrease in ADHDRS-IV-Parent:Inv total score was significantly greater in the ATX group (-13.3 +/- 10.0) compared with the placebo group (-5.1 +/- 9.9; p < 0.001). Atomoxetine Hydrochloride 83-86 inversin Homo sapiens 34-37 17515448-0 2007 Norepinephrine transporter blockade with atomoxetine induces hypertension in patients with impaired autonomic function. Atomoxetine Hydrochloride 41-52 solute carrier family 6 member 2 Homo sapiens 0-26 17515448-1 2007 Atomoxetine, a selective norepinephrine transporter blocker, could increase blood pressure by elevating norepinephrine concentration in peripheral sympathetic neurons. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 25-51 16890481-1 2006 OBJECTIVE: To evaluate the use of P300 in predicting treatment response to medicines in patients with Attention-Deficit/Hyperactivity Disorder (ADHD), and to confirm previous reports that 31-electrode mean auditory P300 amplitude (AA) predicts response to atomoxetine; and right fronto-central to parietal AA ratio predicts response to methylphenidate. Atomoxetine Hydrochloride 256-267 E1A binding protein p300 Homo sapiens 215-219 17201614-2 2006 Atomoxetine, a potent inhibitor of the presynaptic norepinephrine transporter, is used to treat attention-deficit/hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 51-77 16890481-8 2006 31-electrode mean P300 visual latency (VL) also predicted response to atomoxetine, as previously reported with imipramine. Atomoxetine Hydrochloride 70-81 E1A binding protein p300 Homo sapiens 18-22 16896954-0 2006 Atomoxetine occupies the norepinephrine transporter in a dose-dependent fashion: a PET study in nonhuman primate brain using (S,S)-[18F]FMeNER-D2. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 25-51 16896954-1 2006 RATIONALE: Atomoxetine is a potent and selective norepinephrine transporter (NET) reuptake inhibitor acting as a nonstimulant for the treatment of attention-deficit/hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 11-22 solute carrier family 6 member 2 Homo sapiens 49-75 16896954-1 2006 RATIONALE: Atomoxetine is a potent and selective norepinephrine transporter (NET) reuptake inhibitor acting as a nonstimulant for the treatment of attention-deficit/hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 11-22 solute carrier family 6 member 2 Homo sapiens 77-80 16896954-3 2006 OBJECTIVES: To determine if atomoxetine occupies NET in a dose-dependent fashion using (S,S)-[18F]FMeNER-D2 in nonhuman primate brain. Atomoxetine Hydrochloride 28-39 solute carrier family 6 member 2 Homo sapiens 49-52 16896954-11 2006 After administration of atomoxetine, a dose-dependent occupancy from 38 to 82% was observed for various brain regions known to contain high densities of NET. Atomoxetine Hydrochloride 24-35 solute carrier family 6 member 2 Homo sapiens 153-156 15717291-3 2005 NET1 is implicated in ADHD because of the efficacy of atomoxetine, a selective noradrenergic reuptake inhibitor, in the treatment of ADHD. Atomoxetine Hydrochloride 54-65 neuroepithelial cell transforming gene 1 Mus musculus 0-4 16771600-9 2006 Venlafaxine, aripiprazole, duloxetine, and atomoxetine are newer drugs metabolized by CYP2D6 but studies of the clinical relevance of CYP2D6 genotypes are needed. Atomoxetine Hydrochloride 43-54 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 86-92 16771600-9 2006 Venlafaxine, aripiprazole, duloxetine, and atomoxetine are newer drugs metabolized by CYP2D6 but studies of the clinical relevance of CYP2D6 genotypes are needed. Atomoxetine Hydrochloride 43-54 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 134-140 16384813-9 2006 The next few years will determine whether CYP2D6 genotyping is beneficial for patients taking the new drugs aripiprazole, duloxetine, and atomoxetine. Atomoxetine Hydrochloride 138-149 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 42-48 16202140-1 2005 BACKGROUND: The objective of this study was to evaluate the efficacy of atomoxetine, a new and highly selective inhibitor of the norepinephrine transporter, in reducing symptoms of attention-deficit/hyperactivity disorder (ADHD) among adults by using drug-placebo response curve methods. Atomoxetine Hydrochloride 72-83 solute carrier family 6 member 2 Homo sapiens 129-155 16190797-1 2005 OBJECTIVE: The aim of this study was to assess the efficacy of atomoxetine, a new and highly selective inhibitor of the norepinephrine transporter, for executive functioning in adults with attention-deficit/hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 63-74 solute carrier family 6 member 2 Homo sapiens 120-146 15968233-5 2005 RESULTS: ADHDRS-IV-Teacher:Inv total scores were significantly lower for children treated with atomoxetine compared with those treated with placebo (p = .001). Atomoxetine Hydrochloride 95-106 inversin Homo sapiens 27-30 16142049-8 2005 Although pharmacokinetics of atomoxetine is influenced by the polymorphism of the CYP2D6 metabolic pathway, safety and -tolerability data reported during clinical trials did not show any difference in poor versus extensive metabolizers. Atomoxetine Hydrochloride 29-40 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 82-88 14709944-3 2004 Atomoxetine, a new drug approved in the United States for treatment of attention deficit/hyperactivity disorder (ADHD), is associated with blockade of the presynaptic norepinephrine transporter. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 167-193 15910008-1 2005 Atomoxetine (Strattera, a potent and selective inhibitor of the presynaptic norepinephrine transporter, is used clinically for the treatment of attention-deficit hyperactivity disorder (ADHD) in children, adolescents and adults. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 76-102 15910008-7 2005 This results in two distinct populations of individuals: those exhibiting active metabolic capabilities (CYP2D6 extensive metabolisers) and those exhibiting poor metabolic capabilities (CYP2D6 poor metabolisers) for atomoxetine. Atomoxetine Hydrochloride 216-227 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 186-192 15910008-8 2005 The oral bioavailability and clearance of atomoxetine are influenced by the activity of CYP2D6; nonetheless, plasma pharmacokinetic parameters are predictable in extensive and poor metaboliser patients. Atomoxetine Hydrochloride 42-53 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 88-94 15910008-17 2005 In extensive metabolisers, potent and selective CYP2D6 inhibitors reduce atomoxetine clearance; however, administration of CYP inhibitors to poor metabolisers has no effect on the steady-state plasma concentrations of atomoxetine. Atomoxetine Hydrochloride 73-84 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 48-54 16871320-3 2004 Some potent and selective or mixed NET inhibitors (e.g., reboxetine and atomoxetine) have been successfully developed to treat a variety of mental disorders such as depression and attention deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 72-83 solute carrier family 6 member 2 Homo sapiens 35-38 15581262-2 2004 SUMMARY: Atomoxetine is a methylphenoxy-benzenepropanamine derivative with antidepressant activity and is thought to enhance noradrenergic function via selective inhibition of the presynaptic norepinephrine transporter. Atomoxetine Hydrochloride 9-20 solute carrier family 6 member 2 Homo sapiens 192-218 14610241-1 2004 In the studies reported here, the ability of atomoxetine hydrochloride (Strattera) to inhibit or induce the metabolic capabilities of selected human isoforms of cytochrome P450 was evaluated. Atomoxetine Hydrochloride 45-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-176 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. Atomoxetine Hydrochloride 28-39 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 164-170 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. Atomoxetine Hydrochloride 28-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 14610241-3 2004 Although little inhibition of CYP1A2 and CYP2C9 activity was observed, inhibition was predicted for CYP3A (56% predicted inhibition) and CYP2D6 (60% predicted inhibition) at concentrations representative of high therapeutic doses of atomoxetine. Atomoxetine Hydrochloride 233-244 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 100-105 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. Atomoxetine Hydrochloride 28-39 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 180-186 14610241-3 2004 Although little inhibition of CYP1A2 and CYP2C9 activity was observed, inhibition was predicted for CYP3A (56% predicted inhibition) and CYP2D6 (60% predicted inhibition) at concentrations representative of high therapeutic doses of atomoxetine. Atomoxetine Hydrochloride 233-244 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 137-143 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. Atomoxetine Hydrochloride 28-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 192-197 14610241-4 2004 The ability of atomoxetine to induce the catalytic activities of CYP1A2 and CYP3A in human hepatocytes was also evaluated; however, atomoxetine did not induce either isoenzyme. Atomoxetine Hydrochloride 15-26 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 65-71 14610241-4 2004 The ability of atomoxetine to induce the catalytic activities of CYP1A2 and CYP3A in human hepatocytes was also evaluated; however, atomoxetine did not induce either isoenzyme. Atomoxetine Hydrochloride 15-26 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-81 12547466-2 2003 To assess the efficacy of atomoxetine, a new and highly selective inhibitor of the norepinephrine transporter, we conducted two large, multicenter treatment trials. Atomoxetine Hydrochloride 26-37 solute carrier family 6 member 2 Homo sapiens 83-109 14610241-10 2004 Although at high therapeutic doses of atomoxetine inhibition of CYP2D6 and CYP3A was predicted, definitive in vivo studies clearly indicate that atomoxetine administration with substrates of CYP2D6 and CYP3A does not result in clinically significant drug interactions. Atomoxetine Hydrochloride 38-49 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 64-70 14610241-10 2004 Although at high therapeutic doses of atomoxetine inhibition of CYP2D6 and CYP3A was predicted, definitive in vivo studies clearly indicate that atomoxetine administration with substrates of CYP2D6 and CYP3A does not result in clinically significant drug interactions. Atomoxetine Hydrochloride 38-49 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 75-80 14610241-10 2004 Although at high therapeutic doses of atomoxetine inhibition of CYP2D6 and CYP3A was predicted, definitive in vivo studies clearly indicate that atomoxetine administration with substrates of CYP2D6 and CYP3A does not result in clinically significant drug interactions. Atomoxetine Hydrochloride 145-156 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 191-197 14681910-2 2004 The mechanism of action of the new selective noradrenergic reuptake inhibitor, atomoxetine, efficacious in the treatment of ADHD symptoms, suggests that the norepinephrine transporter (NET) may be involved in ADHD. Atomoxetine Hydrochloride 79-90 solute carrier family 6 member 2 Homo sapiens 157-183 14681910-2 2004 The mechanism of action of the new selective noradrenergic reuptake inhibitor, atomoxetine, efficacious in the treatment of ADHD symptoms, suggests that the norepinephrine transporter (NET) may be involved in ADHD. Atomoxetine Hydrochloride 79-90 solute carrier family 6 member 2 Homo sapiens 185-188 12621383-1 2003 BACKGROUND AND OBJECTIVES: Atomoxetine is a treatment for attention-deficit/hyperactivity disorder and is primarily eliminated via cytochrome P4502D6 (CYP2D6). Atomoxetine Hydrochloride 27-38 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 131-149 12621383-1 2003 BACKGROUND AND OBJECTIVES: Atomoxetine is a treatment for attention-deficit/hyperactivity disorder and is primarily eliminated via cytochrome P4502D6 (CYP2D6). Atomoxetine Hydrochloride 27-38 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 151-157 12621383-13 2003 Decreased atomoxetine clearance in patients with hepatic impairment was clearly correlated with decreased CYP2D6 activity and decreased hepatic blood flow. Atomoxetine Hydrochloride 10-21 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 106-112 12485958-1 2003 The role of the polymorphic cytochrome p450 2D6 (CYP2D6) in the pharmacokinetics of atomoxetine hydrochloride [(-)-N-methyl-gamma-(2-methylphenoxy)benzenepropanamine hydrochloride; LY139603] has been documented following both single and multiple doses of the drug. Atomoxetine Hydrochloride 181-189 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 49-55 34811360-1 2021 Many antidepressants, atomoxetine, and several antipsychotics are metabolized by the cytochrome P450 enzymes CYP2D6 and CYP2C19, and guidelines for prescribers based on genetic variants exist. Atomoxetine Hydrochloride 22-33 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 109-115 12412820-0 2002 Effect of potent CYP2D6 inhibition by paroxetine on atomoxetine pharmacokinetics. Atomoxetine Hydrochloride 52-63 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 17-23 12412820-1 2002 The purpose of this study was to characterize the effect of potent CYP2D6 inhibition byparoxetine on atomoxetine disposition in extensive metabolizers. Atomoxetine Hydrochloride 101-112 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 67-73 12412820-3 2002 In period 1, 20 mg atomoxetine bid was administered to steady state. Atomoxetine Hydrochloride 19-30 BH3 interacting domain death agonist Homo sapiens 31-34 12412820-11 2002 It was concluded that inhibition of CYP2D6 by paroxetine markedly affected atomoxetine disposition, resulting in pharmacokinetics similar to poor metabolizers of CYP2D6 substrates. Atomoxetine Hydrochloride 75-86 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 36-42 11854152-10 2002 Utilizing studies similar to those outlined above, CYP2C19 was identified as the primary enzyme responsible for the biotransformation of atomoxetine to N-desmethylatomoxetine. Atomoxetine Hydrochloride 137-148 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 51-58 11854152-11 2002 In summary, CYP2D6 was found to be the primary P450 responsible for the formation of the major oxidative metabolite of atomoxetine, 4-hydroxyatomoxetine. Atomoxetine Hydrochloride 119-130 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 12-18 34958348-4 2022 Atomoxetine is a NET inhibitor but also has some affinity for the serotonin reuptake transporter (SERT). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 4 Homo sapiens 66-96 34958348-4 2022 Atomoxetine is a NET inhibitor but also has some affinity for the serotonin reuptake transporter (SERT). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 4 Homo sapiens 98-102 34919634-16 2021 Atomoxetine treatment also significantly altered CSF abundances of protein panels linked to brain pathophysiologies, including synaptic, metabolism, and glial immunity, as well as inflammation-related CDCP1, CD244, TWEAK, and OPG proteins. Atomoxetine Hydrochloride 0-11 basic transcription factor 3 pseudogene 11 Homo sapiens 226-229 34919634-21 2021 Atomoxetine significantly reduced CSF Tau and pTau, normalized CSF protein biomarker panels linked to synaptic function, brain metabolism, and glial immunity, and increased brain activity and metabolism in key temporal lobe circuits. Atomoxetine Hydrochloride 0-11 microtubule associated protein tau Homo sapiens 38-41 12485958-0 2003 Disposition and metabolic fate of atomoxetine hydrochloride: the role of CYP2D6 in human disposition and metabolism. Atomoxetine Hydrochloride 34-59 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 73-79 12485958-1 2003 The role of the polymorphic cytochrome p450 2D6 (CYP2D6) in the pharmacokinetics of atomoxetine hydrochloride [(-)-N-methyl-gamma-(2-methylphenoxy)benzenepropanamine hydrochloride; LY139603] has been documented following both single and multiple doses of the drug. Atomoxetine Hydrochloride 84-109 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 28-47 12485958-1 2003 The role of the polymorphic cytochrome p450 2D6 (CYP2D6) in the pharmacokinetics of atomoxetine hydrochloride [(-)-N-methyl-gamma-(2-methylphenoxy)benzenepropanamine hydrochloride; LY139603] has been documented following both single and multiple doses of the drug. Atomoxetine Hydrochloride 84-109 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 49-55 12485958-1 2003 The role of the polymorphic cytochrome p450 2D6 (CYP2D6) in the pharmacokinetics of atomoxetine hydrochloride [(-)-N-methyl-gamma-(2-methylphenoxy)benzenepropanamine hydrochloride; LY139603] has been documented following both single and multiple doses of the drug. Atomoxetine Hydrochloride 111-179 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 28-47 12485958-1 2003 The role of the polymorphic cytochrome p450 2D6 (CYP2D6) in the pharmacokinetics of atomoxetine hydrochloride [(-)-N-methyl-gamma-(2-methylphenoxy)benzenepropanamine hydrochloride; LY139603] has been documented following both single and multiple doses of the drug. Atomoxetine Hydrochloride 111-179 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 49-55 12431845-2 2002 We investigated the mechanism of action of atomoxetine in ADHD by evaluating the interaction of atomoxetine with monoamine transporters, the effects on extracellular levels of monoamines, and the expression of the neuronal activity marker Fos in brain regions. Atomoxetine Hydrochloride 43-54 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 239-242 12431845-7 2002 The expression of the neuronal activity marker Fos was increased 3.7-fold in PFC by atomoxetine administration, but was not increased in the striatum or nucleus accumbens, consistent with the regional distribution of increased DA(EX). Atomoxetine Hydrochloride 84-95 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 47-50 7562595-6 1995 Pretreatment with the alpha-1 adrenoceptor blocker prazosin antagonized the DS effects of cocaine under both training conditions as well as the cocaine-like effects of talsupram and tomoxetine, but not GBR 12909, under the low-dose training condition. Atomoxetine Hydrochloride 182-192 adrenoceptor alpha 1D Homo sapiens 22-29 33944622-0 2021 Role of the norepinephrine transporter polymorphisms in atomoxetine treatment: From response to side effects in children with ADHD. Atomoxetine Hydrochloride 56-67 solute carrier family 6 member 2 Homo sapiens 12-38 33944622-1 2021 OBJECTIVE: Atomoxetine (ATX), one of the most commonly used drugs after stimulants in attention deficit hyperactivity disorder (ADHD) treatment, is an inhibitor of the norepinephrine transporter (NET/SLC6A2), which is also associated with the etiology of ADHD. Atomoxetine Hydrochloride 11-22 solute carrier family 6 member 2 Homo sapiens 168-194 33944622-1 2021 OBJECTIVE: Atomoxetine (ATX), one of the most commonly used drugs after stimulants in attention deficit hyperactivity disorder (ADHD) treatment, is an inhibitor of the norepinephrine transporter (NET/SLC6A2), which is also associated with the etiology of ADHD. Atomoxetine Hydrochloride 11-22 solute carrier family 6 member 2 Homo sapiens 200-206 33944622-1 2021 OBJECTIVE: Atomoxetine (ATX), one of the most commonly used drugs after stimulants in attention deficit hyperactivity disorder (ADHD) treatment, is an inhibitor of the norepinephrine transporter (NET/SLC6A2), which is also associated with the etiology of ADHD. Atomoxetine Hydrochloride 24-27 solute carrier family 6 member 2 Homo sapiens 168-194 33944622-1 2021 OBJECTIVE: Atomoxetine (ATX), one of the most commonly used drugs after stimulants in attention deficit hyperactivity disorder (ADHD) treatment, is an inhibitor of the norepinephrine transporter (NET/SLC6A2), which is also associated with the etiology of ADHD. Atomoxetine Hydrochloride 24-27 solute carrier family 6 member 2 Homo sapiens 200-206 34919634-15 2021 Atomoxetine was associated with a significant reduction in CSF Tau and pTau181 compared to placebo, but not associated with change in Abeta42. Atomoxetine Hydrochloride 0-11 microtubule associated protein tau Homo sapiens 63-66 34919634-16 2021 Atomoxetine treatment also significantly altered CSF abundances of protein panels linked to brain pathophysiologies, including synaptic, metabolism, and glial immunity, as well as inflammation-related CDCP1, CD244, TWEAK, and OPG proteins. Atomoxetine Hydrochloride 0-11 CUB domain containing protein 1 Homo sapiens 201-206 34919634-16 2021 Atomoxetine treatment also significantly altered CSF abundances of protein panels linked to brain pathophysiologies, including synaptic, metabolism, and glial immunity, as well as inflammation-related CDCP1, CD244, TWEAK, and OPG proteins. Atomoxetine Hydrochloride 0-11 CD244 molecule Homo sapiens 208-213 34919634-16 2021 Atomoxetine treatment also significantly altered CSF abundances of protein panels linked to brain pathophysiologies, including synaptic, metabolism, and glial immunity, as well as inflammation-related CDCP1, CD244, TWEAK, and OPG proteins. Atomoxetine Hydrochloride 0-11 TNF superfamily member 12 Homo sapiens 215-220 34811360-1 2021 Many antidepressants, atomoxetine, and several antipsychotics are metabolized by the cytochrome P450 enzymes CYP2D6 and CYP2C19, and guidelines for prescribers based on genetic variants exist. Atomoxetine Hydrochloride 22-33 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 120-127 32878260-0 2020 Digital Light Processing (DLP) 3D Printing of Atomoxetine Hydrochloride Tablets Using Photoreactive Suspensions. Atomoxetine Hydrochloride 46-71 dynein axonemal heavy chain 3 Homo sapiens 0-32 34176285-0 2021 Predictors of the Pressor Response to the Norepinephrine Transporter Inhibitor, Atomoxetine, in Neurogenic Orthostatic Hypotension. Atomoxetine Hydrochloride 80-91 solute carrier family 6 member 2 Homo sapiens 42-68 35222104-4 2021 Atomoxetine inhibits the presynaptic norepinephrine transporter (NET), preventing the reuptake of NE throughout the brain along with inhibiting the reuptake of dopamine in specific brain regions such as the prefrontal cortex (PFC). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 37-63 35222104-12 2021 The latest guideline updated that clinical dose selection of atomoxetine was recommended based on both CYP2D6 genotype and the peak concentration. Atomoxetine Hydrochloride 61-72 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 103-109 35043773-5 2022 OBJECTIVE: The objective of this work was to test the hypothesis that atomoxetine saliva levels (sATX) can be used to predict ATX brain extracellular fluid (bECF) levels and their pharmacological effects in healthy subjects and those with end-stage renal disease (ESRD). Atomoxetine Hydrochloride 70-81 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 126-129 33732157-9 2021 Atomoxetine had only a weak affinity for the resting state of the hNav1.5 (Kr: ~ 120 microM). Atomoxetine Hydrochloride 0-11 sodium voltage-gated channel alpha subunit 5 Homo sapiens 66-73 33245517-8 2020 After paroxetine treatment, AUC0-24 of atomoxetine was increased by 2.3-, 1.7-, and 1.3-fold, in CYP2D6*wt/*wt, CYP2D6*wt/*10, and CYP2D6*10/*10 groups in comparison to atomoxetine phase, respectively. Atomoxetine Hydrochloride 39-50 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 97-103 33245517-8 2020 After paroxetine treatment, AUC0-24 of atomoxetine was increased by 2.3-, 1.7-, and 1.3-fold, in CYP2D6*wt/*wt, CYP2D6*wt/*10, and CYP2D6*10/*10 groups in comparison to atomoxetine phase, respectively. Atomoxetine Hydrochloride 39-50 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 112-118 33245517-8 2020 After paroxetine treatment, AUC0-24 of atomoxetine was increased by 2.3-, 1.7-, and 1.3-fold, in CYP2D6*wt/*wt, CYP2D6*wt/*10, and CYP2D6*10/*10 groups in comparison to atomoxetine phase, respectively. Atomoxetine Hydrochloride 39-50 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 112-118 33245517-8 2020 After paroxetine treatment, AUC0-24 of atomoxetine was increased by 2.3-, 1.7-, and 1.3-fold, in CYP2D6*wt/*wt, CYP2D6*wt/*10, and CYP2D6*10/*10 groups in comparison to atomoxetine phase, respectively. Atomoxetine Hydrochloride 169-180 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 112-118 33245517-8 2020 After paroxetine treatment, AUC0-24 of atomoxetine was increased by 2.3-, 1.7-, and 1.3-fold, in CYP2D6*wt/*wt, CYP2D6*wt/*10, and CYP2D6*10/*10 groups in comparison to atomoxetine phase, respectively. Atomoxetine Hydrochloride 169-180 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 112-118 33245517-11 2020 When atomoxetine was administered alone, Cmax, AUC0-24 and CL/F of atomoxetine were significantly different among the three CYP2D6 genotype groups. Atomoxetine Hydrochloride 5-16 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 124-130 34716917-2 2022 As there are limited and inconsistent data regarding the utility of this distant "enhancer" single nucleotide polymorphism (SNP), our goal was to further assess the impact of rs5758550 on CYP2D6 activity toward two probe substrates, atomoxetine (ATX) and dextromethorphan (DM), using in vivo urinary metabolite (DM; n=188) and pharmacokinetic (ATX; n=70) and in vitro metabolite formation (ATX and DM; n=166) data. Atomoxetine Hydrochloride 233-244 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 188-194 34716917-2 2022 As there are limited and inconsistent data regarding the utility of this distant "enhancer" single nucleotide polymorphism (SNP), our goal was to further assess the impact of rs5758550 on CYP2D6 activity toward two probe substrates, atomoxetine (ATX) and dextromethorphan (DM), using in vivo urinary metabolite (DM; n=188) and pharmacokinetic (ATX; n=70) and in vitro metabolite formation (ATX and DM; n=166) data. Atomoxetine Hydrochloride 246-249 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 188-194 34679865-0 2021 Salivary Chromogranin A (CgA) Response to the Noradrenaline Transporter Blocker Atomoxetine in Dogs. Atomoxetine Hydrochloride 80-91 chromogranin-A Canis lupus familiaris 9-23 34679865-0 2021 Salivary Chromogranin A (CgA) Response to the Noradrenaline Transporter Blocker Atomoxetine in Dogs. Atomoxetine Hydrochloride 80-91 chromogranin-A Canis lupus familiaris 25-28 34679865-3 2021 A selective noradrenaline transporter blocker, atomoxetine, was orally administered without causing any aversive responses in nine laboratory dogs to see if it would increase salivary CgA. Atomoxetine Hydrochloride 47-58 chromogranin-A Canis lupus familiaris 184-187 34679865-6 2021 The results demonstrated that salivary CgA significantly increased at 90 min in the atomoxetine treatment (p < 0.05), whereas it was not observed in the other two treatments. Atomoxetine Hydrochloride 84-95 chromogranin-A Canis lupus familiaris 39-42 34679865-7 2021 The present study showed that salivary CgA was increased by atomoxetine-induced SAM activation. Atomoxetine Hydrochloride 60-71 chromogranin-A Canis lupus familiaris 39-42 34176285-1 2021 We previously reported that the norepinephrine transporter inhibitor, atomoxetine, improved standing blood pressure and lightheadedness in patients with neurogenic orthostatic hypotension (nOH). Atomoxetine Hydrochloride 70-81 solute carrier family 6 member 2 Homo sapiens 32-58 35486119-10 2022 Also, BDNF has a stronger predictive value for assessing resistance to ATX treatment. Atomoxetine Hydrochloride 71-74 brain derived neurotrophic factor Homo sapiens 6-10 35486119-11 2022 CONCLUSIONS: To our knowledge, this is the first study to assess the effects of CYP2C19 polymorphisms and BDNF levels together on ATX treatment in children. Atomoxetine Hydrochloride 130-133 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 80-87 35486119-11 2022 CONCLUSIONS: To our knowledge, this is the first study to assess the effects of CYP2C19 polymorphisms and BDNF levels together on ATX treatment in children. Atomoxetine Hydrochloride 130-133 brain derived neurotrophic factor Homo sapiens 106-110 35204932-5 2022 Still, the short-term effects of atomoxetine treatment on serum growth parameters, such as IGF-1, IGFBP-3, and thyroid function, are not well documented. Atomoxetine Hydrochloride 33-44 insulin like growth factor 1 Homo sapiens 91-96 35204932-5 2022 Still, the short-term effects of atomoxetine treatment on serum growth parameters, such as IGF-1, IGFBP-3, and thyroid function, are not well documented. Atomoxetine Hydrochloride 33-44 insulin like growth factor binding protein 3 Homo sapiens 98-105 33998618-1 2021 The Clinical Pharmacogenetic Implementation Consortium (CPIC) guidelines for personalized atomoxetine therapy are based on the CYP2D6 genotype information and the peak plasma concentrations of atomoxetine. Atomoxetine Hydrochloride 90-101 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 127-133 33143874-0 2021 Alternations in nuclear factor kappa beta activity (NF-kB) in the rat brain due to long-term use of atomoxetine for treating ADHD: In vivo and in silico studies. Atomoxetine Hydrochloride 100-111 nuclear factor kappa B subunit 1 Rattus norvegicus 52-57 33245517-0 2020 Effects of paroxetine on the pharmacokinetics of atomoxetine and its metabolites in different CYP2D6 genotypes. Atomoxetine Hydrochloride 49-60 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 94-100 33245517-1 2020 The aim of this study was to investigate the effects of paroxetine, a potent inhibitor of CYP2D6, on the pharmacokinetics of atomoxetine and its two metabolites, 4-hydroxyatomoxetine and N-desmethylatomoxetine, in different CYP2D6 genotypes. Atomoxetine Hydrochloride 126-137 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 91-97 32712217-1 2020 Atomoxetine (ATX), a selective and potent inhibitor of the presynaptic norepinephrine transporter, is used mainly to treat attention-deficit hyperactivity disorder. Atomoxetine Hydrochloride 0-11 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 71-97 32712217-1 2020 Atomoxetine (ATX), a selective and potent inhibitor of the presynaptic norepinephrine transporter, is used mainly to treat attention-deficit hyperactivity disorder. Atomoxetine Hydrochloride 13-16 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 71-97 28520366-0 2012 Atomoxetine Therapy and CYP2D6 Genotype Atomoxetine was the first non-stimulant drug to be used in the treatment of attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 24-30 32151742-3 2020 OBJECTIVE: The objective of this systematic review was to evaluate the ability of three NET inhibitors - reboxetine, sibutramine, and atomoxetine - to prevent head-up-tilt-induced vasovagal outcomes in healthy participants and patients with VVS. Atomoxetine Hydrochloride 134-145 solute carrier family 6 member 2 Homo sapiens 88-91 28520366-2 2012 The CYP2D6 enzyme metabolizes a quarter of all prescribed drugs, including atomoxetine. Atomoxetine Hydrochloride 75-86 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 4-10 28520366-3 2012 Individuals who carry two nonfunctional copies of the CYP2D6 gene are known as poor metabolizers and have higher plasma concentrations of atomoxetine compared with individuals who have two copies of normal activity alleles. Atomoxetine Hydrochloride 138-149 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 54-60 28520366-4 2012 The FDA states that the dose of atomoxetine may need to be adjusted in patients known to be CYP2D6 poor metabolizers (1). Atomoxetine Hydrochloride 32-43 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 92-98 32184039-1 2020 Atomoxetine is an approved medicine for attention-deficit/hyperactivity disorder and a cytochrome P450 2D6 (CYP2D6) probe substrate. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 87-106 32184039-1 2020 Atomoxetine is an approved medicine for attention-deficit/hyperactivity disorder and a cytochrome P450 2D6 (CYP2D6) probe substrate. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 108-114 32184039-4 2020 However, 5 participants (with impaired CYP2D6 activity scores based on the CYP2D6 genotypes) showed high plasma concentrations of atomoxetine (0.53-1.5 muM) compared with those of total 4-hydroxyatomoxetine (0.49-1.4 muM). Atomoxetine Hydrochloride 130-141 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 39-45 32184039-4 2020 However, 5 participants (with impaired CYP2D6 activity scores based on the CYP2D6 genotypes) showed high plasma concentrations of atomoxetine (0.53-1.5 muM) compared with those of total 4-hydroxyatomoxetine (0.49-1.4 muM). Atomoxetine Hydrochloride 130-141 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 75-81 31538180-3 2019 Atomoxetine is a potent and highly selective NET inhibitor. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 45-48 31506926-8 2020 Co-treatment with the beta2 -adrenoceptor antagonist ICI 118,551 attenuated the protective effects of atomoxetine. Atomoxetine Hydrochloride 102-113 adrenoceptor beta 2 Rattus norvegicus 22-41 30125623-0 2019 The Atxn7-overexpressing mice showed hyperactivity and impulsivity which were ameliorated by atomoxetine treatment: A possible animal model of the hyperactive-impulsive phenotype of ADHD. Atomoxetine Hydrochloride 93-104 ataxin 7 Mus musculus 4-9 30460884-10 2019 Biological effects of atomoxetine (k=1) and amphetamines (k=1) were cortical activation, without change in beta-endorphin (k=1), improved response to antipsychotics after amphetamine challenge (k=2), and an increase of growth hormone-mediated psychosis with methylphenidate (k=2). Atomoxetine Hydrochloride 22-33 growth hormone 1 Homo sapiens 219-233 30801677-2 2019 Cytochrome P450 (CYP)2D6 polymorphisms influence the metabolism of atomoxetine thereby affecting drug efficacy and safety. Atomoxetine Hydrochloride 67-78 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-24 30801677-3 2019 We summarize evidence from the published literature supporting these associations and provide therapeutic recommendations for atomoxetine based on CYP2D6 genotype (updates at www.cpicpgx.org). Atomoxetine Hydrochloride 126-137 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 147-153 30692908-2 2018 The role of this catecholamine system is also highlighted by the selective norepinephrine transporter blocking atomoxetine, which has proved to be effective in the pharmacological treatment of Attention Deficit Hyperactivity Disorder (ADHD). Atomoxetine Hydrochloride 111-122 solute carrier family 6 member 2 Homo sapiens 75-101 30125623-8 2019 Interestingly, treatment with the ADHD drug, atomoxetine (3 mg/kg, intraperitoneal), attenuated ADHD-like behaviors and reduced Atxn7 gene expression in the PFC and STR of these mice. Atomoxetine Hydrochloride 45-56 ataxin 7 Mus musculus 128-133 30037827-8 2018 The spatial structure of both fluctuation patterns resembled the spatial distribution of the expression of catecholamine receptor genes: alpha1 norepinephrine receptors (for the fluctuation pattern: placebo > atomoxetine), D2-like dopamine receptors (pattern: atomoxetine > placebo), and beta norepinephrine receptors (for both patterns, with correlations of opposite sign). Atomoxetine Hydrochloride 209-220 adrenoceptor beta 2 Homo sapiens 107-129 29608930-18 2018 In addition, atomoxetine treatment was associated with changes of TUNEL, pCREB, and BDNF expression levels, and microglial numbers, morphology, and responses. Atomoxetine Hydrochloride 13-24 brain-derived neurotrophic factor Rattus norvegicus 84-88 30571543-2 2019 For example, norepinephrine transporter blockade with atomoxetine raises blood pressure (BP) in autonomic failure patients by increasing synaptic norepinephrine concentrations; acetylcholinesterase inhibition with pyridostigmine increases BP by facilitating ganglionic cholinergic neurotransmission to increase sympathetic outflow. Atomoxetine Hydrochloride 54-65 solute carrier family 6 member 2 Homo sapiens 13-39 33562949-2 2018 Simple potentiometric carbon paste electrodes (CPEs) based on atomoxetine-derivatized with tetraphenylborate (ATM-TPB) or phosphotungstic acid (ATM-PTA) as ion-pairs decorated with TiO2 nanoparticles and sodium tetraphenylborate (Na-TPB) as additives were most useful. Atomoxetine Hydrochloride 62-73 ATM serine/threonine kinase Homo sapiens 110-113 30037827-8 2018 The spatial structure of both fluctuation patterns resembled the spatial distribution of the expression of catecholamine receptor genes: alpha1 norepinephrine receptors (for the fluctuation pattern: placebo > atomoxetine), D2-like dopamine receptors (pattern: atomoxetine > placebo), and beta norepinephrine receptors (for both patterns, with correlations of opposite sign). Atomoxetine Hydrochloride 260-271 adrenoceptor beta 2 Homo sapiens 107-129 28416812-8 2018 Furthermore, hyperactive-impulsive-like behavior was induced by reducing the expression of the zebrafish gene that is homologous to MICALL2, which could be rescued by tomoxetine (atomoxetine), a clinical medication for ADHD. Atomoxetine Hydrochloride 167-177 mical-like 2a Danio rerio 132-139 29573648-3 2018 This study evaluated the safety, tolerability, and potential efficacy of atomoxetine for treating ATS use disorder. Atomoxetine Hydrochloride 73-84 solute carrier family 2 member 10 Homo sapiens 98-101 29573648-7 2018 The proportion of ATS-negative urine tests was higher in atomoxetine- compared to placebo-treated participants: 0.77 (0.63-0.91) vs. 0.67 (0.53-0.81, d = 0.26) in the ITT sample and 0.90 (0.75-1.00) vs. 0.64 (0.51-0.78, d = 0.56) in the higher adherence subsample. Atomoxetine Hydrochloride 57-68 solute carrier family 2 member 10 Homo sapiens 18-21 29573648-11 2018 CONCLUSIONS: The findings support clinical tolerability and safety and suggest potential efficacy of atomoxetine for treating ATS use disorder in this population. Atomoxetine Hydrochloride 101-112 solute carrier family 2 member 10 Homo sapiens 126-129 30120390-0 2018 Physiologically based pharmacokinetic modelling of atomoxetine with regard to CYP2D6 genotypes. Atomoxetine Hydrochloride 51-62 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 78-84 30120390-3 2018 Clinical trials have shown that decreased CYP2D6 activity leads to substantially elevated atomoxetine exposure and increase in adverse reactions. Atomoxetine Hydrochloride 90-101 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 42-48 30120390-4 2018 The aim of this study was to to develop a pharmacologically based pharmacokinetic (PBPK) model of atomoxetine in different CYP2D6 genotypes. Atomoxetine Hydrochloride 98-109 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 123-129 30120390-5 2018 A single 20 mg dose of atomoxetine was given to 19 healthy Korean individuals with CYP2D6*wt/*wt (*wt = *1 or *2) or CYP2D6*10/*10 genotype. Atomoxetine Hydrochloride 23-34 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 83-89 30120390-5 2018 A single 20 mg dose of atomoxetine was given to 19 healthy Korean individuals with CYP2D6*wt/*wt (*wt = *1 or *2) or CYP2D6*10/*10 genotype. Atomoxetine Hydrochloride 23-34 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 117-123 30120390-9 2018 The presented PBPK model describes the pharmacokinetics after single and repeated oral atomoxetine doses with regard to CYP2D6 genotype and phenotype. Atomoxetine Hydrochloride 87-98 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 120-126 30120390-10 2018 This model could be utilized for identification of appropriate dosages of atomoxetine in patients with reduced CYP2D6 activity to minimize the adverse events, and to enable personalised medicine. Atomoxetine Hydrochloride 74-85 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 111-117 28416812-8 2018 Furthermore, hyperactive-impulsive-like behavior was induced by reducing the expression of the zebrafish gene that is homologous to MICALL2, which could be rescued by tomoxetine (atomoxetine), a clinical medication for ADHD. Atomoxetine Hydrochloride 179-190 mical-like 2a Danio rerio 132-139 29339319-4 2018 Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-alpha whilst increasing the expression of neurotrophic factors. Atomoxetine Hydrochloride 15-26 tumor necrosis factor Rattus norvegicus 206-215 29203337-0 2018 Shati/Nat8l knockout mice show behavioral deficits ameliorated by atomoxetine and methylphenidate. Atomoxetine Hydrochloride 66-77 N-acetyltransferase 8-like Mus musculus 0-5 29203337-0 2018 Shati/Nat8l knockout mice show behavioral deficits ameliorated by atomoxetine and methylphenidate. Atomoxetine Hydrochloride 66-77 N-acetyltransferase 8-like Mus musculus 6-11 28509573-2 2017 We describe the rate and duration of adverse events in a randomized controlled trial of atomoxetine (ATX) and parent training (PT) for ADHD symptoms and noncompliance in children with ASD. Atomoxetine Hydrochloride 88-99 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 101-104 28860113-0 2017 Physiologically Based Pharmacokinetic Model of the CYP2D6 Probe Atomoxetine: Extrapolation to Special Populations and Drug-Drug Interactions. Atomoxetine Hydrochloride 64-75 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 51-57 28860113-5 2017 A full PBPK model of atomoxetine was developed using a training set of pharmacokinetic (PK) data from CYP2D6 genotyped individuals. Atomoxetine Hydrochloride 21-32 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 102-108 27857045-0 2016 Long term pharmacotherapy by methylfenidate or atomoxetine DAT 1 10/10 ADHD children in correlation with results of the imaging methods. Atomoxetine Hydrochloride 47-58 solute carrier family 6 member 3 Homo sapiens 59-64 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Atomoxetine Hydrochloride 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Atomoxetine Hydrochloride 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-98 28232237-3 2017 Atomoxetine administration significantly increased SAA secretion and concentrations at 75-180min after treatment (more than doubling baseline levels). Atomoxetine Hydrochloride 0-11 amylase alpha 1A Homo sapiens 51-54 28236285-22 2017 CONCLUSIONS: Statistically significant pre-post increases of SBP, DBP and HR were associated with AMP and ATX treatment in children and adolescents with ADHD, while MPH treatment had a statistically significant effect only on SBP in these patients. Atomoxetine Hydrochloride 106-109 selenium binding protein 1 Homo sapiens 61-64 28236285-22 2017 CONCLUSIONS: Statistically significant pre-post increases of SBP, DBP and HR were associated with AMP and ATX treatment in children and adolescents with ADHD, while MPH treatment had a statistically significant effect only on SBP in these patients. Atomoxetine Hydrochloride 106-109 D-box binding PAR bZIP transcription factor Homo sapiens 66-69 27755221-0 2016 Dihydroxyphenylglycol as a Biomarker of Norepinephrine Transporter Inhibition by Atomoxetine: Human Model to Assess Central and Peripheral Effects of Dosing. Atomoxetine Hydrochloride 81-92 solute carrier family 6 member 2 Homo sapiens 40-66 28744604-6 2017 Treatment with methylphenidate and/or atomoxetine increased choice of the large, delayed reward in SHR/NCrl and Wistar rats and changed, in varying degrees, mRNA levels of Nr4a2, Btg2, and Homer2, genes with previously described roles in neuropsychiatric disorders characterized by impulsivity. Atomoxetine Hydrochloride 38-49 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 172-177 28744604-6 2017 Treatment with methylphenidate and/or atomoxetine increased choice of the large, delayed reward in SHR/NCrl and Wistar rats and changed, in varying degrees, mRNA levels of Nr4a2, Btg2, and Homer2, genes with previously described roles in neuropsychiatric disorders characterized by impulsivity. Atomoxetine Hydrochloride 38-49 BTG anti-proliferation factor 2 Rattus norvegicus 179-183 28744604-6 2017 Treatment with methylphenidate and/or atomoxetine increased choice of the large, delayed reward in SHR/NCrl and Wistar rats and changed, in varying degrees, mRNA levels of Nr4a2, Btg2, and Homer2, genes with previously described roles in neuropsychiatric disorders characterized by impulsivity. Atomoxetine Hydrochloride 38-49 homer scaffold protein 2 Rattus norvegicus 189-195 29404031-0 2017 Multi-spectroscopic characterization of bovine serum albumin upon interaction with atomoxetine. Atomoxetine Hydrochloride 83-94 albumin Homo sapiens 47-60 29404031-1 2017 The quenching interaction of atomoxetine (ATX) with bovine serum albumin (BSA) was studied in vitro under optimal physiological condition (pH=7.4) by multi-spectroscopic techniques. Atomoxetine Hydrochloride 29-40 albumin Homo sapiens 59-72 29404031-1 2017 The quenching interaction of atomoxetine (ATX) with bovine serum albumin (BSA) was studied in vitro under optimal physiological condition (pH=7.4) by multi-spectroscopic techniques. Atomoxetine Hydrochloride 42-45 albumin Homo sapiens 59-72 27673638-0 2016 Drug-Drug Interaction of Paroxetine and Atomoxetine in Different CYP2D6 Genotypes. Atomoxetine Hydrochloride 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 65-71 26859445-3 2016 These differences can be substantial, resulting in 8-10-fold differences in atomoxetine exposure between CYP2D6 poor metabolizers and extensive metabolizers. Atomoxetine Hydrochloride 76-87 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 105-111 26774596-0 2016 Age-related changes in prefrontal norepinephrine transporter density: The basis for improved cognitive flexibility after low doses of atomoxetine in adolescent rats. Atomoxetine Hydrochloride 134-145 solute carrier family 6 member 2 Rattus norvegicus 34-60 26774596-4 2016 While both of these drugs act as norepinephrine reuptake inhibitors, higher doses of atomoxetine and all doses of methylphenidate also block dopamine transporters (DAT). Atomoxetine Hydrochloride 85-96 solute carrier family 6 member 3 Rattus norvegicus 141-162 26774596-4 2016 While both of these drugs act as norepinephrine reuptake inhibitors, higher doses of atomoxetine and all doses of methylphenidate also block dopamine transporters (DAT). Atomoxetine Hydrochloride 85-96 solute carrier family 6 member 3 Rattus norvegicus 164-167 26660002-0 2016 Single dose, CYP2D6 genotype-stratified pharmacokinetic study of atomoxetine in children with ADHD. Atomoxetine Hydrochloride 65-76 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 13-19 26660002-1 2016 The effect of CYP2D6 genotype on the dose-exposure relationship for atomoxetine has not been well characterized in children. Atomoxetine Hydrochloride 68-79 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 14-20 27052878-2 2016 Inconsistent therapeutic efficacy has been reported with ATX, which may be related to variable CYP2D6-mediated drug clearance. Atomoxetine Hydrochloride 57-60 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 95-101 27052878-3 2016 We characterized ATX metabolism in a panel of human liver samples as a basis for a bottom-up PBPK model to aid in ATX exposure prediction and control. Atomoxetine Hydrochloride 17-20 activation induced cytidine deaminase Homo sapiens 107-110 27052878-3 2016 We characterized ATX metabolism in a panel of human liver samples as a basis for a bottom-up PBPK model to aid in ATX exposure prediction and control. Atomoxetine Hydrochloride 114-117 activation induced cytidine deaminase Homo sapiens 107-110 27052878-10 2016 Competing pathways of ATX metabolism [N-desmethylatomoxetine (NDM-ATX) and 2-CH2OH-ATX formation] had increasing importance in livers with lesser CYP2D6 activity, but, overall ATX clearance was still compromised. Atomoxetine Hydrochloride 22-25 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 146-152 27052878-11 2016 Modeling ATX exposure to individualize therapy would require comprehensive knowledge of factors that affect CYP2D6 activity as well as an understanding of competing pathways, particularly for individuals with lower CYP2D6 activity. Atomoxetine Hydrochloride 9-12 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 108-114 27052878-11 2016 Modeling ATX exposure to individualize therapy would require comprehensive knowledge of factors that affect CYP2D6 activity as well as an understanding of competing pathways, particularly for individuals with lower CYP2D6 activity. Atomoxetine Hydrochloride 9-12 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 215-221 26859445-5 2016 The present review focuses on atomoxetine metabolism, disposition, and genetic polymorphisms of CYP2D6 as they specifically relate to atomoxetine, and provides an in-depth discussion of the fundamental pharmacokinetics of the drug including its absorption, distribution, metabolism, and excretion in pediatric and adult populations. Atomoxetine Hydrochloride 30-41 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 96-102 26859445-5 2016 The present review focuses on atomoxetine metabolism, disposition, and genetic polymorphisms of CYP2D6 as they specifically relate to atomoxetine, and provides an in-depth discussion of the fundamental pharmacokinetics of the drug including its absorption, distribution, metabolism, and excretion in pediatric and adult populations. Atomoxetine Hydrochloride 134-145 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 96-102 26859445-6 2016 Further, a summary of relationships between genetic variants of CYP2D6 and to some degree, CYP2C19, are provided with respect to atomoxetine plasma concentrations, central nervous system (CNS) pharmacokinetics, and associated clinical implications for pharmacotherapy. Atomoxetine Hydrochloride 129-140 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 64-70 26859445-6 2016 Further, a summary of relationships between genetic variants of CYP2D6 and to some degree, CYP2C19, are provided with respect to atomoxetine plasma concentrations, central nervous system (CNS) pharmacokinetics, and associated clinical implications for pharmacotherapy. Atomoxetine Hydrochloride 129-140 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 91-98 27518170-7 2016 CONCLUSIONS: These results demonstrated that the effect of bupropion on CYP2D6 activity was responsible for an increased systemic exposure to atomoxetine (5.1-fold) and also for a decreased exposure to its main metabolite (1.5-fold). Atomoxetine Hydrochloride 142-153 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 72-78 26666748-0 2016 Effect of 24 Cytochrome P450 2D6 Variants Found in the Chinese Population on Atomoxetine Metabolism in vitro. Atomoxetine Hydrochloride 77-88 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 13-32 27114797-7 2016 The administration of atomoxetine significantly reduced tissue tumour necrosis factor alpha (TNF-alpha), and nitric oxide (NO) levels compared to the trauma group (P<0.001). Atomoxetine Hydrochloride 22-33 tumor necrosis factor Rattus norvegicus 93-102 27114797-8 2016 Treatment with atomoxetine also decreased the tissue myeloperoxidase (MPO) activity (P=0.026) and increased the tissue superoxide dismutase (SOD) activity compared to the trauma group (P=0.001 and P=0.004, respectively). Atomoxetine Hydrochloride 15-26 myeloperoxidase Rattus norvegicus 53-68 27114797-8 2016 Treatment with atomoxetine also decreased the tissue myeloperoxidase (MPO) activity (P=0.026) and increased the tissue superoxide dismutase (SOD) activity compared to the trauma group (P=0.001 and P=0.004, respectively). Atomoxetine Hydrochloride 15-26 myeloperoxidase Rattus norvegicus 70-73 26666748-1 2016 OBJECTIVE: The aim of this article was to assess the catalytic activities of 24 cytochrome P450 2D6 (CYP2D6) variants found in the Chinese population toward atomoxetine in vitro as well as CYP2D6.1. Atomoxetine Hydrochloride 157-168 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 80-99 26666748-1 2016 OBJECTIVE: The aim of this article was to assess the catalytic activities of 24 cytochrome P450 2D6 (CYP2D6) variants found in the Chinese population toward atomoxetine in vitro as well as CYP2D6.1. Atomoxetine Hydrochloride 157-168 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 101-107 26666748-2 2016 METHODS: In this study, the co-expression enzyme of human recombinant CYPOR, CYPb5, and CYP2D6.1 or other CYP2D6 variants with the baculovirus-mediated insect cells (Sf21) was used to study the catalytic activities of 24 CYP2D6 variants toward atomoxetine metabolism. Atomoxetine Hydrochloride 244-255 cytochrome p450 oxidoreductase Homo sapiens 70-75 26666748-2 2016 METHODS: In this study, the co-expression enzyme of human recombinant CYPOR, CYPb5, and CYP2D6.1 or other CYP2D6 variants with the baculovirus-mediated insect cells (Sf21) was used to study the catalytic activities of 24 CYP2D6 variants toward atomoxetine metabolism. Atomoxetine Hydrochloride 244-255 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 88-94 26730199-4 2016 However, with the approval of atomoxetine (Strattera( ), [ATX]) by the US Food and Drug Administration in late 2002, an effective non-stimulant option became available. Atomoxetine Hydrochloride 30-41 ectonucleotide pyrophosphatase/phosphodiesterase 2 Homo sapiens 58-61 26671145-4 2015 In addition, pre-treatment with 30 mg/kg ATX, which had neuroprotective effect against ischemic damage, distinctly attenuated the activation of astrocytes and microglia in the ischemic CA1 region compared with the vehicle-treated ischemia group by glial fibrillary acidic protein (for astrocytes) and ionized calcium-binding adapter molecule 1 (for microglia) immunohistochemistry. Atomoxetine Hydrochloride 41-44 allograft inflammatory factor 1 Homo sapiens 301-343 26693006-3 2015 This pilot study explores the effects of atomoxetine on gait in PD patients with dopa-unresponsive FoG using a novel paradigm for objective gait assessment. Atomoxetine Hydrochloride 41-52 zinc finger protein, FOG family member 1 Homo sapiens 99-102 26640376-5 2015 Here we report the results of a pilot study conducted to evaluate changes in striatal DAT after 8 weeks of atomoxetine treatment. Atomoxetine Hydrochloride 107-118 solute carrier family 6 member 3 Homo sapiens 86-89 26640376-6 2015 Our results suggest that 8 weeks of atomoxetine treatment may change striatal DAT bioavailability as measured via SPECT but that change was not correlated with genotype or clinical improvement. Atomoxetine Hydrochloride 36-47 solute carrier family 6 member 3 Homo sapiens 78-81 26447643-0 2015 Variants of Dopamine Beta Hydroxylase Gene Moderate Atomoxetine Response in Children with Attention-Deficit/Hyperactivity Disorder. Atomoxetine Hydrochloride 52-63 dopamine beta-hydroxylase Homo sapiens 12-37 26254792-0 2015 Effects of the CYP2D6*10 allele on the pharmacokinetics of atomoxetine and its metabolites. Atomoxetine Hydrochloride 59-70 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 15-21 26254792-8 2015 The concentration of active moieties of atomoxetine (atomoxetine + 4-HAT) in the CYP2D6*10/*10 group was 3.32-fold higher than that in the CYP2D6*wt/*wt group. Atomoxetine Hydrochloride 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 81-87 26254792-8 2015 The concentration of active moieties of atomoxetine (atomoxetine + 4-HAT) in the CYP2D6*10/*10 group was 3.32-fold higher than that in the CYP2D6*wt/*wt group. Atomoxetine Hydrochloride 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 139-145 26254792-8 2015 The concentration of active moieties of atomoxetine (atomoxetine + 4-HAT) in the CYP2D6*10/*10 group was 3.32-fold higher than that in the CYP2D6*wt/*wt group. Atomoxetine Hydrochloride 53-64 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 81-87 26254792-9 2015 The mean exposure to active moieties of atomoxetine was markedly higher in subjects with the CYP2D6*10/*10 genotype compared to that in those with the CYP2D6*wt/*wt genotype. Atomoxetine Hydrochloride 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 93-99 26254792-9 2015 The mean exposure to active moieties of atomoxetine was markedly higher in subjects with the CYP2D6*10/*10 genotype compared to that in those with the CYP2D6*wt/*wt genotype. Atomoxetine Hydrochloride 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 151-157 26254792-10 2015 The higher systemic exposure of the active atomoxetine moieties in CYP2D6*10/*10 individuals may increase the risk of concentration-related adverse events of atomoxetine, although this has not yet been clinically confirmed. Atomoxetine Hydrochloride 43-54 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 67-73 26254792-10 2015 The higher systemic exposure of the active atomoxetine moieties in CYP2D6*10/*10 individuals may increase the risk of concentration-related adverse events of atomoxetine, although this has not yet been clinically confirmed. Atomoxetine Hydrochloride 158-169 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 67-73 26447643-4 2015 This study aimed to investigate whether variants in the DBH gene have an effect on the differential response to atomoxetine. Atomoxetine Hydrochloride 112-123 dopamine beta-hydroxylase Homo sapiens 56-59 26447643-15 2015 CONCLUSIONS: Variants in DBH genes were associated with atomoxetine response in the treatment of ADHD. Atomoxetine Hydrochloride 56-67 dopamine beta-hydroxylase Homo sapiens 25-28 25919121-0 2015 CYP2D6 predicted metabolizer status and safety in adult patients with attention-deficit hyperactivity disorder participating in a large placebo-controlled atomoxetine maintenance of response clinical trial. Atomoxetine Hydrochloride 155-166 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 25919121-1 2015 Atomoxetine, which is indicated for treatment of attention-deficit hyperactivity disorder (ADHD), is predominantly metabolized by genetically polymorphic cytochrome P450 2D6 (CYP2D6). Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 154-173 25919121-1 2015 Atomoxetine, which is indicated for treatment of attention-deficit hyperactivity disorder (ADHD), is predominantly metabolized by genetically polymorphic cytochrome P450 2D6 (CYP2D6). Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 175-181 25919121-9 2015 These results suggest that data from CYP2D6 intermediate and extensive/ultrarapid metabolizers can be combined when considering safety analyses related to atomoxetine. Atomoxetine Hydrochloride 155-166 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 37-43 26314574-1 2015 Atomoxetine is indicated for the treatment of attention deficit hyperactivity disorder and is predominantly metabolized by the CYP2D6 enzyme. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 127-133 26088654-0 2015 Translating Pharmacogenetics to Clinical Practice: Do Cytochrome P450 2D6 Ultrarapid Metabolizers Need Higher Atomoxetine Doses? Atomoxetine Hydrochloride 110-121 cytochrome P450 2D6 Homo sapiens 54-73 25841321-0 2015 Atomoxetine reverses locomotor hyperactivity, impaired novel object recognition, and prepulse inhibition impairment in mice lacking pituitary adenylate cyclase-activating polypeptide. Atomoxetine Hydrochloride 0-11 adenylate cyclase activating polypeptide 1 Mus musculus 132-182 25841321-4 2015 ATX treatment significantly alleviated hyperactivity of pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP(-/-)) mice with C57BL/6J and 129S6/SvEvTac hybrid background. Atomoxetine Hydrochloride 0-3 adenylate cyclase activating polypeptide 1 Mus musculus 56-106 25841321-4 2015 ATX treatment significantly alleviated hyperactivity of pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP(-/-)) mice with C57BL/6J and 129S6/SvEvTac hybrid background. Atomoxetine Hydrochloride 0-3 adenylate cyclase activating polypeptide 1 Mus musculus 108-113 25841321-4 2015 ATX treatment significantly alleviated hyperactivity of pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP(-/-)) mice with C57BL/6J and 129S6/SvEvTac hybrid background. Atomoxetine Hydrochloride 0-3 adenylate cyclase activating polypeptide 1 Mus musculus 126-131 25841321-5 2015 ATX also improved impaired novel object recognition memory and prepulse inhibition in PACAP(-/-) mice with CD1 background. Atomoxetine Hydrochloride 0-3 adenylate cyclase activating polypeptide 1 Mus musculus 86-91 25841321-5 2015 ATX also improved impaired novel object recognition memory and prepulse inhibition in PACAP(-/-) mice with CD1 background. Atomoxetine Hydrochloride 0-3 CD1 antigen complex Mus musculus 107-110 25841321-6 2015 The ATX-induced increases in extracellular noradrenaline and dopamine levels were significantly higher in the prefrontal cortex of PACAP(-/-) mice compared to wild-type mice with C57BL/6J and 129S6/SvEvTac hybrid background. Atomoxetine Hydrochloride 4-7 adenylate cyclase activating polypeptide 1 Mus musculus 131-136 25841321-7 2015 These results suggest that ATX treatment-induced increases in central monoamine metabolism may be involved in the rescue of ADHD-related abnormalities in PACAP(-/-) mice. Atomoxetine Hydrochloride 27-30 adenylate cyclase activating polypeptide 1 Mus musculus 154-159 25554436-2 2015 Based on the evidence from treatment effect of atomoxetine, which interacts directly with the norepinephrine transporter, on visual memory in children with ADHD, this study examined the linkage disequilibrium structure of the norepinephrine transporter gene (SLC6A2) and the association between SLC6A2 and ADHD and visual memory, a promising endophenotype for ADHD. Atomoxetine Hydrochloride 47-58 solute carrier family 6 member 2 Homo sapiens 226-252 26293742-11 2015 After controlling for comorbidities, IRR was still significantly higher in the atomoxetine group compared with the methylphenidate group for a number of mild (decreased appetite, weight loss, abdominal pain, dyspepsia, stomach ache, irritability, mood disorder and dizziness) and severe (gastrointestinal, neuropsychiatric, and cardiovascular) AEs. Atomoxetine Hydrochloride 79-90 insulin receptor related receptor Homo sapiens 37-40 25185131-3 2014 We previously reported in a proof of concept study that pediatric doses of the norepinephrine transporter blockade, atomoxetine, increases blood pressure in autonomic failure patients with residual sympathetic activity compared with placebo. Atomoxetine Hydrochloride 116-127 solute carrier family 6 member 2 Homo sapiens 79-105 24597602-10 2014 Whereas, administration of vanillin as well as atomoxetine has significantly attenuated 2VO induced impaired locomotion, motor coordination, learning and memory, brain damage, brain oxidative stress and higher AChE activity. Atomoxetine Hydrochloride 47-58 acetylcholinesterase Mus musculus 210-214 24703228-6 2014 Cox proportional hazards models with propensity score adjustment assessed differences in atomoxetine persistence among initial-dose cohorts. Atomoxetine Hydrochloride 89-100 cytochrome c oxidase subunit 8A Homo sapiens 0-3 24703228-7 2014 RESULTS: In patients treated with atomoxetine who had available dosing information (N = 134), Cox proportional hazards revealed lower (< 0.5 mg/kg) initial dose was significantly associated with shorter medication persistence (p < 0.01). Atomoxetine Hydrochloride 34-45 cytochrome c oxidase subunit 8A Homo sapiens 94-97 25292181-8 2014 Atomoxetine increased p-mTOR 24 hours after treatment in naive mice, but did not change any other biomarkers. Atomoxetine Hydrochloride 0-11 mechanistic target of rapamycin kinase Mus musculus 24-28 25292181-10 2014 In contrast to what was observed with acute treatment, chronic treatment (7 days) with atomoxetine increased p-Akt and p-FoxO3a, and sustained PGC-1alpha expression and skeletal muscle mass in dexamethasone-treated mice, in a manner comparable to formoterol. Atomoxetine Hydrochloride 87-98 thymoma viral proto-oncogene 1 Mus musculus 111-114 25292181-10 2014 In contrast to what was observed with acute treatment, chronic treatment (7 days) with atomoxetine increased p-Akt and p-FoxO3a, and sustained PGC-1alpha expression and skeletal muscle mass in dexamethasone-treated mice, in a manner comparable to formoterol. Atomoxetine Hydrochloride 87-98 forkhead box O3 Mus musculus 121-127 25292181-10 2014 In contrast to what was observed with acute treatment, chronic treatment (7 days) with atomoxetine increased p-Akt and p-FoxO3a, and sustained PGC-1alpha expression and skeletal muscle mass in dexamethasone-treated mice, in a manner comparable to formoterol. Atomoxetine Hydrochloride 87-98 peroxisome proliferative activated receptor, gamma, coactivator 1 alpha Mus musculus 143-153 25450119-4 2014 RESULTS: Atomoxetine reduced the hyperactivity displayed by NK1R(-/-) mice in the LDEB at a dose (3mg/kg) which did not affect the locomotor activity of wildtypes. Atomoxetine Hydrochloride 9-20 tachykinin receptor 1 Mus musculus 60-64 25450119-5 2014 Atomoxetine (10mg/kg) also reduced impulsivity in NK1R(-/-) mice, but not wildtypes, in the 5-CSRTT. Atomoxetine Hydrochloride 0-11 tachykinin receptor 1 Mus musculus 50-54 25450119-7 2014 CONCLUSIONS: This evidence that atomoxetine reduces hyperactive/impulsive behaviours in NK1R(-/-) mice consolidates the validity of using NK1R(-/-) mice in research of the aetiology and treatment of ADHD. Atomoxetine Hydrochloride 32-43 tachykinin receptor 1 Mus musculus 88-92 25450119-7 2014 CONCLUSIONS: This evidence that atomoxetine reduces hyperactive/impulsive behaviours in NK1R(-/-) mice consolidates the validity of using NK1R(-/-) mice in research of the aetiology and treatment of ADHD. Atomoxetine Hydrochloride 32-43 tachykinin receptor 1 Mus musculus 138-142 24582917-7 2014 Systemic administration of norepinephrine reuptake inhibitor atomoxetine increased c-Fos protein expression in BLA neurons following fear conditioning training and promoted the expression of conditioned fear in resistant mice. Atomoxetine Hydrochloride 61-72 FBJ osteosarcoma oncogene Mus musculus 83-88 24202115-0 2014 Changes in the serum levels of brain-derived neurotrophic factor in adults with attention deficit hyperactivity disorder after treatment with atomoxetine. Atomoxetine Hydrochloride 142-153 brain derived neurotrophic factor Homo sapiens 31-64 24202115-3 2014 OBJECTIVES: The aim of this study was to evaluate the possible changes in serum levels of BDNF in adults treated with ATX and its relationship with clinical improvement. Atomoxetine Hydrochloride 118-121 brain derived neurotrophic factor Homo sapiens 90-94 24202115-2 2014 Although animal models have provided evidence that brain-derived neurotrophic factor (BDNF) is involved in the effects of ATX in the brain, there are no studies of BDNF in ADHD patients undergoing treatment with ATX. Atomoxetine Hydrochloride 122-125 brain derived neurotrophic factor Homo sapiens 51-84 24202115-2 2014 Although animal models have provided evidence that brain-derived neurotrophic factor (BDNF) is involved in the effects of ATX in the brain, there are no studies of BDNF in ADHD patients undergoing treatment with ATX. Atomoxetine Hydrochloride 122-125 brain derived neurotrophic factor Homo sapiens 86-90 24202115-8 2014 The inattentive subgroup of ATX responders showed a decrease of serum BDNF after 3 months of ATX treatment (p = 0.05) not present in the combined subtype (p = 0.82). Atomoxetine Hydrochloride 28-31 brain derived neurotrophic factor Homo sapiens 70-74 24202115-8 2014 The inattentive subgroup of ATX responders showed a decrease of serum BDNF after 3 months of ATX treatment (p = 0.05) not present in the combined subtype (p = 0.82). Atomoxetine Hydrochloride 93-96 brain derived neurotrophic factor Homo sapiens 70-74 23933039-1 2014 BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Atomoxetine Hydrochloride 12-23 solute carrier family 6 member 2 Homo sapiens 104-130 24202115-10 2014 The differences between the combined and inattentive subtypes in serum BDNF changes suggest selective ATX-induced effects in the function of brain circuitry. Atomoxetine Hydrochloride 102-105 brain derived neurotrophic factor Homo sapiens 71-75 25252662-7 2014 Thus, although pharmacological studies suggest that the response to atomoxetine in adults with ADHD is not directly mediated by the effect on the BDNF, reductions in BDNF levels in the plasma of adult patients with ADHD have been reported. Atomoxetine Hydrochloride 68-79 brain derived neurotrophic factor Homo sapiens 166-170 24346747-0 2014 Effects of CYP2C19 genetic polymorphisms on atomoxetine pharmacokinetics. Atomoxetine Hydrochloride 44-55 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 11-18 24346747-2 2014 Atomoxetine metabolism is mediated by CYP2D6 and CYP2C19. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 38-44 24346747-2 2014 Atomoxetine metabolism is mediated by CYP2D6 and CYP2C19. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 49-56 24346747-3 2014 This study aimed to investigate the effect of the CYP2C19 genetic polymorphism on the pharmacokinetics of atomoxetine and its metabolites, 4-hydroxyatomoxetine and N-desmethylatomoxetine. Atomoxetine Hydrochloride 106-117 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 50-57 24346747-4 2014 A single 40-mg oral dose of atomoxetine was administered to 40 subjects with different CYP2C19 genotypes (all participants carried the CYP2D6*1/*10 genotype). Atomoxetine Hydrochloride 28-39 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 135-141 24346747-6 2014 For atomoxetine, the CYP2C19 poor metabolizer (PM) group showed significantly increased maximum plasma concentration and AUC0- (area under the plasma concentration-time curve from 0 to infinity) and decreased apparent oral clearance compared with samples of the CYP2C19 extensive metabolizer (EM) and intermediate metabolizer (IM) groups (P < 0.001 for all). Atomoxetine Hydrochloride 4-15 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 21-28 24346747-7 2014 The half-life of atomoxetine in the CYP2C19PM group was also significantly longer than in the other genotype groups (P < 0.01 for CYP2C19EM and P < 0.05 for CYP2C19IM groups). Atomoxetine Hydrochloride 17-28 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 36-43 24346747-7 2014 The half-life of atomoxetine in the CYP2C19PM group was also significantly longer than in the other genotype groups (P < 0.01 for CYP2C19EM and P < 0.05 for CYP2C19IM groups). Atomoxetine Hydrochloride 17-28 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 133-140 24346747-7 2014 The half-life of atomoxetine in the CYP2C19PM group was also significantly longer than in the other genotype groups (P < 0.01 for CYP2C19EM and P < 0.05 for CYP2C19IM groups). Atomoxetine Hydrochloride 17-28 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 133-140 24346747-9 2014 These results suggest that the genetic polymorphisms of CYP2C19 significantly affect the pharmacokinetics of atomoxetine. Atomoxetine Hydrochloride 109-120 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 56-63 23933039-1 2014 BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Atomoxetine Hydrochloride 12-23 solute carrier family 6 member 4 Homo sapiens 259-280 23933039-1 2014 BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Atomoxetine Hydrochloride 12-23 solute carrier family 6 member 4 Homo sapiens 282-286 23933039-1 2014 BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Atomoxetine Hydrochloride 25-28 solute carrier family 6 member 2 Homo sapiens 104-130 23933039-1 2014 BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Atomoxetine Hydrochloride 25-28 solute carrier family 6 member 4 Homo sapiens 259-280 23933039-1 2014 BACKGROUND: Atomoxetine (ATX), a drug for treatment of depression and ADHD, has a high affinity for the norepinephrine transporter (NET); however, our previous study showed it had a blocking effect similar to fluoxetine on binding of [(11)C]DASB, a selective serotonin transporter (SERT) ligand. Atomoxetine Hydrochloride 25-28 solute carrier family 6 member 4 Homo sapiens 282-286 23933039-9 2014 RESULTS: ATX displayed dose-dependent occupancy on both NET and SERT, with a higher occupancy on NET: IC50 of 31+-10 and 99+-21ng/mL plasma for NET and SERT, respectively. Atomoxetine Hydrochloride 9-12 solute carrier family 6 member 4 Homo sapiens 64-68 23933039-9 2014 RESULTS: ATX displayed dose-dependent occupancy on both NET and SERT, with a higher occupancy on NET: IC50 of 31+-10 and 99+-21ng/mL plasma for NET and SERT, respectively. Atomoxetine Hydrochloride 9-12 solute carrier family 6 member 4 Homo sapiens 152-156 23933039-11 2014 300-600ng/mL plasma), ATX would occupy >90% of NET and >85% of SERT. Atomoxetine Hydrochloride 22-25 solute carrier family 6 member 4 Homo sapiens 69-73 23933039-13 2014 CONCLUSION: Our data suggests that ATX at clinically relevant doses greatly occupies both NET and SERT. Atomoxetine Hydrochloride 35-38 solute carrier family 6 member 4 Homo sapiens 98-102 23822950-10 2013 Rather, atomoxetine decreased V(max) and DAT cell surface expression in SHR OFC, indicating that inhibition of NET by atomoxetine treatment during adolescence indirectly reduced DAT function and trafficking to the cell surface in OFC, specifically in the ADHD model. Atomoxetine Hydrochloride 8-19 solute carrier family 6 member 3 Rattus norvegicus 41-44 24090673-3 2014 METHODS: A beta3-adrenoreceptor selective agonist (CL 316243), an adenylyl cyclase enzyme activator (forskolin) and a potent blocker of presynaptic norepinephrine transporter (atomoxetine), were injected through the tail vein of Swiss Webster mice 30 minutes before intravenous (iv) administration of [(18)F]FDG. Atomoxetine Hydrochloride 176-187 histocompatibility 2, P region beta locus Mus musculus 9-16 24090673-12 2014 Atomoxetine increased [(18)F]FDG SUV of IBAT 1.7-fold greater than that in placebo-treated mice. Atomoxetine Hydrochloride 0-11 solute carrier family 10, member 2 Mus musculus 40-44 23912772-0 2014 Dissociations between cognitive and motor effects of psychostimulants and atomoxetine in hyperactive DAT-KO mice. Atomoxetine Hydrochloride 74-85 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 101-104 23912772-8 2014 Further investigation of the potential involvement of other monoamine systems in the regulation of cognitive functions showed that the norepinephrine transporter blocker atomoxetine restored cognitive performances in DAT-KO mice without affecting hyperactivity. Atomoxetine Hydrochloride 170-181 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 217-220 24348020-0 2013 Atomoxetine affects transcription/translation of the NMDA receptor and the norepinephrine transporter in the rat brain--an in vivo study. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Rattus norvegicus 75-101 24348020-2 2013 The norepinephrine transporter (NET) inhibitor atomoxetine, the first nonstimulant drug licensed for ADHD treatment, also acts as an N-methyl-D-aspartate receptor (NMDAR) antagonist. Atomoxetine Hydrochloride 47-58 solute carrier family 6 member 2 Rattus norvegicus 4-30 24348020-2 2013 The norepinephrine transporter (NET) inhibitor atomoxetine, the first nonstimulant drug licensed for ADHD treatment, also acts as an N-methyl-D-aspartate receptor (NMDAR) antagonist. Atomoxetine Hydrochloride 47-58 solute carrier family 6 member 2 Rattus norvegicus 32-35 23822950-4 2013 This study investigates the effects of atomoxetine ((R)-N-methyl-gamma-(2-methylphenoxy)-benzenepropanamine hydrochloride) treatment, a selective NET inhibitor, during adolescence on cocaine self-administration and on DAT function and cell-surface expression in mPFC and OFC during adulthood. Atomoxetine Hydrochloride 39-50 solute carrier family 6 member 3 Rattus norvegicus 218-221 23822950-10 2013 Rather, atomoxetine decreased V(max) and DAT cell surface expression in SHR OFC, indicating that inhibition of NET by atomoxetine treatment during adolescence indirectly reduced DAT function and trafficking to the cell surface in OFC, specifically in the ADHD model. Atomoxetine Hydrochloride 118-129 solute carrier family 6 member 3 Rattus norvegicus 178-181 24206099-6 2013 The atomoxetine-treated dyslexia-only subjects significantly improved from baseline to week 32 on ADHDRS-IV-Parent:Inv Inattentive subscale, K-SCT Parent and Teacher subscales, and WMTB-C Phonological Loop and Central Executive component scores. Atomoxetine Hydrochloride 4-15 leishmanolysin like peptidase Homo sapiens 115-118 24206099-7 2013 The atomoxetine-treated ADHD-only subjects significantly improved from baseline to Week 32 on ADHDRS-Parent:Inv Total and subscales, ADHDRS-IV-Teacher-Version Hyperactive/Impulsive subscale, LPS Self-Control and Total, all K-SCT subscales, and MSCS Academic and Competence subscale scores. Atomoxetine Hydrochloride 4-15 leishmanolysin like peptidase Homo sapiens 108-111 24098676-6 2013 Atomoxetine, a monoamine reuptake inhibitor that achieves higher levels of norepinephrine than serotonin transporter occupancy, exhibited robust antinociceptive synergy with morphine. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 4 Rattus norvegicus 95-116 23775507-2 2013 Atomoxetine is a selective inhibitor of the presynaptic norepinephrine transporter. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 56-82 23266789-1 2013 Atomoxetine, a selective inhibitor of the norepinephrine transporter, exerts its therapeutic effect for attention-deficit hyperactivity disorder (ADHD) by increasing the concentration of synaptic norepinephrine. Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 42-68 23266789-12 2013 The results of this study suggest that DNA variants of both SLC6A2 and ADRA2A in the adrenergic neurotransmitter system might alter the response to atomoxetine, though further replication study in larger sample for validation of these findings is still needed. Atomoxetine Hydrochloride 148-159 solute carrier family 6 member 2 Homo sapiens 60-66 23266789-12 2013 The results of this study suggest that DNA variants of both SLC6A2 and ADRA2A in the adrenergic neurotransmitter system might alter the response to atomoxetine, though further replication study in larger sample for validation of these findings is still needed. Atomoxetine Hydrochloride 148-159 adrenoceptor alpha 2A Homo sapiens 71-77 23493066-0 2013 Successful treatment with atomoxetine of an adolescent boy with attention deficit/hyperactivity disorder, extreme obesity, and reduced melanocortin 4 receptor function. Atomoxetine Hydrochloride 26-37 melanocortin 4 receptor Homo sapiens 135-158 23566813-0 2013 Daily methylphenidate and atomoxetine treatment impacts on clock gene protein expression in the mouse brain. Atomoxetine Hydrochloride 26-37 circadian locomotor output cycles kaput Mus musculus 59-64 23566813-4 2013 In the present study we have examined the effects of daily methylphenidate or atomoxetine treatment across 7 days on circadian clock gene product expression across numerous brain regions in the male mouse to test the potential impact of such compounds on circadian timing. Atomoxetine Hydrochloride 78-89 circadian locomotor output cycles kaput Mus musculus 127-132 22803597-2 2013 METHODS: Atomoxetine doses were selected to result in plasma concentrations that approximated expected plasma concentrations at both the maximum recommended dose and at a supratherapeutic dose in CYP2D6 extensive metabolizers. Atomoxetine Hydrochloride 9-20 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 196-202 22960082-7 2013 SNAP-25 (synaptosomal-associated protein of 25 kDa), which is an ADHD candidate gene and an important vesicle protein involved in axonal growth, synaptic plasticity and regulation of neurotransmitter release was also significantly upregulated on RNA- and protein level after atomoxetine treatment. Atomoxetine Hydrochloride 275-286 synaptosome associated protein 25 Rattus norvegicus 0-7 22960082-7 2013 SNAP-25 (synaptosomal-associated protein of 25 kDa), which is an ADHD candidate gene and an important vesicle protein involved in axonal growth, synaptic plasticity and regulation of neurotransmitter release was also significantly upregulated on RNA- and protein level after atomoxetine treatment. Atomoxetine Hydrochloride 275-286 synaptosome associated protein 25 Rattus norvegicus 9-50 22960082-9 2013 Especially the increased expression of SNAP-25 and GABA-A receptor subunits may indicate additional active therapeutic mechanisms for atomoxetine. Atomoxetine Hydrochloride 134-145 synaptosome associated protein 25 Rattus norvegicus 39-46 23493066-7 2013 CONCLUSION: Atomoxetine proved to efficiently reduce weight in a severely obese MC4R mutation carrier with ADHD. Atomoxetine Hydrochloride 12-23 melanocortin 4 receptor Homo sapiens 80-84 22311903-4 2012 We hypothesized that increasing residual sympathetic outflow with the alpha-2 antagonist yohimbine would potentiate the pressor effect of norepinephrine transporter blockade with atomoxetine and improve orthostatic tolerance in peripheral autonomic failure. Atomoxetine Hydrochloride 179-190 solute carrier family 6 member 2 Homo sapiens 138-164 22311903-11 2012 In conclusion, the combination of yohimbine and atomoxetine had a synergistic effect on blood pressure and orthostatic tolerance in peripheral autonomic failure, which may be explained by an increased release of norepinephrine in peripheral sympathetic neurons by alpha-2 antagonism combined with a reduced norepinephrine clearance by norepinephrine transporter blockade. Atomoxetine Hydrochloride 48-59 solute carrier family 6 member 2 Homo sapiens 335-361 22311903-2 2012 Norepinephrine transporter blockade with atomoxetine raises blood pressure in autonomic failure by increasing synaptic norepinephrine concentrations in postganglionic sympathetic neurons. Atomoxetine Hydrochloride 41-52 solute carrier family 6 member 2 Homo sapiens 0-26 21963947-0 2011 Regulation of CCL2/MCP-1 production in astrocytes by desipramine and atomoxetine: involvement of alpha2 adrenergic receptors. Atomoxetine Hydrochloride 69-80 C-C motif chemokine ligand 2 Rattus norvegicus 14-18 21543662-1 2012 Atomoxetine is a cytochrome P4502D6 (CYP2D6) substrate. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 17-35 21543662-1 2012 Atomoxetine is a cytochrome P4502D6 (CYP2D6) substrate. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 37-43 21809008-2 2012 The use of the norepinephrine reuptake inhibitor, atomoxetine, to treat ADHD suggests that the activity of the norepinephrine transporter (NET) may be important in regulating impulsive behavior. Atomoxetine Hydrochloride 50-61 solute carrier family 6 member 2 Rattus norvegicus 111-137 21809008-2 2012 The use of the norepinephrine reuptake inhibitor, atomoxetine, to treat ADHD suggests that the activity of the norepinephrine transporter (NET) may be important in regulating impulsive behavior. Atomoxetine Hydrochloride 50-61 solute carrier family 6 member 2 Rattus norvegicus 139-142 21809008-9 2012 Ex vivo analysis of brain tissue indicated that chronic atomoxetine decreased phosphorylation of CREB and ERK in the orbitofrontal cortex and decreased mRNA for BDNF and cdk5. Atomoxetine Hydrochloride 56-67 cAMP responsive element binding protein 1 Rattus norvegicus 97-101 21809008-9 2012 Ex vivo analysis of brain tissue indicated that chronic atomoxetine decreased phosphorylation of CREB and ERK in the orbitofrontal cortex and decreased mRNA for BDNF and cdk5. Atomoxetine Hydrochloride 56-67 Eph receptor B1 Rattus norvegicus 106-109 21809008-9 2012 Ex vivo analysis of brain tissue indicated that chronic atomoxetine decreased phosphorylation of CREB and ERK in the orbitofrontal cortex and decreased mRNA for BDNF and cdk5. Atomoxetine Hydrochloride 56-67 brain-derived neurotrophic factor Rattus norvegicus 161-165 21809008-9 2012 Ex vivo analysis of brain tissue indicated that chronic atomoxetine decreased phosphorylation of CREB and ERK in the orbitofrontal cortex and decreased mRNA for BDNF and cdk5. Atomoxetine Hydrochloride 56-67 cyclin-dependent kinase 5 Rattus norvegicus 170-174 22234242-6 2012 At that time, there were fewer contraindications reported in the SPC for atomoxetine than methylphenidate and the specific contraindications varied considerably amongst methylphenidate formulations. Atomoxetine Hydrochloride 73-84 proline rich protein gene cluster Homo sapiens 65-68 22234242-8 2012 Between September and November 2011, there were three changes to the atomoxetine SPC that resulted in revised prescribing information. Atomoxetine Hydrochloride 69-80 proline rich protein gene cluster Homo sapiens 81-84 22119060-10 2012 Further, atomoxetine treatment at CT6 induced a downregulation of c-Fos and CLOCK in the SCN, but did not alter the expression of PER2 and BMAL1. Atomoxetine Hydrochloride 9-20 FBJ osteosarcoma oncogene Mus musculus 66-71 22119060-12 2012 Atomoxetine treatment preceding a light pulse at CT15 enhanced the magnitude of the photic-phase shift, whereas it altered photic induction of the immediate early gene products c-Fos and ARC in the SCN. Atomoxetine Hydrochloride 0-11 jun proto-oncogene Mus musculus 147-162 22119060-12 2012 Atomoxetine treatment preceding a light pulse at CT15 enhanced the magnitude of the photic-phase shift, whereas it altered photic induction of the immediate early gene products c-Fos and ARC in the SCN. Atomoxetine Hydrochloride 0-11 FBJ osteosarcoma oncogene Mus musculus 177-182 22277677-5 2012 Based on the variability and reported frequency of the CYP2D6 genotype in Asians and Caucasians, the inter-individual variability of CL(int,h,2D6) of extensive metabolizers was estimated at 60-70%, which provided comparable variability of AUC with the literature values of DM, tolterodine, risperidone and atomoxetine. Atomoxetine Hydrochloride 306-317 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 55-61 21963947-0 2011 Regulation of CCL2/MCP-1 production in astrocytes by desipramine and atomoxetine: involvement of alpha2 adrenergic receptors. Atomoxetine Hydrochloride 69-80 C-C motif chemokine ligand 2 Rattus norvegicus 19-24 21963947-2 2011 The treatment of primary rat astrocyte cultures with the noradrenaline transporter inhibitors desipramine or atomoxetine induced the expression and synthesis of CCL2/MCP-1 in these cells. Atomoxetine Hydrochloride 109-120 C-C motif chemokine ligand 2 Rattus norvegicus 161-165 21963947-2 2011 The treatment of primary rat astrocyte cultures with the noradrenaline transporter inhibitors desipramine or atomoxetine induced the expression and synthesis of CCL2/MCP-1 in these cells. Atomoxetine Hydrochloride 109-120 chemokine (C-C motif) ligand 2 Mus musculus 166-171 21109233-2 2011 We have recently shown that impulsivity predating drug-taking increases the susceptibility to relapse to cocaine seeking and that treatment with the anti-impulsivity drug atomoxetine (ATO), a selective norepinephrine re-uptake inhibitor (norepinephrine transporter), prevents relapse. Atomoxetine Hydrochloride 171-182 solute carrier family 6 member 2 Rattus norvegicus 238-264 21852991-3 2011 The primary efficacy measure was change in ADHDRS-IV-Parent: Inv total score after 6 weeks of atomoxetine treatment. Atomoxetine Hydrochloride 94-105 inversin Homo sapiens 61-64 21852991-6 2011 A graded dose response was apparent with mean change in ADHDRS-IV-Parent: Inv total scores of -9.6, -12.3 and -14.5 with atomoxetine 0.2, 0.5 and 1.2 mg/kg/day, respectively (p=0.024 - F-test). Atomoxetine Hydrochloride 121-132 inversin Homo sapiens 74-77 21168122-4 2011 METHODS: Twelve healthy, male volunteers were included in a randomized double-blind, placebo-controlled, within-subjects design to examine the effects of a single dose of atomoxetine on neural activities during a combined Eriksen flanker-Go/NoGo task as measured by functional magnetic resonance imaging. Atomoxetine Hydrochloride 171-182 reticulon 4 Homo sapiens 241-245 21168122-6 2011 Functionally, interaction analysis revealed a significant increase of the error signal (incorrect minus correct NoGo trials) under atomoxetine in bilateral inferior frontal cortex and presupplementary motor area. Atomoxetine Hydrochloride 131-142 reticulon 4 Homo sapiens 112-116 21739117-2 2011 The norepinephrine transporter facilitates the reuptake of norepinephrine and dopamine in the prefrontal cortex and represents the main target of atomoxetine, an effective drug in the treatment of ADHD. Atomoxetine Hydrochloride 146-157 solute carrier family 6 member 2 Homo sapiens 4-30 21109233-2 2011 We have recently shown that impulsivity predating drug-taking increases the susceptibility to relapse to cocaine seeking and that treatment with the anti-impulsivity drug atomoxetine (ATO), a selective norepinephrine re-uptake inhibitor (norepinephrine transporter), prevents relapse. Atomoxetine Hydrochloride 184-187 solute carrier family 6 member 2 Rattus norvegicus 238-264 21109233-10 2011 CONCLUSIONS: Selective norepinephrine transporter inhibition by ATO might be an effective treatment for the prevention of relapse to both stimulant and opiate addiction. Atomoxetine Hydrochloride 64-67 solute carrier family 6 member 2 Rattus norvegicus 23-49 20393461-9 2010 Atomoxetine and reboxetine also inhibited GIRK currents induced by activation of cloned A(1) adenosine receptors or by intracellularly applied GTPgammaS, a nonhydrolyzable GTP analogue. Atomoxetine Hydrochloride 0-11 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 42-46 20691787-0 2010 Sub-chronic exposure to atomoxetine up-regulates BDNF expression and signalling in the brain of adolescent spontaneously hypertensive rats: comparison with methylphenidate. Atomoxetine Hydrochloride 24-35 brain-derived neurotrophic factor Rattus norvegicus 49-53 20691787-3 2010 Atomoxetine (ATX) up-regulated BDNF mRNA levels in the hippocampus whereas methylphenidate (MPH) increased BDNF gene expression in the nucleus accumbens and caudate-putamen. Atomoxetine Hydrochloride 0-11 brain-derived neurotrophic factor Rattus norvegicus 31-35 20691787-3 2010 Atomoxetine (ATX) up-regulated BDNF mRNA levels in the hippocampus whereas methylphenidate (MPH) increased BDNF gene expression in the nucleus accumbens and caudate-putamen. Atomoxetine Hydrochloride 13-16 brain-derived neurotrophic factor Rattus norvegicus 31-35 20691787-5 2010 Analysis of BDNF-mediated signalling in the prefrontal cortex revealed that ATX enhanced AKT and GSK3beta phosphorylation whereas MPH reduced the synaptic levels of trkB, the high-affinity BDNF receptor, and ERK1/2 activation. Atomoxetine Hydrochloride 76-79 brain-derived neurotrophic factor Rattus norvegicus 12-16 20691787-5 2010 Analysis of BDNF-mediated signalling in the prefrontal cortex revealed that ATX enhanced AKT and GSK3beta phosphorylation whereas MPH reduced the synaptic levels of trkB, the high-affinity BDNF receptor, and ERK1/2 activation. Atomoxetine Hydrochloride 76-79 AKT serine/threonine kinase 1 Rattus norvegicus 89-92 20691787-5 2010 Analysis of BDNF-mediated signalling in the prefrontal cortex revealed that ATX enhanced AKT and GSK3beta phosphorylation whereas MPH reduced the synaptic levels of trkB, the high-affinity BDNF receptor, and ERK1/2 activation. Atomoxetine Hydrochloride 76-79 glycogen synthase kinase 3 beta Rattus norvegicus 97-105 20691935-0 2010 Recognition of impaired atomoxetine metabolism because of low CYP2D6 activity. Atomoxetine Hydrochloride 24-35 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 62-68 20691935-5 2010 Four children with compromised cytochrome P450 2D6 activity responded better after decreasing their atomoxetine dose. Atomoxetine Hydrochloride 100-111 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 31-50 20691935-8 2010 Physicians should be aware of the typical pattern of adverse effects and late response in atomoxetine treatment, possibly indicating compromised cytochrome P450 2D6 activity. Atomoxetine Hydrochloride 90-101 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 145-164 20691935-10 2010 Further research is needed to establish the cost versus benefit ratio of prospective cytochrome P450 2D6 genotyping in atomoxetine treatment. Atomoxetine Hydrochloride 119-130 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 85-104 20661027-0 2010 Treatment of chronic akinetic mutism with atomoxetine: subtraction analysis of brain f-18 fluorodeoxyglucose positron emission tomographic images before and after medication: a case report. Atomoxetine Hydrochloride 42-53 mastermind like domain containing 1 Homo sapiens 85-89 20403082-9 2010 Acute and chronic atomoxetine 1 mg/kg and methylphenidate 3 mg/kg increased the expression of c-Fos in the prefrontal cortex, but not in the striatum, to a similar extent. Atomoxetine Hydrochloride 18-29 FBJ osteosarcoma oncogene Mus musculus 94-99 20393461-5 2010 In this study, we investigated the effects of atomoxetine and reboxetine on GIRK channels using the Xenopus oocyte expression assay. Atomoxetine Hydrochloride 46-57 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 76-80 20393461-6 2010 In oocytes injected with mRNA for GIRK1/GIRK2, GIRK2, or GIRK1/GIRK4 subunits, extracellular application of atomoxetine or reboxetine reversibly reduced GIRK currents. Atomoxetine Hydrochloride 108-119 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 34-39 20393461-6 2010 In oocytes injected with mRNA for GIRK1/GIRK2, GIRK2, or GIRK1/GIRK4 subunits, extracellular application of atomoxetine or reboxetine reversibly reduced GIRK currents. Atomoxetine Hydrochloride 108-119 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 57-62 20393461-6 2010 In oocytes injected with mRNA for GIRK1/GIRK2, GIRK2, or GIRK1/GIRK4 subunits, extracellular application of atomoxetine or reboxetine reversibly reduced GIRK currents. Atomoxetine Hydrochloride 108-119 potassium inwardly rectifying channel subfamily J member 5 L homeolog Xenopus laevis 63-68 20393461-6 2010 In oocytes injected with mRNA for GIRK1/GIRK2, GIRK2, or GIRK1/GIRK4 subunits, extracellular application of atomoxetine or reboxetine reversibly reduced GIRK currents. Atomoxetine Hydrochloride 108-119 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 34-38 20393461-10 2010 Furthermore, the GIRK currents induced by ethanol were concentration-dependently inhibited by extracellularly applied atomoxetine but not by intracellularly applied atomoxetine. Atomoxetine Hydrochloride 118-129 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 17-21 20393461-11 2010 The present results suggest that atomoxetine and reboxetine inhibit brain- and cardiac-type GIRK channels, revealing a novel characteristic of clinically used NRIs. Atomoxetine Hydrochloride 33-44 potassium inwardly rectifying channel subfamily J member 3 L homeolog Xenopus laevis 92-96 20018569-4 2010 Atomoxetine decreased (p<0.05) baseline mBP from 128+/-11 mm Hg (mean+/-SD) to 117+/-19 mm Hg; baseline HR slowed from 380+/-23 bpm to 351+/-21 bpm. Atomoxetine Hydrochloride 0-11 myelin basic protein Mus musculus 43-46 19568826-5 2010 Compared with patients in the placebo group (n = 33), patients treated with atomoxetine (n = 72) with a mean final dose of 1.4 mg/kg showed significantly greater improvement in ADHDRS-IV-Parent:Inv total score (least-squares mean: atomoxetine, -15.8; placebo, -11.4; p = 0.013). Atomoxetine Hydrochloride 76-87 inversin Homo sapiens 194-197 20004658-2 2010 The noradrenalin transporter blocker atomoxetine was more effective in reducing stress-induced hyperthermia, induced by an injection, in serotonin transporter (SERT) knockout (SERT(-/-)) rats compared to SERT(+/+) rats. Atomoxetine Hydrochloride 37-48 solute carrier family 6 member 4 Rattus norvegicus 137-158 20004658-2 2010 The noradrenalin transporter blocker atomoxetine was more effective in reducing stress-induced hyperthermia, induced by an injection, in serotonin transporter (SERT) knockout (SERT(-/-)) rats compared to SERT(+/+) rats. Atomoxetine Hydrochloride 37-48 solute carrier family 6 member 4 Rattus norvegicus 160-164 20004658-2 2010 The noradrenalin transporter blocker atomoxetine was more effective in reducing stress-induced hyperthermia, induced by an injection, in serotonin transporter (SERT) knockout (SERT(-/-)) rats compared to SERT(+/+) rats. Atomoxetine Hydrochloride 37-48 solute carrier family 6 member 4 Rattus norvegicus 176-180 20004658-2 2010 The noradrenalin transporter blocker atomoxetine was more effective in reducing stress-induced hyperthermia, induced by an injection, in serotonin transporter (SERT) knockout (SERT(-/-)) rats compared to SERT(+/+) rats. Atomoxetine Hydrochloride 37-48 solute carrier family 6 member 4 Rattus norvegicus 176-180 19733552-3 2009 Genetic analyses of MPH metabolizing carboxylesterase 1 enzyme (CES1) have not been carried out, whereas, meta-analysis of CYP2D6 genetic variants has been already indicated significant pharmacogenetic differences in atomoxetine treatment. Atomoxetine Hydrochloride 217-228 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 123-129 19361809-5 2009 Further studies in a larger population of patients with FoG are needed to determine whether atomoxetine is a useful drug in the treatment of this, often disabling, feature of PD. Atomoxetine Hydrochloride 92-103 zinc finger protein, FOG family member 1 Homo sapiens 56-59 19785510-9 2009 Atomoxetine-treated patients showed greater reductions from baseline to week 12 of total ADHDRS-IV-Parent:Inv score than placebo-treated patients (least square mean difference: -7.9 [95% CI: -11.0 to -4.8], corresponding to a large effect size of 0.8). Atomoxetine Hydrochloride 0-11 inversin Homo sapiens 106-109 19785510-11 2009 At the end of the study, 50% of atomoxetine-treated patients (14% with placebo) showed a reduction > or =40% in total ADHDRS-IV-Parent:Inv score, and only 29% (46% with placebo) were severely ill (by CGI-ADHD-S). Atomoxetine Hydrochloride 32-43 inversin Homo sapiens 138-141 19700948-0 2009 Atomoxetine augmentation of cholinesterase inhibitor therapy in patients with Alzheimer disease: 6-month, randomized, double-blind, placebo-controlled, parallel-trial study. Atomoxetine Hydrochloride 0-11 butyrylcholinesterase Homo sapiens 28-42 19300997-0 2009 Saturated norepinephrine transporter occupancy by atomoxetine relevant to clinical doses: a rhesus monkey study with (S,S)-[(18)F]FMeNER-D (2). Atomoxetine Hydrochloride 50-61 solute carrier family 6 member 2 Macaca mulatta 10-36 19300997-1 2009 PURPOSE: In a previous PET study on norepinephrine transporter (NET) occupancy in the nonhuman primate brain, the relationship between NET occupancy and atomoxetine plasma concentration, and occupancies among different brain regions, were not demonstrated adequately. Atomoxetine Hydrochloride 153-164 solute carrier family 6 member 2 Macaca mulatta 135-138 19300997-6 2009 RESULTS: NET occupancy increased regionally and uniformly as the plasma concentration of atomoxetine increased. Atomoxetine Hydrochloride 89-100 solute carrier family 6 member 2 Macaca mulatta 9-12 19300997-8 2009 CONCLUSION: The results indicate that clinical doses of atomoxetine would occupy NET almost completely. Atomoxetine Hydrochloride 56-67 solute carrier family 6 member 2 Macaca mulatta 81-84 19387424-2 2009 We investigated whether polymorphisms in the NET/SLC6A2 gene may influence atomoxetine response in ADHD. Atomoxetine Hydrochloride 75-86 solute carrier family 6 member 2 Homo sapiens 45-48 19387424-2 2009 We investigated whether polymorphisms in the NET/SLC6A2 gene may influence atomoxetine response in ADHD. Atomoxetine Hydrochloride 75-86 solute carrier family 6 member 2 Homo sapiens 49-55 19387424-6 2009 Significant (p<0.05) associations between 20 NET/SLC6A2 single nucleotide polymorphisms (SNPs) and clinical efficacy in atomoxetine responders, compared with non-responders, were observed. Atomoxetine Hydrochloride 123-134 solute carrier family 6 member 2 Homo sapiens 48-51 19387424-0 2009 A haplotype of the norepinephrine transporter (Net) gene Slc6a2 is associated with clinical response to atomoxetine in attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 104-115 solute carrier family 6 member 2 Homo sapiens 19-45 19387424-6 2009 Significant (p<0.05) associations between 20 NET/SLC6A2 single nucleotide polymorphisms (SNPs) and clinical efficacy in atomoxetine responders, compared with non-responders, were observed. Atomoxetine Hydrochloride 123-134 solute carrier family 6 member 2 Homo sapiens 52-58 19387424-0 2009 A haplotype of the norepinephrine transporter (Net) gene Slc6a2 is associated with clinical response to atomoxetine in attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 104-115 solute carrier family 6 member 2 Homo sapiens 47-50 19387424-0 2009 A haplotype of the norepinephrine transporter (Net) gene Slc6a2 is associated with clinical response to atomoxetine in attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 104-115 solute carrier family 6 member 2 Homo sapiens 57-63 19387424-8 2009 Clinical efficacy of atomoxetine treatment in ADHD shows potential dependence upon a series of genetic polymorphisms of its mechanistic target, the norepinephrine transporter. Atomoxetine Hydrochloride 21-32 solute carrier family 6 member 2 Homo sapiens 148-174 19387424-1 2009 Atomoxetine is a specific inhibitor of the norepinephrine transporter (NET) that has demonstrated efficacy in the treatment of children with attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 43-69 18811678-9 2009 In addition, the NE transporter (NET) inhibitor atomoxetine attenuates some of d-amphetamine"s subjective and physiological effects in humans. Atomoxetine Hydrochloride 48-59 solute carrier family 6 member 2 Homo sapiens 17-31 19387424-1 2009 Atomoxetine is a specific inhibitor of the norepinephrine transporter (NET) that has demonstrated efficacy in the treatment of children with attention-deficit hyperactivity disorder (ADHD). Atomoxetine Hydrochloride 0-11 solute carrier family 6 member 2 Homo sapiens 71-74 19404616-10 2009 CONCLUSIONS: The results suggest that atomoxetine exerts its beneficial effects on SSRT via its action on noradrenaline re-uptake, as the specific DAT blocker GBR-12909 and serotonin reuptake blockade had only minor effects on SSRT. Atomoxetine Hydrochloride 38-49 solute carrier family 6 member 3 Rattus norvegicus 147-150 18811678-9 2009 In addition, the NE transporter (NET) inhibitor atomoxetine attenuates some of d-amphetamine"s subjective and physiological effects in humans. Atomoxetine Hydrochloride 48-59 solute carrier family 6 member 2 Homo sapiens 33-36 19445548-17 2009 CYP2D6 inhibitors, such as paroxetine, are associated with changes in atomoxetine pharmacokinetics similar to those observed among poor CYP2D6 metabolizers. Atomoxetine Hydrochloride 70-81 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 19154426-0 2009 Selective noradrenaline reuptake inhibitor atomoxetine directly blocks hERG currents. Atomoxetine Hydrochloride 43-54 ETS transcription factor ERG Homo sapiens 71-75 19154426-4 2009 We therefore analysed acute and subacute effects of atomoxetine on cloned human Ether-a-Go-Go-Related Gene (hERG) channels. Atomoxetine Hydrochloride 52-63 ETS transcription factor ERG Homo sapiens 108-112 19154426-8 2009 KEY RESULTS: In human embryonic kidney cells, atomoxetine inhibited hERG current with an IC(50) of 6.3 micromol.L(-1). Atomoxetine Hydrochloride 46-57 ETS transcription factor ERG Homo sapiens 68-72 19445548-32 2009 Atomoxetine appeared better tolerated among extensive CYP2D6 metabolizers than among poor metabolizers. Atomoxetine Hydrochloride 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 54-60 19012633-1 2008 An 11 year old boy developed acute agitation and suicidal ideation after commencing atomoxetine shortly after it was approved for use in Australia. Atomoxetine Hydrochloride 84-95 DDB1 and CUL4 associated factor 7 Homo sapiens 0-5