Page last updated: 2024-12-11

dextromethorphan

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Description

Dextromethorphan: Methyl analog of DEXTRORPHAN that shows high affinity binding to several regions of the brain, including the medullary cough center. This compound is an NMDA receptor antagonist (RECEPTORS, N-METHYL-D-ASPARTATE) and acts as a non-competitive channel blocker. It is one of the widely used ANTITUSSIVES, and is also used to study the involvement of glutamate receptors in neurotoxicity. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

dextromethorphan : A 6-methoxy-11-methyl-1,3,4,9,10,10a-hexahydro-2H-10,4a-(epiminoethano)phenanthrene in which the sterocenters at positions 4a, 10 and 10a have S-configuration. It is a prodrug of dextrorphan and used as an antitussive drug for suppressing cough. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Cross-References

ID SourceID
PubMed CID5360696
CHEMBL ID52440
CHEBI ID4470
CHEBI ID92579
SCHEMBL ID29949
MeSH IDM0006138

Synonyms (118)

Synonym
BRD-K33211335-337-03-7
(+)-3-methoxy-n-methylmorphinan
dextromethorphane
dextrometorfano
destrometerfano
ba 2666
benylin dm
balminil dm
(4as,10s,10as)-6-methoxy-11-methyl-1,3,4,9,10,10a-hexahydro-2h-10,4a-(epiminoethano)phenanthrene
dextromethorphanum
ba-2666
dextromethorfan
CHEBI:4470 ,
(9alpha,13alpha,14alpha)-3-methoxy-17-methylmorphinan
d-methorphan
calmylin
albutussin
3-methoxy-17-methyl-9alpha,13alpha,14alpha-morphinan
dextromorphan
romilar
dxm ,
(9alpha,13alpha,14alpha)-17-methyl-3-(methyloxy)morphinan
dextromethorphan (usp)
D03742
dextromethorfan [czech]
dextromethorphane [inn-french]
morphinan, 3-methoxy-17-methyl-, (9-alpha,13-alpha,14-alpha)-
dextrometorfano [inn-spanish]
einecs 204-752-2
destrometerfano [dcit]
dextromethorphan [usp:inn:ban]
brn 0088549
dextromethorphanum [inn-latin]
9alpha,13alpha,14alpha-morphinan, 3-methoxy-17-methyl-
9-alpha,13-alpha,14-alpha-morphinan, 3-methoxy-17-methyl-
morphinan, 3-methoxy-17-methyl-, (9.alpha.,13.alpha.,14.alpha.)-
NCGC00015333-01
lopac-d-2531
BSPBIO_000457
PRESTWICK2_000359
BPBIO1_000503
( )-3-methoxy-n-methylmorphinon
morphinan, 3-methoxy-17-methyl-, (9alpha,13alpha,14alpha)-
(+)-3-methoxy-17-methylmorphinan
( )-cis-1,3,4,9,10,10a-hexahydro-6-methoxy-11-methyl-2h-10,4alpha-iminoethanophenanthren
9alpha,13alpha,14alpha-morphinan, 3-methoxy-17-methyl- (8ci)
hsdb 3056
delta-methorphan
C06947
dextromethorphan
125-71-3
(+)-dextromethorphan
DB00514
SPBIO_002378
PRESTWICK0_000359
PRESTWICK1_000359
PRESTWICK3_000359
NCGC00015333-02
HMS2090C08
nsc-751452
CHEMBL52440
dextrometorphan
nsc 751452
(+)-3-methoxy-n-methylmorphinon
unii-7355x3rots
4-21-00-01367 (beilstein handbook reference)
dxm [antitussive]
(+)-cis-1,3,4,9,10,10a-hexahydro-6-methoxy-11-methyl-2h-10,4alpha-iminoethanophenanthren
7355x3rots ,
bdbm50366613
nodex
dextromethorphan [hsdb]
dextromethorphan [who-dd]
dextromethorphan [vandf]
dextromethorphan [inn]
dextromethorphan [mart.]
dextromethorphan [usp monograph]
dextromethorphan [usp-rs]
dextromethorphan [mi]
gtpl6953
AKOS025311415
SCHEMBL29949
3-methoxy-17-methyl-9.alpha.,13.alpha.,14.alpha.-morphinan
romilar (salt/mix)
tusilan (salt/mix)
9.alpha.,13.alpha.,14.alpha.-morphinan, 3-methoxy-17-methyl-
3-methoxy-17-methylmorphinan-, (9.alpha.,13.alpha.,14.alpha.)- #
3-methoxy-17-methyl-9.alpha.,13.alpha.-morphinan
racemethorphan [as d-form]
delsym (salt/mix)
medicon (salt/mix)
DTXSID3022908 ,
(1r,9r,10r)-4-methoxy-17-methyl-17-azatetracyclo[7.5.3.0^{1,10}.0^{2,7}]heptadeca-2,4,6-triene
(+)-3-methoxy-17-methyl-9alpha,13alpha,14alpha-morphinan
(1s,9s,10s)-4-methoxy-17-methyl-17-azatetracyclo[7.5.3.01,10.02,7]heptadeca-2(7),3,5-triene
J-005274
3-methoxy-17-methyl-9a,13a,14a-morphinan
(9a,13a,14a)-3-methoxy-17-methylmorphinan
lsm-2726
CHEBI:92579
4-methoxy-12-methyl-12-azatetracyclo[9.3^1.10^.0^2.7^] heptadeca-2(7),3,5-triene
Q407781
morphinan, 3-methoxy-17-methyl-, (9?,13?,14?)-
robotablets
soothe comfort ii cough syrupherbal mint
dextromethorphan (usp monograph)
dextromethorphan (usp:inn:ban)
dextrometorfano (inn-spanish)
dxm (antitussive)
dextromethorphane (inn-french)
dtxcid202908
soothe comfort i cough syruporiginal
dextromethorphan (usp-rs)
dextromethorphan (mart.)
dextromethorphanum (inn-latin)
dextromethorphanum (latin)
delsym (dextromethorphan)
dextromethorphan, 1mg/ml in methanol

Research Excerpts

Overview

Dextromethorphan (DXM) is an effective over-the-counter antitussive with an alarming increase as an abused drug for recreational purposes. It is a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist that has produced antidepressant-like effects in forced swim and tail suspension tests.

ExcerptReferenceRelevance
"Dextromethorphan polistirex is an extended-release formulation of dextromethorphan hydrobromide, marketed as Delsym® (Reckitt; Parsippany, NJ), with a duration of action roughly two to three times that of the standard formulation. "( Life-threatening pediatric dextromethorphan polistirex overdose.
Foreman, E; Friedland, S; Lasoff, DR; McDaniel, MA; Raviendran, R; Seltzer, JA; Sheth, SK; Toney, C, 2022
)
2.46
"Dextromethorphan is an over-the-counter antitussive drug, whereas levomethorphan is strictly controlled as a narcotic drug."( Chiral analysis of dextromethorphan and levomethorphan in human hair by liquid chromatography-tandem mass spectrometry.
Ji, JJ; Shen, M; Xiang, P; Yan, H; Zhao, J, 2022
)
1.77
"Dextromethorphan (DXM) is an effective over-the-counter antitussive with an alarming increase as an abused drug for recreational purposes. "( Targeting mediodorsal thalamic CB1 receptors to inhibit dextromethorphan-induced anxiety/exploratory-related behaviors in rats: The post-weaning effect of exercise and enriched environment on adulthood anxiety.
Alijanpour, S; Banaei-Boroujeni, G; Nazari-Serenjeh, F; Rezayof, A, 2023
)
2.6
"Dextromethorphan (DXM) is a safe and effective antitussive agent present in several over the counter cough and cold medications. "( Pharmacokinetics and pharmacodynamics of dextromethorphan: clinical and forensic aspects.
Dinis-Oliveira, RJ; Silva, AR, 2020
)
2.27
"Dextromethorphan is a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist that has produced antidepressant-like effects in forced swim and tail suspension tests (TST); however, the rapid and sustained antidepressant-like effects of dextromethorphan have not been evaluated."( Assessment of the rapid and sustained antidepressant-like effects of dextromethorphan in mice.
Garrett, PI; Hillhouse, TM; Honeycutt, SC; Peterson, AM; Saavedra, JS; White, JW, 2020
)
1.51
"Dextromethorphan acts as an antagonist at the glutamate N-methyl-d-aspartate (NMDA) receptor, in addition to other pharmacodynamics properties that include activity at sigma-1 receptors."( Dextromethorphan/quinidine pharmacotherapy in patients with treatment resistant depression: A proof of concept clinical trial.
Ahle, G; Charney, DS; Collins, KA; Iosifescu, DV; Kiraly, DD; Murrough, JW; Sayed, S; Soleimani, L; Wade, E; Welch, A, 2017
)
2.62
"Dextromethorphan (DM) is a dextrorotatory isomer of levorphanol, a typical morphine-like opioid. "( MK-801, but not naloxone, attenuates high-dose dextromethorphan-induced convulsive behavior: Possible involvement of the GluN2B receptor.
Chung, YH; Jang, CG; Jeong, JH; Kim, HC; Nabeshima, T; Nah, SY; Shin, EJ; Tran, HQ; Tran, TV; Yamada, K, 2017
)
2.15
"Dextromethorphan is a potent noncompetitive antagonist of the NMDA receptor channel that is safe for use in young girls with Rett syndrome. "( Randomized open-label trial of dextromethorphan in Rett syndrome.
Bibat, G; Brereton, N; Ewen, JB; Gupta, S; Hong, M; Johnston, MV; Kelley, R; Kratz, L; Naidu, S; Sanyal, A; Smith-Hicks, CL; Tierney, E; Vaurio, R; Yenokyan, G, 2017
)
2.18
"Dextromethorphan is a centrally acting antitussive drug, while its enantiomer levomethorphan is an illicit drug with opioid analgesic effects. "( Development of a capillary electrophoresis method for the determination of the chiral purity of dextromethorphan by a dual selector system using quality by design methodology.
Heuermann, M; Krait, S; Scriba, GKE, 2018
)
2.14
"Dextromethorphan is an antitussive with a high margin of safety that has been hypothesized to display rapid-acting antidepressant activity based on pharmacodynamic similarities to the N-methyl-D-aspartate (NMDA) receptor antagonist ketamine. "( Involvement of sigma-1 receptors in the antidepressant-like effects of dextromethorphan.
Healy, JR; Matsumoto, RR; Nguyen, L; Robson, MJ; Scandinaro, AL, 2014
)
2.08
"Dextromethorphan is an over-the-counter antitussive agent that may be a rapidly acting treatment for bipolar depression. "( The utility of the combination of dextromethorphan and quinidine in the treatment of bipolar II and bipolar NOS.
Kelly, TF; Lieberman, DZ, 2014
)
2.12
"Dextromethorphan (DXM) is a derivative of codeine with an antitussive properties. "( [Age and gender trends of inpatient recreational acute dextromethorphan intoxication hospitalized in Pomeranian Center of Toxicology between 2009-2011].
Schetz, D; Sein Anand, J; Zając, M, 2014
)
2.09
"Dextromethorphan (DM) is an antitussive with rapid acting antidepressant potential based on pharmacodynamic similarities to ketamine. "( Involvement of AMPA receptors in the antidepressant-like effects of dextromethorphan in mice.
Matsumoto, RR; Nguyen, L, 2015
)
2.1
"Dextromethorphan (DXM) is a widely available antitussive that has, at elevated dose levels, euphoric and dissociative effects. "( Patterns and Perceptions of Dextromethorphan Use in Adult Members of an Online Dextromethorphan Community.
Gabriel, KI; McDonald, MP; Pringle, G,
)
1.87
"Dextromethorphan (DM) is a commonly used antitussive and is currently the only FDA-approved pharmaceutical treatment for pseudobulbar affect. "( Dextromethorphan: An update on its utility for neurological and neuropsychiatric disorders.
Cavendish, JZ; Crowe, MS; Lucke-Wold, BP; Matsumoto, RR; Nguyen, L; Thomas, KL, 2016
)
3.32
"Dextromethorphan (DM) is a non-competitive antagonist of NMDA receptors and a widely used component of cough medicine. "( Prevention of Hippocampal Neuronal Damage and Cognitive Function Deficits in Vascular Dementia by Dextromethorphan.
Deng, J; Lu, K; Xu, X; Zhang, B; Zhao, BQ; Zhao, F; Zhao, Y, 2016
)
2.09
"Dextromethorphan (DXM) is an antitussive drug found in commonly used nonprescription cold and cough medications. "( Determination of Dextromethorphan in Oral Fluid by LC-MS-MS.
Amaratunga, P; Clothier, M; Lemberg, D; Lorenz Lemberg, B, 2016
)
2.22
"Dextromethorphan is a safe, effective cough suppressant, available without a prescription in the United States since 1958. "( Dextromethorphan: a case study on addressing abuse of a safe and effective drug.
Loyd, CM; Skor, EE; Spangler, DC, 2016
)
3.32
"Dextromethorphan is a cough suppressant agent with potential antidepressant activity reported in mouse force swimming test."( The role of NMDA receptor and nitric oxide/cyclic guanosine monophosphate pathway in the antidepressant-like effect of dextromethorphan in mice forced swimming test and tail suspension test.
Akbarian, R; Chamanara, M; Dehpour, AR; Khan, MI; Norouzi-Javidan, A; Ostadhadi, S; Sakhaee, E; Yousefi, F; Zolfaghari, S, 2017
)
1.38
"Dextromethorphan (DM) is a widely used antitussive reported to exert anti-inflammatory effect in vivo."( Effects of dextromethorphan on MDMA-induced serotonergic aberration in the brains of non-human primates using [
Chang, KW; Chen, CF; Cheng, CY; Chueh, SH; Hsu, TH; Huang, WS; Huang, YS; Liao, MH; Liu, TT; Ma, KH; Sung, CC; Weng, SJ, 2016
)
1.55
"Dextromethorphan is a widely available over-the-counter antitussive that produces intoxicating, hallucinogenic, and dissociative effects at doses significantly exceeding the therapeutic range. "( Five deaths resulting from abuse of dextromethorphan sold over the internet.
Goldfogel, G; Hamilton, R; Kuhlman, J; Logan, BK, 2009
)
2.07
"Dextromethorphan is a nonprescription antitussive which has been gaining in popularity as an abused drug, because of the hallucinogenic, dissociative, and intoxicating effects it produces at high doses. "( Combined dextromethorphan and chlorpheniramine intoxication in impaired drivers.
Logan, BK, 2009
)
2.21
"Dextromethorphan is a psychotropic substance that carries a potential for abuse and dependence. "( Dextromethorphan withdrawal and dependence syndrome.
Grosshans, M; Hermann, D; Kiefer, F; Koopmann, A; Mann, K; Mutschler, J, 2010
)
3.25
"Dextromethorphan (DEX) is a widely used non-opioid antitussive. "( Dextromethorphan inhibits the glutamatergic synaptic transmission in the nucleus tractus solitarius of guinea pigs.
Haji, A; Ohi, Y; Tsunekawa, S, 2011
)
3.25
"Dextromethorphan (DEX) is an antitussive agent used in many cough and cold medications, and dextrorphan (DOR) is its metabolite. "( Simultaneous determination of dextromethorphan and its metabolite dextrorphan in plasma samples using second-order calibration coupled with excitation-emission matrix fluorescence.
Liu, YJ; Nie, JF; Ouyang, LQ; Wang, JY; Wu, HL; Yu, RQ, 2011
)
2.1
"Dextromethorphan (DM) is a dextrorotary morphinan and a widely used component of cough medicine. "( Low dose dextromethorphan attenuates moderate experimental autoimmune encephalomyelitis by inhibiting NOX2 and reducing peripheral immune cells infiltration in the spinal cord.
Chechneva, OV; Daugherty, DJ; Deng, W; Hong, JS; Mayrhofer, F; Pleasure, DE, 2011
)
2.23
"Dextromethorphan is a low-affinity N-methyl-D-aspartate receptor inhibitor."( Comparative effects of triflusal, S-adenosylmethionine, and dextromethorphan over intestinal ischemia/reperfusion injury.
Alarcón-Galván, G; Cámara-Lemarroy, CR; Cordero-Pérez, P; Fernández-Garza, NE; Guzmán-de la Garza, FJ; Muñoz-Espinosa, LE; Torres-Gonzalez, L, 2011
)
1.33
"Dextromethorphan (DM) is a well-known probe drug for CYP2D6 and metabolic ratio (MR) is often used to measure the enzyme activity in vivo."( Inter-individual variability of in vivo CYP2D6 activity in different genotypes.
Chiba, K; Ito, T; Kato, M; Sugiyama, Y; Suwa, T, 2012
)
1.1
"Dextromethorphan (DM) is a well-known antitussive dextrorotatory morphinan. "( Dextromethorphan-induced psychotoxic behaviors cause sexual dysfunction in male mice via stimulation of σ-1 receptors.
Bach, JH; Chung, YH; Jeong, JH; Kim, HC; Kim, KW; Kwon, YB; Li, Z; Nabeshima, T; Nam, Y; Park, ES; Shin, EJ; Yang, BK, 2012
)
3.26
"Dextromethorphan (DM) is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist that may be neuroprotective for monoamine neurons. "( The DRD2/ANKK1 gene is associated with response to add-on dextromethorphan treatment in bipolar disorder.
Chang, YH; Chen, SH; Chen, SL; Chu, CH; Huang, SY; Lee, IH; Lee, SY; Lu, RB; Tzeng, NS; Wang, CL; Yang, YK; Yeh, TL, 2012
)
2.07
"Dextromethorphan is a commonly encountered antitussive medication which has found additional therapeutic use in the treatment of pseudobulbar disorder and as an adjunct to opiate use in pain management. "( Dextromethorphan abuse leading to assault, suicide, or homicide.
Frost, MP; Goldfogel, G; Logan, BK; Sandstrom, G; Wickham, DJ; Yeakel, JK, 2012
)
3.26
"Dextromethorphan is a noncompetitive NMDA-receptor antagonist."( Dextromethorphan for phantom pain attenuation in cancer amputees: a double-blind crossover trial involving three patients.
Ben Abraham, R; Kollender, Y; Marouani, N; Meller, I; Weinbroum, AA,
)
2.3
"Dextromethorphan is a widely used anti-tussive drug with non-competitive antagonistic effects on excitatory amino acid receptors of the N-methyl-D-aspartate (NMDA) type. "( Dextromethorphan alters gene expression in rat brain hippocampus and cortex.
Ahn, JI; Kim, SH; Koh, HC; Lee, KH; Lee, YS; Yang, BH; Yu, DH, 2003
)
3.2
"Dextromethorphan is an N-methyl-D-aspartic acid antagonist which can attenuate acute pain with few side-effects. "( Dextromethorphan and intrathecal morphine for analgesia after Caesarean section under spinal anaesthesia.
Choi, DM; Douglas, MJ; Kliffer, AP, 2003
)
3.2
"Dextromethorphan is an effective and safe antitussive, but has liabilities with respect to its abuse potential at doses above the therapeutic dose. "( Determination of dextromethorphan and its metabolites in rat serum by liquid-liquid extraction and liquid chromatography with fluorescence detection.
Gurley, BJ; Hendrickson, HP; Wessinger, WD, 2003
)
2.1
"Dextromethorphan is a weak noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist. "( Comparison of the effects of dextromethorphan, dextrorphan, and levorphanol on the hypothalamo-pituitary-adrenal axis.
Pechnick, RN; Poland, RE, 2004
)
2.06
"Dextromethorphan is an N-methyl-D-aspartate (NMDA) noncompetitive antagonist which has been used as an antitussive, analgesic adjunct, probe drug, experimentally to attenuate acute opiate and ethanol withdrawal, and as an anticonvulsant. "( Plasma profile and pharmacokinetics of dextromethorphan after intravenous and oral administration in healthy dogs.
Kukanich, B; Papich, MG, 2004
)
2.04
"Dextromethorphan is an over-the-counter dissociative agent of increasing popularity as a drug of abuse among younger adolescents. "( Dextromethorphan abuse.
Boyer, EW, 2004
)
3.21
"Dextromethorphan (DM) is a dextrorotatory morphinan and an over-the-counter non-opioid cough suppressant. "( Protective effect of dextromethorphan against endotoxic shock in mice.
Block, ML; Cui, G; Hong, JS; Li, G; Liu, B; Liu, J; Liu, Y; Qin, L; Tzeng, NS; Wang, T; Wilson, B, 2005
)
2.09
"Dextromethorphan is a commonly used antitussive agent that can be purchased over the counter. "( Dextromethorphan-induced delirium and possible methadone interaction.
Lotrich, FE; Pollock, BG; Rosen, J, 2005
)
3.21
"Dextromethorphan is an opiod-derived, easily available cough remedy that, when used in large quantities, can have stimulatory effects which mimic that of amphetamine and other psychedelic drugs. "( Dextromethorphan abuse in Thai adolescents: A report of two cases and review of literature.
Chomchai, C; Manaboriboon, B, 2005
)
3.21
"Dextromethorphan (DXM) is a cough-suppressing ingredient in a variety of over-the-counter cough and cold medications. "( Effects of dextromethorphan on in vitro contractile responses of mouse and rat urinary bladders.
Levin, RM; Millington, WR; Sourial, MW; Tadrous, M; Whitbeck, C, 2006
)
2.17
"Dextromethorphan (DM) is a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist, which is widely used as an antitussive agent. "( Dextromethorphan: a review of N-methyl-d-aspartate receptor antagonist in the management of pain.
Drachtman, R; Siu, A, 2007
)
3.23
"Dextromethorphan (DXM) is a common component of combination cold medications that has become a popular drug of abuse for young adults. "( "Crystal dex:" free-base dextromethorphan.
Cloutier, RL; Hendrickson, RG, 2007
)
2.09
"Dextromethorphan is a synthetic analogue of codeine used in hundreds of over-the-counter medications for its antitussive effects. "( Dextromethorphan in Wisconsin drivers.
Cochems, A; Harding, P; Liddicoat, L, 2007
)
3.23
"Dextromethorphan is a widely used antitussive agent, also showing increased recreational abuse. "( Oral administration of dextromethorphan does not produce neuronal vacuolation in the rat brain.
Carliss, RD; Chengelis, CP; O'Neill, TP; Radovsky, A; Shuey, DL, 2007
)
2.09
"Dextromethorphan (DXM) is a common ingredient in several prescriptions and over-the-counter cough preparations. "( Dextromethorphan-induced near-fatal suicide attempt in a slow metabolizer at cytochrome P450 2D6.
Kennedy, CA; Kurani, A; Liu, QY; Matin, N, 2007
)
3.23
"Dextromethorphan (DM) is a widely-used antitussive. "( Dextromethorphan as a potential neuroprotective agent with unique mechanisms of action.
Calef, U; Lauterbach, EC; Werling, LL, 2007
)
3.23
"Dextromethorphan (DEM) is a widely used probe drug for human cytochrome P450 2D6 isozyme activity assessment by measuring the ratio between DEM and its N-demethylated metabolite dextrorphan (DOR). "( Development and validation of a chemical hydrolysis method for dextromethorphan and dextrophan determination in urine samples: application to the assessment of CYP2D6 activity in fibromyalgia patients.
Cherkaoui, S; Daali, Y; Dayer, P; Desmeules, JA; Doffey-Lazeyras, F, 2008
)
2.03
"Dextromethorphan (DMP) is an effective and widely used antitussive drug. "( An evaluation of the genotoxicity of the antitussive drug Dextromethorphan.
Aardema, MJ; Gatehouse, D; Johnston, G; Robison, SH, 2008
)
2.03
"Dextromethorphan (DM) is a low-affinity, non-competitive NMDA receptor antagonist that has shown promise in preclinical and preliminary clinical studies for the reduction of opioid withdrawal symptoms, but when used at higher doses, it is associated with deleterious side effects attributed to its metabolite, dextrorphan. "( Dextromethorphan and quinidine combination for heroin detoxification.
Akerele, E; Bisaga, A; Comer, SD; Garawi, F; Kleber, HD; Nunes, EV; Sullivan, MA; Thomas, AA,
)
3.02
"Dextromethorphan is a dextrorotary morphinan without affinity for opioid receptors, commonly used as an antitussive medication. "( Dextromethorphan: cellular effects reducing neuronal hyperactivity.
Netzer, R; Trube, G, 1994
)
3.17
"1. Dextromethorphan is a widely used antitussive agent which is a non-narcotic codeine analogue. "( The effect of inhaled and oral dextromethorphan on citric acid induced cough in man.
Grattan, TJ; Higgins, KS; Marshall, AE; Morice, AH, 1995
)
1.2
"Dextromethorphan is a weak noncompetitive N-methyl-D-aspartate (NMDA) antagonist and higher doses or other potent NMDA receptor antagonists should be tested."( A controlled one-year trial of dextromethorphan in amyotrophic lateral sclerosis.
Azulay, JP; Besse, D; Billé-Turc, F; Blin, O; Braguer, D; Branger, E; Crevat, A; Desnuelle, C; Pouget, JY; Serratrice, G, 1996
)
1.3
"Dextromethorphan is a widely used antitussive agent with non-competitive antagonistic effects at the excitatory amino acid receptors of the NMDA type. "( Dextromethorphan reduces intravenous cocaine self-administration in the rat.
Balducci, C; Koob, GF; Pulvirenti, L, 1997
)
3.18
"Dextromethorphan is a NMDA-glutamate receptor antagonist with neuroprotective properties."( A clinical trial of dextromethorphan in amyotrophic lateral sclerosis.
Bak, S; Boysen, G; Christensen, PB; Gredal, O; Hinge, HH; Jensen, TS; Jespersen, JH; Kristensen, MO; Regeur, L; Werdelin, L, 1997
)
1.34
"Dextromethorphan is a noncompetitive antagonist of this receptor with a favorable safety profile."( The effect of the N-methyl-D-aspartate receptor antagonist dextromethorphan on perioperative brain injury in children undergoing cardiac surgery with cardiopulmonary bypass: results of a pilot study.
Arbenz, U; Bandtlow, C; Bauersfeld, U; Baumann, P; Boltshauser, E; Fanconi, S; Huisman, TA; Martin, E; Molinari, L; Schmid, ER; Schmitt, B; Superti-Furga, A; Turina, M; Wohlrab, G, 1997
)
1.26
"Dextromethorphan is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist known to inhibit wind-up and central hyperexcitability of dorsal horn neurones. "( Effect of systemic N-methyl-D-aspartate receptor antagonist (dextromethorphan) on primary and secondary hyperalgesia in humans.
Brennum, J; Dahl, JB; Dirks, J; Ilkjaer, S; Wernberg, M, 1997
)
1.98
"Dextromethorphan is a nonopioid antitussive metabolized by cytochrome P450 2D6 (CYP2D6) to an active metabolite, dextrorphan. "( Psychotropic effects of dextromethorphan are altered by the CYP2D6 polymorphism: a pilot study.
Busto, UE; Kaplan, HL; Sellers, EM; Tyndale, RF; Zawertailo, LA, 1998
)
2.05
"Dextromethorphan is an N-methyl-D-aspartate (NMDA) receptor antagonist which has been shown to inhibit the development of cutaneous secondary hyperalgesia after tissue trauma. "( Dextromethorphan and pain after total abdominal hysterectomy.
Campbell, WI; McCaughey, W; McConaghy, PM; McSorley, P, 1998
)
3.19
"Dextromethorphan 1 is an effective neuroprotectant in animal models of epilepsy and ischemia but showed side-effects during clinical trials limiting its potential use in a clinical setting. "( Synthesis and preliminary biological evaluation of new alpha-amino amide anticonvulsants incorporating a dextromethorphan moiety.
Bonsignori, A; Caccia, C; Maj, R; McArthur, RA; Pevarello, P; Salvati, P; Traquandi, G; Varasi, M, 1999
)
1.96
"Dextromethorphan is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist known to inhibit wind-up and NMDA-mediated nociceptive responses of dorsal horn neurons. "( Effect of preoperative oral dextromethorphan on immediate and late postoperative pain and hyperalgesia after total abdominal hysterectomy.
Bach, LF; Dahl, JB; Ilkjaer, S; Nielsen, PA; Wernberg, M, 2000
)
2.04
"Dextromethorphan is a weak N-methyl-d-aspartate (NMDA) receptor antagonist that inhibits spinal cord sensitization in animal models of pain and also inhibits the development of cutaneous secondary hyperalgesia after tissue trauma. "( Large-dose oral dextromethorphan as an adjunct to patient-controlled analgesia with morphine after knee surgery.
Clarke, D; Goodchild, CS; Wadhwa, A; Young, D, 2001
)
2.1
"Dextromethorphan syrup is a safe, non-narcotic medication that significantly reduced the requirement of intravenous morphine after pediatric adenotonsillectomy. "( Improved postoperative pain control in pediatric adenotonsillectomy with dextromethorphan.
Dawson, GS; Ramadan, HH; Seidman, P, 2001
)
1.98
"Dextromethorphan (DXM) is a widely used probe drug for human CYP2D6 activity both in vitro and in vivo. "( Comparative contribution to dextromethorphan metabolism by cytochrome P450 isoforms in vitro: can dextromethorphan be used as a dual probe for both CTP2D6 and CYP3A activities?
Haining, RL; Yu, A, 2001
)
2.05
"Dextromethorphan (DXM) is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist shown to prevent the development of tolerance to the antinociceptive effects of morphine in rodents. "( Dextromethorphan potentiates the antinociceptive effects of morphine and the delta-opioid agonist SNC80 in squirrel monkeys.
Allen, RM; Dykstra, LA; Granger, AL, 2002
)
3.2
"Dextromethorphan is a probe substrate to determine CYP2D6 phenotype. "( Development of a rapid and sensitive high-performance liquid chromatographic method to determine CYP2D6 phenotype in human liver microsomes.
March, C; Rege, B; Sarkar, MA, 2002
)
1.76
"Dextromethorphan (DM) is a neuroprotective agent. "( Nasal delivery of [14C]dextromethorphan hydrochloride in rats: levels in plasma and brain.
Behl, CR; Char, H; Iqbal, K; Kumar, S; Malick, AW; Patel, S; Piemontese, D; Salvador, RA, 1992
)
2.04
"Dextromethorphan is a highly effective and widely used nonopioid antitussive drug. "( Dextromethorphan. An overview of safety issues.
Bem, JL; Peck, R,
)
3.02
"Dextromethorphan (DM) is an antitussive with anticonvulsant activity that binds to high- and low-affinity sites in guinea pig brain homogenates. "( Autoradiographic localization of [3H]dextromethorphan in guinea pig brain: allosteric enhancement by ropizine.
Canoll, PD; Gottesman, S; Musacchio, JM; Smith, PR, 1989
)
1.99

Effects

Dextromethorphan is a weak antagonist of the ion channel associated with the NMDA receptor. It has been used as an antitussive for more than 40 years. recreational use of high doses has been found to cause mania and hallucinations.

ExcerptReferenceRelevance
"Dextromethorphan (DXM) has been re-purposed several times over the past 7 decades: first as a cough suppressant, then as a compounded formulation with quinidine for treatment of pseudobulbar affect, and most recently as a compounded formulation with bupro-pion for treatment of major depressive disorder. "( Dextromethorphan: From Cough Suppressant to Antidepressant.
Kverno, K, 2022
)
3.61
"Dextromethorphan has been used as an abortive and prophylactic treatment for MTX-NT."( Dextromethorphan Administration on Day 0 and Day 7 for Secondary Prevention of Methotrexate-induced Neurotoxicity in Childhood Acute Lymphoblastic Leukemia: A Retrospective Case Series.
Fustino, NJ; Juhl, K; Leister, J, 2021
)
2.79
"Dextromethorphan (DM) has been reported to possess neuroprotective effects in lipopolysaccharide- (LPS-) stimulated animals; however, it remains unclear whether epigenetic regulatory mechanisms in microglial cells are involved in such DM-mediated neuroprotective effects."( Dextromethorphan Suppresses Lipopolysaccharide-Induced Epigenetic Histone Regulation in the Tumor Necrosis Factor-
Kuo, KC; Wu, PL; Yang, CH; Yang, SN; Yang, YN; Yang, YSH, 2020
)
2.72
"Dextromethorphan (DXM) has overtaken codeine as the most widely used cough suppressant due to its availability, efficacy, and safety profile at directed doses."( Dextromethorphan in Cough Syrup: The Poor Man's Psychosis.
Birur, B; Black, JR; Bolis, RA; Fargason, RE; Martinak, B, 2017
)
2.62
"Dextromethorphan and memantine have been administered to animals after spinal nerve ligation (SNL) to evaluate their antinociceptive/cognitive effects and associated molecular events, including the phosphorylation of several tyrosine (pTyr(1336), pTyr(1472)) residues in the NR2B NMDAR subunit."( Low doses of dextromethorphan have a beneficial effect in the treatment of neuropathic pain.
Chalus, M; Daulhac, L; Dupuis, A; Eschalier, A; Etienne, M; Morel, V; Pickering, G; Privat, AM, 2014
)
1.49
"Dextromethorphan has been reported to ameliorate opioid withdrawal symptoms in both animal and human subjects. "( A double-blind, placebo-controlled trial of dextromethorphan combined with clonidine in the treatment of heroin withdrawal.
Chen, CH; Lin, SK; Pan, CH, 2014
)
2.11
"Dextromethorphan (DM) has been used for more than 50years as an over-the-counter antitussive. "( Pharmacology of dextromethorphan: Relevance to dextromethorphan/quinidine (Nuedexta®) clinical use.
Matsumoto, RR; Pope, LE; Siffert, J; Taylor, CP; Traynelis, SF, 2016
)
2.22
"Dextromethorphan (DM) has been well-characterized as a neuroprotective agent in experimental models of CNS injury. "( Neuroprotective profile of dextromethorphan in an experimental model of penetrating ballistic-like brain injury.
Lu, XC; Sharrow, K; Shear, DA; Tortella, FC; Williams, AJ, 2009
)
2.09
"Dextromethorphan (DXM) has unique toxicity that may be difficult to diagnose. "( Dextromethorphan abuse masquerading as a recurrent seizure disorder.
Ali, SS; Lee, DC; Majlesi, N, 2011
)
3.25
"Dextromethorphan (DM) has been shown to protect against endotoxic shock in mice. "( Attenuating heat-induced acute lung inflammation and injury by dextromethorphan in rats.
Chang, CP; Hou, CC; Lin, MT; Yang, HH, 2012
)
2.06
"Dextromethorphan at high doses has phencyclidine-like effects on the NMDA receptor system; recreational use of high doses has been found to cause mania and hallucinations."( Dextromethorphan abuse leading to assault, suicide, or homicide.
Frost, MP; Goldfogel, G; Logan, BK; Sandstrom, G; Wickham, DJ; Yeakel, JK, 2012
)
2.54
"Dextromethorphan has been reported to decrease the self-administration of several drugs of abuse, including morphine, methamphetamine, cocaine, and nicotine. "( Effects of dextromethorphan on dopamine release in the nucleus accumbens: Interactions with morphine.
Glick, SD; Maisonneuve, IM; Steinmiller, CL, 2003
)
2.15
"Dextromethorphan has moderate affinities for phencyclidine sites, while dimemorfan has very low affinities for such sites, suggesting that these sites are not essential for the anticonvulsant actions of dimemorfan."( The dextromethorphan analog dimemorfan attenuates kainate-induced seizures via sigma1 receptor activation: comparison with the effects of dextromethorphan.
Cha, JY; Chen, CF; Jhoo, WK; Kim, HC; Kim, WK; Ko, KH; Lim, YK; Nah, SY; Shin, EJ, 2005
)
1.61
"Dextromethorphan has been used as an antitussive for more than 40 years and is considered a drug with a good margin of safety. "( Dextromethorphan, 3-methoxymorphinan, and dextrorphan have local anaesthetic effect on sciatic nerve blockade in rats.
Chen, YW; Hou, CH; Lin, CN; Lin, MT; Tu, CH; Tzeng, JI; Wang, JJ, 2006
)
3.22
"Dextromethorphan has been reported to be a weak antagonist of the ion channel associated with the NMDA receptor, and to have putative antiparkinsonian activity in man. "( Antiparkinsonian action of dextromethorphan in the reserpine-treated mouse.
Kaur, S; Starr, MS, 1995
)
2.03
"Dextromethorphan has been shown to protect against ischemic tissue damage. "( Dextromethorphan attenuates the effects of ischemia on rabbit electroretinographic oscillatory potentials.
Brunette, JR; Cao, W; Drumheller, A; Jolicoeur, FB; Lafond, G; Zaharia, M, 1993
)
3.17
"Dextromethorphan has analgesic efficacy in the experimental formalin test, blocks the nociceptive activation of the immediate-early gene, c-fos proto-oncogene, and prevents and reverses the development of opiate analgesic tolerance in experimental models."( Dextromethorphan shows efficacy in experimental pain (nociception) and opioid tolerance.
Brodsky, M; Elliott, KJ; Foley, KM; Hyanansky, A; Inturrisi, CE, 1995
)
2.46
"Dextromethorphan (DM) has been observed to afford neuroprotection in a variety of in vitro and in vivo experimental models of CNS injury. "( Dextromethorphan protects against cerebral injury following transient, but not permanent, focal ischemia in rats.
Britton, P; Laskosky, MS; Lu, XC; Tortella, FC, 1997
)
3.18
"Dextromethorphan has been on the market for many years and has never been implicated as a human teratogen."( Dextromethorphan. Extrapolation of findings from reproductive studies in animals to humans.
Einarson, A; Koren, G, 1999
)
2.47
"Dextromethorphan has been proposed as a test compound to assess drug oxidation polymorphism."( Effect of liver disease on dextromethorphan oxidation capacity and phenotype: a study in 107 patients.
Amouyal, G; Babany, G; Freneaux, E; Habersetzer, F; Larrey, D; Letteron, P; Pessayre, D; Tinel, M, 1989
)
1.3

Actions

Dextromethorphan produced lower ED(50) values for morphine, fentanyl and sufentanil but exerted no effect on the potency of SNC80 or U50,488H. The drug can produce serotonin syndrome in the absence of another serotonergic drug.

ExcerptReferenceRelevance
"Dextromethorphan produced lower ED(50) values for morphine, fentanyl and sufentanil but exerted no effect on the potency of SNC80 or U50,488H."( Dextromethorphan and ketamine potentiate the antinociceptive effects of mu- but not delta- or kappa-opioid agonists in a mouse model of acute pain.
Baker, AK; Hoffmann, VL; Meert, TF, 2002
)
2.48
"Dextromethorphan produced lower ED50 values for morphine, fentanyl and sufentanil but exerted no effect on SNC80 or U50,488H."( Interactions of NMDA antagonists and an alpha 2 agonist with mu, delta and kappa opioids in an acute nociception assay.
Baker, AK; Hoffmann, VL; Meert, TF, 2002
)
1.04
"Dextromethorphan can produce psychoactive effects similar to that of marijuana, and higher doses will produce dissociative effects, including sensory enhancement and hallucinations."( Dextromethorphan in Wisconsin drivers.
Cochems, A; Harding, P; Liddicoat, L, 2007
)
2.5
"Dextromethorphan can produce serotonin syndrome in the absence of another serotonergic drug."( Serotonin syndrome in dextromethorphan ingestion responsive to propofol therapy.
Babu, KM; Boyer, EW; Ganetsky, M, 2007
)
1.38

Treatment

Dextromethorphan treatment led to a significant but modest reduction in morphine requirements (29.3% P < 0.05) but no reduction in postoperative pain levels. Dextromethmorphan-pretreated animals required a significantly larger dose of PTZ than did controls to produce the first myoclonic jerk.

ExcerptReferenceRelevance
"The dextromethorphan-treated rats were given 12, 24 and 48 mg/kg (low, medium, high) of dextromethorphan by intragastric administration each day for 3 days."( Serum metabolic changes in rats after intragastric administration of dextromethorphan.
Bao, S; Chen, L; Hong, L; Hu, L; Lin, G; Lin, Z; Mo, J; Qian, S; Su, K; Sun, F; Wang, X; Wen, C; Wu, Q; Zhang, J, 2017
)
1.17
"Dextromethorphan pretreatment significantly suppressed the production of tumour necrosis factor-alpha, monocyte chemoattractant protein-1, interleukin-6, interleukin-10, and superoxide in macrophage cell culture after stimulation."( Dextromethorphan reduces oxidative stress and inhibits atherosclerosis and neointima formation in mice.
Hong, JS; Huang, CW; Jiang, SJ; Li, YH; Liu, PY; Liu, SL; Shi, GY; Tang, SH; Wu, HL, 2009
)
2.52
"Dextromethorphan treatment induced a slight inhibition of peak currents in human K(v)1.2 and K(v)1.3 channels, whereas dextromethorphan profoundly inhibited the steady-state currents of human K(v)1.3 channels compared to K(v)1.2 channel currents."( Effect of dextromethorphan on human K(v)1.3 channel activity: involvement of C-type inactivation.
Choi, SH; Hwang, SH; Kim, HC; Lee, BH; Lee, JH; Nah, SY; Shin, TJ, 2011
)
1.49
"Dextromethorphan treatment led to a significant but modest reduction in morphine requirements (29.3% P < 0.05) but no reduction in postoperative pain levels."( Large-dose oral dextromethorphan as an adjunct to patient-controlled analgesia with morphine after knee surgery.
Clarke, D; Goodchild, CS; Wadhwa, A; Young, D, 2001
)
1.38
"Dextromethorphan-treated patients reported significantly (p < 0.05) less pain and sedation, and felt better."( Combined pre-incisional oral dextromethorphan and epidural lidocaine for postoperative pain reduction and morphine sparing: a randomised double-blind study on day-surgery patients.
Ben-Abraham, R; Flaishon, R; Lalayev, G; Niv, D; Weinbroum, AA; Yashar, T, 2001
)
1.32
"Dextromethorphan-treated animals (n = 10) showed less lactate formation during ischemia than untreated animals (n = 11, p less than 0.001)."( Effects of dextromethorphan on rat brain during ischemia and reperfusion assessed by magnetic resonance spectroscopy.
Balázs, R; Lewis, P; Nasim, MM; Tulleken, CA; van Echteld, CJ; van Rijen, PC; Verheul, HB, 1991
)
1.39
"Dextromethorphan-pretreated animals required a significantly larger dose of PTZ than did controls to produce the first myoclonic jerk, but a significantly smaller dose of the convulsant to produce maximal seizures."( Effect of orally administered dextromethorphan on theophylline- and pentylenetetrazol-induced seizures in rats.
Levy, G; Zhi, JG, 1990
)
1.29
"The dextromethorphan-treated rabbits also demonstrated significantly smaller areas of cortical edema (10.2%) on magnetic resonance imaging than the controls (38.6%, p less than 0.01)."( Dextromethorphan protects against cerebral injury following transient focal ischemia in rabbits.
DeLaPaz, R; George, CP; Gross, T; Shibata, DK; Steinberg, GK, 1988
)
2.2
"Treatment of dextromethorphan 1 with various alkylating agents followed by base treatment led to Hoffman-type elimination reactions to produce a series of tricyclic derivatives, 6. "( Synthesis of potent sigma-1 receptor ligands via fragmentation of dextromethorphan.
Arrington, MP; Brown, C; Schwartz, CE, 2004
)
0.93
"Treatment of dextromethorphan (32 mg/kg) alone or in combination with CYP2D6 enzyme inhibitor Quinidine (32 mg/kg) failed to produce rapid antidepressant-like effects by increasing the latency to drink in the NIH test rather than decreasing the latency to drink."( Assessment of the rapid and sustained antidepressant-like effects of dextromethorphan in mice.
Garrett, PI; Hillhouse, TM; Honeycutt, SC; Peterson, AM; Saavedra, JS; White, JW, 2020
)
1.15
"Treatment with Dextromethorphan/Quinidine (DM/Q) has demonstrated benefit on pseudobulbar affect and bulbar function in amyotrophic lateral sclerosis (ALS). "( Effect of one-year dextromethorphan/quinidine treatment on management of respiratory impairment in amyotrophic lateral sclerosis.
Burés, E; Ferrer, S; Luis Díaz, J; Sancho, J; Servera, E; Signes-Costa, J; Torrecilla, T, 2021
)
1.3
"Pretreatment with dextromethorphan (20 mg/kg s.c., 20 min prior) potentiated the effects of acute morphine, while attenuating the effects of chronic morphine on nucleus accumbens DA levels."( Effects of dextromethorphan on dopamine release in the nucleus accumbens: Interactions with morphine.
Glick, SD; Maisonneuve, IM; Steinmiller, CL, 2003
)
1.03
"Pretreatment with dextromethorphan at 60 mg/kg s.c., 30 min prior to formalin resulted in a suppression of c-fos induction, so that c-fos mRNA levels in the ipsilateral spinal dorsal horn of animals receiving dextromethorphan prior to formalin did not differ from controls."( Dextromethorphan suppresses both formalin-induced nociceptive behavior and the formalin-induced increase in spinal cord c-fos mRNA.
Brodsky, M; Elliott, KJ; Foley, KM; Hynansky, AD; Inturrisi, CE, 1995
)
2.06
"Treatment with dextromethorphan, an NMDA receptor antagonist, was started between 48 hours and 14 days after the critical incident."( Drug refractory epilepsy in brain damage: effect of dextromethorphan on EEG in four patients.
Baumann, P; Boltshauser, E; Fanconi, S; Netzer, R; Schmitt, B, 1994
)
0.88
"Treatment with dextromethorphan or levemopamil reduced the deficit in spatial learning by limiting the increase in swim distance due to ischemia."( Global ischemic neuronal damage relates to behavioural deficits: a pharmacological approach.
Block, F; Schwarz, M, 1998
)
0.64
"Pretreatment with dextromethorphan was associated with a significant decrease in the concentrations of theophylline in the cerebrospinal fluid and serum at the pharmacologic endpoint."( Effect of orally administered dextromethorphan on theophylline- and pentylenetetrazol-induced seizures in rats.
Levy, G; Zhi, JG, 1990
)
0.89

Toxicity

Dextromethorphan (DM) and its metabolite dextrorphan (DX) have antiepileptic and neuroprotective properties that we evaluated in mice against 2 LD50 of soman in a three-drug pretreatment. DM is widely used as a probe drug for CYP2D6 phenotyping and for the assessment of CYP1D6 activity. We sought to characterize adverse events associated with dextromethmorphan in children <12 years old.

ExcerptReferenceRelevance
" An anthology of adverse drug events has been analysed, drawn both from published case records and a data base recording dextromethorphan-related adverse events spontaneously reported by physicians or pharmacists."( Dextromethorphan. An overview of safety issues.
Bem, JL; Peck, R,
)
1.78
" A literature review, brief review of pharmacology, and report of two cases of adverse reactions to dextromethorphan-containing preparations are presented."( Toxicity with dextromethorphan-containing preparations: a literature review and report of two additional cases.
Parks, BR; Pender, ES, 1991
)
0.86
" They were given individually to guinea pigs prior to poisoning with 2 x LD50 soman to test their efficacy against organophosphorus-induced convulsions, brain damage, and lethality."( Prevention of soman neurotoxicity by non-opioid antitussives.
Braitman, DJ; Brennecke, LH; Sparenborg, S, 1990
)
0.28
"The anticonvulsant and adverse effects of dextromethorphan, a non-opioid antitussive, and its metabolite dextrorphan were examined in amygdala-kindled rats."( Differences in anticonvulsant potency and adverse effects between dextromethorphan and dextrorphan in amygdala-kindled and non-kindled rats.
Hönack, D; Löscher, W, 1993
)
0.79
" All symptoms were reversible and no patient suffered severe adverse reactions."( Dose escalation safety and tolerance study of the N-methyl-D-aspartate antagonist dextromethorphan in neurosurgery patients.
Bell, TE; Steinberg, GK; Yenari, MA, 1996
)
0.52
"Extracellular levels of endogenous glutamate are relatively high in the developing rabbit retina but nonetheless appear to promote cell survival and developmental processes at concentrations considered toxic in the adult."( N-methyl-D-aspartate-mediated glutamate toxicity in the developing rabbit retina.
Haberecht, MF; Lo, GJ; Mitchell, CK; Redburn, DA, 1997
)
0.3
" Slow DM metabolizers took significantly lower daily doses of MorphiDex than rapid metabolizers without a significant difference in the incidence of adverse events."( Long-term safety of MorphiDex.
Goldblum, R, 2000
)
0.31
"In this study, we have focused our investigation of the facts whether co-administration of a NMDA antagonist dextromethorphan (DM) with morphine during pregnancy and throughout lactation could prevent the adverse effects associated with chronic morphine administration in rat offspring."( Co-administration of dextromethorphan during pregnancy and throughout lactation significantly decreases the adverse effects associated with chronic morphine administration in rat offspring.
Su, CH; Tao, PL; Wu, YH; Yeh, GC, 2001
)
0.84
" Hcy is directly toxic to vascular endothelium and it and its metabolites are excitatory agonists of the N-methyl-D-aspartate (NMDA) receptor."( Dextromethorphan is effective in the treatment of subacute methotrexate neurotoxicity.
Aisner, J; Cole, PD; Drachtman, RA; Golden, CB; James, SJ; Kamen, BA; Melnyk, S,
)
1.57
" Participants in the DM group reported many more adverse events than did subjects on placebo (173 vs."( A randomized, double-blind, placebo-controlled safety study of high-dose dextromethorphan in methadone-maintained male inpatients.
Bead, V; Caruso, FS; Childress, AR; Cornish, JW; Ehrman, RN; Esmonde, CA; Herman, BH; Martz, K; O'Brien, CP; Poole, S; Robbins, SJ, 2002
)
0.55
" However, there have been reports of patients experiencing adverse events with related agents."( Safety of selegiline with cold medications.
Jacob, JE; Sage, JI; Wagner, ML, 2003
)
0.32
" If selegiline is used with these medications, watch for adverse events or replace selegiline with another drug."( Safety of selegiline with cold medications.
Jacob, JE; Sage, JI; Wagner, ML, 2003
)
0.32
"Dextromethorphan (DEM) shares part of the adverse event profile of opioids and is widely used as a probe drug for CYP2D6 phenotyping and for the assessment of CYP2D6 activity."( Measurement of CYP2D6 and CYP3A4 activity in vivo with dextromethorphan: sources of variability and predictors of adverse effects in 419 healthy subjects.
Becquemont, L; Boëlle, PY; Bornert, C; Funck-Brentano, C; Poirier, JM; Verstuyft, C, 2005
)
2.02
" Logistic regression was performed to examine the factors associated with changes in metabolic ratios and with the occurrence of adverse events."( Measurement of CYP2D6 and CYP3A4 activity in vivo with dextromethorphan: sources of variability and predictors of adverse effects in 419 healthy subjects.
Becquemont, L; Boëlle, PY; Bornert, C; Funck-Brentano, C; Poirier, JM; Verstuyft, C, 2005
)
0.58
" Based on multivariate analysis, the CYP2D6 metabolic ratio was a stronger independent predictor of adverse events (p<0."( Measurement of CYP2D6 and CYP3A4 activity in vivo with dextromethorphan: sources of variability and predictors of adverse effects in 419 healthy subjects.
Becquemont, L; Boëlle, PY; Bornert, C; Funck-Brentano, C; Poirier, JM; Verstuyft, C, 2005
)
0.58
" The CYP2D6 metabolic ratio predicts adverse events to DEM as does CYP2D6 phenotype, and extensive metabolizer subjects are not protected against adverse events."( Measurement of CYP2D6 and CYP3A4 activity in vivo with dextromethorphan: sources of variability and predictors of adverse effects in 419 healthy subjects.
Becquemont, L; Boëlle, PY; Bornert, C; Funck-Brentano, C; Poirier, JM; Verstuyft, C, 2005
)
0.58
" Adverse effects of these medications and the lack of evidence of their efficacy in children make their use a serious matter."( Toxicity and use of over-the-counter cough and cold medication in the pediatric population.
Montgomery, EJ; Wasserman, GS,
)
0.13
"3,4-Methylenedioxymethamphetamine (MDMA)'s O-demethylenated metabolite, 3,4-dihydroxymethamphetamine (HHMA), has been hypothesized to serve as a precursor for the formation of toxic catechol-thioether metabolites (e."( Inhibition of 3,4-methylenedioxymethamphetamine metabolism leads to marked decrease in 3,4-dihydroxymethamphetamine formation but no change in serotonin neurotoxicity: implications for mechanisms of neurotoxicity.
Maldonado Adrian, C; McCann, UD; Mueller, M; Ricaurte, GA; Yuan, J, 2011
)
0.37
" Adverse events were mostly mild or moderate and of expected types."( Efficacy and safety of dextromethorphan/quinidine at two dosage levels for diabetic neuropathic pain: a double-blind, placebo-controlled, multicenter study.
Hepner, A; Pope, LE; Shaibani, AI; Thisted, R, 2012
)
0.69
" Safety measures included adverse events (AEs), laboratory tests, electrocardiograms (ECGs), vital signs, and physical examinations."( An open-label multicenter study to assess the safety of dextromethorphan/quinidine in patients with pseudobulbar affect associated with a range of underlying neurological conditions.
Appel, SH; Formella, AE; Lomen-Hoerth, C; Pattee, GL; Pope, LE; Wymer, JP, 2014
)
0.65
" Adverse events included headache (7."( An open-label study to assess safety, tolerability, and effectiveness of dextromethorphan/quinidine for pseudobulbar affect in dementia: PRISM II results.
Cutler, AJ; D'Amico, S; Davis, CS; Doody, RS; Ledon, F; Shin, P; Siffert, J; Yonan, C, 2016
)
0.67
"DM/Q significantly reduced PBA symptoms in patients with dementia; reported adverse events were consistent with the known safety profile of DM/Q."( An open-label study to assess safety, tolerability, and effectiveness of dextromethorphan/quinidine for pseudobulbar affect in dementia: PRISM II results.
Cutler, AJ; D'Amico, S; Davis, CS; Doody, RS; Ledon, F; Shin, P; Siffert, J; Yonan, C, 2016
)
0.67
" We sought to characterize adverse events associated with dextromethorphan in children <12 years old from a surveillance program of OTC cough/cold medication exposures."( Adverse events associated with pediatric exposures to dextromethorphan.
Banner, W; Bond, GR; Burnham, RI; Green, JL; Kauffman, RE; Palmer, RB; Paul, IM; Reynolds, KM, 2017
)
0.95
"1716 cases contained ≥1 adverse event deemed at least potentially related to dextromethorphan; 1417 were single product exposures."( Adverse events associated with pediatric exposures to dextromethorphan.
Banner, W; Bond, GR; Burnham, RI; Green, JL; Kauffman, RE; Palmer, RB; Paul, IM; Reynolds, KM, 2017
)
0.93
" Adverse events were predominantly associated with overdose, most commonly affecting the central nervous and autonomic systems."( Adverse events associated with pediatric exposures to dextromethorphan.
Banner, W; Bond, GR; Burnham, RI; Green, JL; Kauffman, RE; Palmer, RB; Paul, IM; Reynolds, KM, 2017
)
0.7
" Adverse events (AEs) were consistent with the known DM/Q safety profile; the most common AE was diarrhea (8."( Safety, Tolerability, and Effectiveness of Dextromethorphan/Quinidine for Pseudobulbar Affect Among Study Participants With Traumatic Brain Injury: Results From the PRISM-II Open Label Study.
Davis, C; Formella, AE; Hammond, FM; Ledon, F; Sauve, W, 2018
)
0.74
"Initial research following regulatory changes addressing the pediatric safety of cough and cold medications (CCMs) demonstrated decreases in adverse events (AEs)."( Trends in adverse events and related health-care facility utilization from cough and cold medication exposures in children.
Banner, W; Bond, GR; Dart, RC; Green, JL; Kauffman, RE; Palmer, RB; Paul, IM; Rapp-Olsson, M; Reynolds, KM; Wang, GS, 2021
)
0.62
" No serious adverse events/deaths were reported."( Napabucasin Drug-Drug Interaction Potential, Safety, Tolerability, and Pharmacokinetics Following Oral Dosing in Healthy Adult Volunteers.
Brantley, SJ; Dai, X; Goulet, MT; Hard, ML; Hitron, M; Karol, MD; McLaughlin, CF, 2021
)
0.62
" The treatment was well-tolerated, with some transient adverse events reported."( Dextromethorphan-Bupropion for the Treatment of Depression: A Systematic Review of Efficacy and Safety in Clinical Trials.
Akbar, D; Cao, B; Ceban, F; Ho, R; Kwan, ATH; McIntyre, RS; Rhee, TG; Rosenblat, JD; Subramaniapillai, M; Teopiz, KM, 2023
)
2.35

Pharmacokinetics

The pharmacokinetic interactions of sertraline and fluoxetine with the tricyclic antidepressant desipramine were studied in 18 healthy male volunteers phenotyped as extensive metabolizers of dextromethorphan.

ExcerptReferenceRelevance
" To examine whether this effect is related to the chemical structure or pharmacokinetic characteristics of the inhibiting agent, we determined the effect of dextromethorphan (a compound which exhibits pharmacokinetic similarities to, but is chemically dissimilar from, previously studied agents) on the disposition of antipyrine."( Dextromethorphan pretreatment induces antipyrine clearance in the rat.
Svensson, CK; Ware, JA, 1988
)
1.91
" No therapeutically significant differences were detected between the three preparations tested, and there were no great differences between the pharmacokinetic profiles of dextromethorphan and dextrorphan."( Pharmacokinetics of dextromethorphan and dextrorphan: a single dose comparison of three preparations in human volunteers.
Hänninen, U; Karttunen, P; Kokkonen, P; Nykänen, S; Silvasti, M; Tukiainen, H, 1987
)
0.79
" Detailed pharmacokinetic and neuropsychology studies were performed."( High-dose dextromethorphan in amyotrophic lateral sclerosis: phase I safety and pharmacokinetic studies.
Evans, WE; Hollander, D; Kaplan, R; McLeod, HL; Munsat, TL; Pradas, J, 1994
)
0.69
"The pharmacokinetic interactions of sertraline and fluoxetine with the tricyclic antidepressant desipramine were studied in 18 healthy male volunteers phenotyped as extensive metabolizers of dextromethorphan."( Pharmacokinetics of desipramine coadministered with sertraline or fluoxetine.
Alderman, J; Chung, M; Harris, S; Harrison, W; Messig, M; Preskorn, SH, 1994
)
0.48
"The present report describes the pharmacokinetic characteristics of dextromethorphan (DM) and its main active metabolite dextrorphan (DX) in a group of epileptic patients receiving comedication."( Pharmacokinetics of dextromethorphan and dextrorphan in epileptic patients.
Kazis, A; Kimiskidis, V; Niopas, I,
)
0.69
" No evidence was seen of a pharmacokinetic interaction between CIT and Li, and this combination was well tolerated."( A double-blind, placebo-controlled study of citalopram with and without lithium in the treatment of therapy-resistant depressive patients: a clinical, pharmacokinetic, and pharmacogenetic investigation.
Amey, M; Baettig, D; Baumann, P; Jonzier-Perey, M; Kasas, A; Lambert, S; Montaldi, S; Nil, R; Souche, A; Uehlinger, C, 1996
)
0.29
" For the 200 mg dose group, the single dose plasma results showed no significant differences in pharmacokinetic parameters for NEF and HO-NEF in EM compared with PM subjects."( Single and multiple dose pharmacokinetics of nefazodone in subjects classified as extensive and poor metabolizers of dextromethorphan.
Barbhaiya, RH; Buch, AB; Greene, DS, 1996
)
0.5
"5 mg/kg in a crossover fashion in a two-sequence pharmacokinetic study with a wash-out period of at least 7 days."( A urine metabolic ratio of dextromethorphan and 3-methoxymorphinan as a probe for CYP3A activity and prediction of cyclosporine clearance in healthy volunteers.
Fleckenstein, LL; Ku, YM; Min, DI; Vichiendilokkul, A, 1999
)
0.6
" The enhancement of MS analgesia by DM is due to a pharmacodynamic interaction, not an effect on MS blood levels or a pharmacokinetic interaction between MS and DM, or their metabolites."( MorphiDex pharmacokinetic studies and single-dose analgesic efficacy studies in patients with postoperative pain.
Caruso, FS, 2000
)
0.31
"The terminal half-life of sabeluzole was significantly prolonged in subjects with severe hepatic dysfunction vs healthy subjects (respectively 39."( Pharmacokinetics of sabeluzole and dextromethorphan oxidation capacity in patients with severe hepatic dysfunction and healthy volunteers.
Blin, O; Bouhours, P; Lacarelle, B; Le Moing, JP; Levron, JC; Micallef, J; Nataf, MB; Pageaux, GP, 2001
)
0.59
" The authors present data evaluating the pharmacokinetic and pharmacodynamic responses to coadministration of oral linezolid with sympathomimetics (pseudoephedrine and phenylpropanolamine) and a serotonin reuptake inhibitor (dextromethorphan)."( Linezolid: pharmacokinetic and pharmacodynamic evaluation of coadministration with pseudoephedrine HCl, phenylpropanolamine HCl, and dextromethorpan HBr.
Antal, EJ; Batts, DH; Hendershot, PE; Hopkins, NK; Welshman, IR, 2001
)
0.5
" An analysis of pharmacokinetic parameters for determining the DM metabolic phenotype was conducted."( Analysis of pharmacokinetic parameters for assessment of dextromethorphan metabolic phenotypes.
Chen, JY; Ho, HO; Lee, YJ; Sheu, MT; Tao, PL; Yeh, GC,
)
0.38
" A multiple-dose study in healthy subjects with an extensive or a poor enzyme metabolizer phenotype evaluated the safety and pharmacokinetic profile of a selected fixed-dose combination (AVP-923)."( Pharmacokinetics of dextromethorphan after single or multiple dosing in combination with quinidine in extensive and poor metabolizers.
Berg, JE; Khalil, MH; Pope, LE; Sellers, EM; Stiles, M; Yakatan, GJ, 2004
)
0.65
" The pharmacokinetic parameters of dextrorphan were calculated by non-compartment model statistics."( [Determination of dextrorphan in human plasma and pharmacokinetic study].
Chen, XY; Gu, Q; Liu, D; Zhang, Y; Zhang, YF; Zhong, DF, 2004
)
0.32
" Pharmacokinetic parameters of dextrorphan was obtained as follows: Tmax was (2."( [Determination of dextrorphan in human plasma and pharmacokinetic study].
Chen, XY; Gu, Q; Liu, D; Zhang, Y; Zhang, YF; Zhong, DF, 2004
)
0.32
") on hyperalgesia and pain after a tissue injury in human volunteers, and to describe the relationship between pharmacokinetic and pharmacodynamic data."( Intravenous dextromethorphan to human volunteers: relationship between pharmacokinetics and anti-hyperalgesic effect.
Dahl, JB; Dirks, J; Duedahl, TH; Larsen, NE; Petersen, KB; Romsing, J, 2005
)
0.71
" Whereas 6 subjects of each phenotype were adequate to achieve 80% power in showing pharmacokinetic differences, the power required to detect a difference in antitussive response was less than 80% with 500 subjects in each study arm."( Incorporating in vitro information on drug metabolism into clinical trial simulations to assess the effect of CYP2D6 polymorphism on pharmacokinetics and pharmacodynamics: dextromethorphan as a model application.
Dickinson, GL; Lennard, MS; Proctor, NJ; Rezaee, S; Rostami-Hodjegan, A; Tucker, GT, 2007
)
0.53
" The enzymatic activity is reflected by various pharmacokinetic metrics such as the partial clearance of a parent compound to the respective CYP2D6-mediated metabolite or metabolic ratios."( Evaluation of probe drugs and pharmacokinetic metrics for CYP2D6 phenotyping.
Frank, D; Fuhr, U; Jaehde, U, 2007
)
0.34
" Pharmacokinetic parameters of midazolam and dextromethorphan were assessed alone and in the presence of AMD070."( Pharmacokinetic effect of AMD070, an Oral CXCR4 antagonist, on CYP3A4 and CYP2D6 substrates midazolam and dextromethorphan in healthy volunteers.
Becker, S; Calandra, GB; Chee, P; Everts, S; Hendrix, CW; MacFarland, RT; Nyunt, MM; Scarborough, R, 2008
)
0.82
" Blood samples were collected periodically in both phases and analyzed for parent drugs and metabolites (dextrorphan, 4'-hydroxy-flurbiprofen and 1'-hydroxy-midazolam) to calculate pharmacokinetic parameters."( Drug metabolism in hemorrhagic shock: pharmacokinetics of selective markers of cytochrome-P450 2C9, 2D6, and 3A4 enzyme activities in a porcine model.
Beilman, GJ; Kumar, A; Mann, HJ; Remmel, RP, 2011
)
0.37
" The aim of this study was to quantify the effects of the CYP2D6*1, *2, and *41 variants on DM metabolism in vivo and to identify other sources of pharmacokinetic variability."( Assessment of activity levels for CYP2D6*1, CYP2D6*2, and CYP2D6*41 genes by population pharmacokinetics of dextromethorphan.
Abduljalil, K; Frank, D; Fuhr, U; Gaedigk, A; Jaehde, U; Jetter, A; Kirchheiner, J; Klaassen, T; Tomalik-Scharte, D, 2010
)
0.57
"The pharmacokinetic parameters and the relative bioavailability of dextromethorphan chewing gum preparation in rabbits were compared with those of the commercially available chewing dextromethorphan tablets using 3P97 software."( [Pharmacokinetic study of a new chewing gum dextromethorphan delivery system].
Liu, BL; Liu, J; Tan, QY; Xu, ML; Zhang, JQ; Zhao, CJ, 2011
)
0.87
" Pharmacokinetic analyses were performed on day 1 without sorafenib and day 28 after steady-state sorafenib exposure; sorafenib pharmacokinetics were evaluated on day 28."( Interaction of sorafenib and cytochrome P450 isoenzymes in patients with advanced melanoma: a phase I/II pharmacokinetic interaction study.
Flaherty, KT; Frye, RF; Lathia, C; O'Dwyer, PJ; Redlinger, M; Rosen, M; Schuchter, L, 2011
)
0.37
"In the first DDI study, coadministration of ketoconazole (a CYP3A4 inhibitor) and clobazam increased clobazam's area under the concentration time curve from time zero extrapolated to infinity (AUC(0-∞) ) 54% and decreased clobazam's maximum plasma concentration (C(max) ) by 15% versus administration of clobazam alone, but the combination affected these pharmacokinetic parameters for N-CLB to a lesser degree."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
" The method has high sensitivity, specificity and allows high throughput analysis required for a pharmacokinetic study."( Simultaneous determination of dextromethorphan, dextrorphan and doxylamine in human plasma by HPLC coupled to electrospray ionization tandem mass spectrometry: application to a pharmacokinetic study.
De Nucci, G; Donato, JL; Koizumi, F; Mendes, GD; Pereira, AS, 2012
)
0.67
" Pharmacokinetic blood sampling was performed to assess the primary endpoints of the 90% confidence intervals (CIs) for the geometric mean ratios of the areas under the plasma concentration-time curves (AUCs) for memantine, dextromethorphan, dextrorphan - the dextromethorphan metabolite - and quinidine during concomitant therapy versus monotherapy."( A study of potential pharmacokinetic and pharmacodynamic interactions between dextromethorphan/quinidine and memantine in healthy volunteers.
Bartlett, C; Pope, LE; Schoedel, KA; Sellers, EM, 2012
)
0.79
"25) for memantine, dextromethorphan and dextrorphan, indicating no pharmacokinetic DDI."( A study of potential pharmacokinetic and pharmacodynamic interactions between dextromethorphan/quinidine and memantine in healthy volunteers.
Bartlett, C; Pope, LE; Schoedel, KA; Sellers, EM, 2012
)
0.94
"Minimal pharmacokinetic and pharmacodynamic interactions were observed between memantine and DMQ, suggesting they can be coadministered without dose adjustment."( A study of potential pharmacokinetic and pharmacodynamic interactions between dextromethorphan/quinidine and memantine in healthy volunteers.
Bartlett, C; Pope, LE; Schoedel, KA; Sellers, EM, 2012
)
0.61
" The developed method was successfully applied to analyze the drug in samples of rat plasma for pharmacokinetic study."( Simultaneous determination of dextromethorphan and dextrophan in rat plasma by LC-MS/MS and its application to a pharmacokinetic study.
Chen, X; Hu, L; Wang, X; Wang, Z; Xu, R; Xu, T; Zhan, H; Zhang, X, 2012
)
0.67
" In contrast, there were no significant differences in any of the pharmacokinetic parameters for warfarin between the two genotypes."( Pharmacogenomic/pharmacokinetic assessment of a four-probe cocktail for CYPs and OATPs following oral microdosing.
Ando, Y; Deguchi, M; Fukae, M; Hirota, T; Ieiri, I; Irie, S; Iwasaki, K; Kanda, E; Kimura, M; Maeda, K; Matsuguma, K; Matsuki, S; Nakamura, T; Sugiyama, Y, 2012
)
0.38
"The present study suggests that 1) the sampling strategy should be optimized according to pharmacokinetic profiles of the test drugs following oral microdosing, and 2) microdosing can be applied to the pharmacogenomic study of CYP-specific drugs."( Pharmacogenomic/pharmacokinetic assessment of a four-probe cocktail for CYPs and OATPs following oral microdosing.
Ando, Y; Deguchi, M; Fukae, M; Hirota, T; Ieiri, I; Irie, S; Iwasaki, K; Kanda, E; Kimura, M; Maeda, K; Matsuguma, K; Matsuki, S; Nakamura, T; Sugiyama, Y, 2012
)
0.38
"We conducted a pharmacokinetic (PK) study and a pharmacodynamic (PD) study to assess whether Roux-en-Y gastric bypass (RYGB) surgery is associated with significant changes to PK and PD of oral medications."( Pharmacokinetic and pharmacodynamic alterations in the Roux-en-Y gastric bypass recipients.
Chalasani, N; Hall, SD; Jones, DR; Mattar, S; Tandra, S; Vuppalanchi, R, 2013
)
0.39
" Compared with controls, the RYGB group had brisk natriuresis, with significantly lower tmax for urine sodium (1."( Pharmacokinetic and pharmacodynamic alterations in the Roux-en-Y gastric bypass recipients.
Chalasani, N; Hall, SD; Jones, DR; Mattar, S; Tandra, S; Vuppalanchi, R, 2013
)
0.39
"RYGB recipients have significantly shorter tmax for the studied orally administered medications, but otherwise no other significant changes in PK were reported."( Pharmacokinetic and pharmacodynamic alterations in the Roux-en-Y gastric bypass recipients.
Chalasani, N; Hall, SD; Jones, DR; Mattar, S; Tandra, S; Vuppalanchi, R, 2013
)
0.39
"Conducting pharmacokinetic (PK) studies in pregnant women is challenging."( A physiologically based pharmacokinetic model to predict disposition of CYP2D6 and CYP1A2 metabolized drugs in pregnant women.
Isoherranen, N; Ke, AB; Nallani, SC; Rostami-Hodjegan, A; Unadkat, JD; Zhao, P, 2013
)
0.39
" To determine the clinical effect of GSE on CYP2D6, the pharmacokinetic interaction between GSE and the sensitive CYP2D6 probe dextromethorphan in healthy adult volunteers was examined."( The effect of grape seed extract on the pharmacokinetics of dextromethorphan in healthy volunteers.
Beijnen, JH; Goey, AK; Meijerman, I; Schellens, JH, 2013
)
0.84
"Population pharmacokinetic models were developed for dextromethorphan, tamoxifen and their metabolites."( Population pharmacokinetic modelling to assess the impact of CYP2D6 and CYP3A metabolic phenotypes on the pharmacokinetics of tamoxifen and endoxifen.
Beijnen, JH; Binkhorst, L; de Bruijn, P; de Graan, AJ; Huitema, AD; Mathijssen, RH; ter Heine, R, 2014
)
0.65
" This study determined, for the first time, the basic pharmacokinetic profile of DM and its active metabolite, dextrorphan (DP) in children and adolescents."( Pharmacokinetic profile of dextromethorphan hydrobromide in a syrup formulation in children and adolescents.
Armogida, M; Guenin, E; Riff, D, 2014
)
0.7
" The terminal half-life (t ½) values were longer in the adolescent group due in part to the single PM subject."( Pharmacokinetic profile of dextromethorphan hydrobromide in a syrup formulation in children and adolescents.
Armogida, M; Guenin, E; Riff, D, 2014
)
0.7
"This is the first evaluation of the pharmacokinetic characteristics of DM in children and adolescents."( Pharmacokinetic profile of dextromethorphan hydrobromide in a syrup formulation in children and adolescents.
Armogida, M; Guenin, E; Riff, D, 2014
)
0.7
"To evaluate felodipine as a potential perpetrator of pharmacokinetic drug-drug interactions (PK-DDIs) involving cytochrome P450 (CYP) enzymes and P-glycoprotein (P-gp)."( Evaluation of felodipine as a potential perpetrator of pharmacokinetic drug-drug interactions.
Doogue, MP; Miners, JO; Polasek, TM; Rowland, A; Snyder, BD, 2014
)
0.4
" Blood samples for pharmacokinetic analysis were collected pre-dose and serially for 72 h post-dose."( Lisdexamfetamine Dimesylate Effects on the Pharmacokinetics of Cytochrome P450 Substrates in Healthy Adults in an Open-Label, Randomized, Crossover Study.
Corcoran, M; Ermer, J; Martin, P, 2015
)
0.42
" Plasma concentrations of slowly eliminated caffeine and R-/S-warfarin and rapidly eliminated omeprazole and midazolam previously observed in cynomolgus monkeys were scaled to human oral biomonitoring equivalents using known species allometric scaling factors and in vitro metabolic clearance data with a simple physiologically based pharmacokinetic (PBPK) model."( Human plasma concentrations of cytochrome P450 probes extrapolated from pharmacokinetics in cynomolgus monkeys using physiologically based pharmacokinetic modeling.
Murayama, N; Shida, S; Shimizu, M; Uno, Y; Utoh, M; Yamazaki, H, 2015
)
0.42
" In this study, we investigated the pharmacokinetic interaction between Zuojin Pill and the sensitive CYP2D6 probe dextromethorphan in healthy Chinese volunteers with CYP2D6*10 genotype."( Effects of the Chinese herbal formula "Zuojin Pill" on the pharmacokinetics of dextromethorphan in healthy Chinese volunteers with CYP2D6*10 genotype.
Jiang, J; Liu, S; Miao, P; Qiu, F; Ye, Y; Zeng, J; Zhao, T; Zhu, L, 2016
)
0.87
" In contrast, no significant change was observed in these pharmacokinetic parameters of the participants with CYP2D6*10/*10 genotype."( Effects of the Chinese herbal formula "Zuojin Pill" on the pharmacokinetics of dextromethorphan in healthy Chinese volunteers with CYP2D6*10 genotype.
Jiang, J; Liu, S; Miao, P; Qiu, F; Ye, Y; Zeng, J; Zhao, T; Zhu, L, 2016
)
0.66
" A physiologically based pharmacokinetic (PBPK) model can be developed to do simulations based on the large virtual Chinese population and evaluate single-point plasma phenotyping method of CYP2D6."( Application of a physiologically based pharmacokinetic model for the evaluation of single-point plasma phenotyping method of CYP2D6.
Berk, D; Chen, R; Hu, P; Rostami-Hodjegan, A; Shi, J; Wang, H, 2016
)
0.43
"A physiologically based pharmacokinetic (PBPK) model was used to simulate the impact of elevated levels of interleukin (IL)-6 on the exposure of several orally administered cytochrome P450 (CYP) probe substrates (caffeine, S-warfarin, omeprazole, dextromethorphan, midazolam, and simvastatin)."( Simulating the Impact of Elevated Levels of Interleukin-6 on the Pharmacokinetics of Various CYP450 Substrates in Patients with Neuromyelitis Optica or Neuromyelitis Optica Spectrum Disorders in Different Ethnic Populations.
Ducray, PS; Endo-Tsukude, C; Gardner, I; Gill, KL; Hatley, OJ; Machavaram, KK; Parrott, N; Terao, K, 2019
)
0.7
"The aim of this work was to predict the extent of Cytochrome P450 2D6 (CYP2D6)-mediated drug-drug interactions (DDIs) in different CYP2D6 genotypes using physiologically-based pharmacokinetic (PBPK) modeling."( Physiologically-Based Pharmacokinetic Modeling for the Prediction of CYP2D6-Mediated Gene-Drug-Drug Interactions.
Daali, Y; Desmeules, J; Storelli, F, 2019
)
0.51
"This study provides a whole-body physiologically-based pharmacokinetic (PBPK) model of dextromethorphan and its metabolites dextrorphan and dextrorphan O-glucuronide for predicting the effects of cytochrome P450 2D6 (CYP2D6) drug-gene interactions (DGIs) on dextromethorphan pharmacokinetics (PK)."( Physiologically-based pharmacokinetic modeling of dextromethorphan to investigate interindividual variability within CYP2D6 activity score groups.
Fuhr, U; Lehr, T; Rüdesheim, S; Schwab, M; Selzer, D, 2022
)
1.2
" This study aimed to build a physiologically based pharmacokinetic (PBPK) model reflecting observed changes in physiological and molecular parameters relevant to drug disposition that are associated with MAFLD."( A Physiologically Based Pharmacokinetic Model to Predict the Impact of Metabolic Changes Associated with Metabolic Associated Fatty Liver Disease on Drug Exposure.
Newman, EM; Rowland, A, 2022
)
0.72
" The application of the method was demonstrated through the estimation of pharmacokinetic parameters that showed good congruence with earlier studies."( Low-volume plasma sampling for determination of dextromethorphan and dextrorphan in rat plasma: LC-MS/MS method and its application in pharmacokinetic study.
Ghosh, G; Kamte, S; Majhi, PK; Mishra, P; Sahu, R; Sayyad, S; Subudhi, BB, 2023
)
1.17

Compound-Compound Interactions

No analgesic effects of oral dextromethorphan 120 mg on pain after surgical termination of labour, and no additive analgesic effect when combined with ibuprofen 400 mg, were observed. The magnitude of drug-drug interaction between dextrome and paroxetine was higher in homozygous than in heterozygous subjects.

ExcerptReferenceRelevance
" In vivo, inhibitory potency is affected by the plasma concentration of the free (unbound) drug, a potentially important consideration since many CYP2D6-metabolised drugs exhibit nonlinear (saturable) kinetics, and by the presence of metabolites, which might accumulate and interact with the CYP system."( Antidepressant drug interactions and the cytochrome P450 system. The role of cytochrome P450 2D6.
Ereshefsky, L; Lam, YW; Riesenman, C, 1995
)
0.29
"No analgesic effects of oral dextromethorphan 120 mg on pain after surgical termination of labour, and no additive analgesic effects when combined with ibuprofen 400 mg, were observed."( The effect of dextromethorphan, alone or in combination with ibuprofen, on postoperative pain after minor gynaecological surgery.
Bach, LF; Dahl, JB; Ilkjaer, S; Nielsen, PA; Wernberg, M, 2000
)
0.96
" To find out whether quinidine (Q), a CYP2D6 inhibitor, could elevate and prolong DM plasma profiles, 2 multiple-dose studies identified the lowest oral dose of Q that could be used in a fixed combination with 3 doses of DM."( Pharmacokinetics of dextromethorphan after single or multiple dosing in combination with quinidine in extensive and poor metabolizers.
Berg, JE; Khalil, MH; Pope, LE; Sellers, EM; Stiles, M; Yakatan, GJ, 2004
)
0.65
" The goal of this study was to investigate the effect of preincisional IM DM combined with intraoperative TEA and postoperative patient-controlled epidural analgesia (PCEA) on pain and bowel function after colonic surgery."( Preincisional dextromethorphan combined with thoracic epidural anesthesia and analgesia improves postoperative pain and bowel function in patients undergoing colonic surgery.
Huh, BK; Jao, SW; White, WD; Wong, CS; Wu, CT; Yang, CP; Yeh, CC, 2005
)
0.69
"Inhibition of cytochrome P450 (CYP) is a principal mechanism for metabolism-based drug-drug interactions (DDIs)."( Development of an in vitro drug-drug interaction assay to simultaneously monitor five cytochrome P450 isoforms and performance assessment using drug library compounds.
Burdette, D; Dunklee, MB; Fahmi, O; Heinle, L; Hyland, R; Lee, C; Miller, H; Smith, D; Thurston, A; Zientek, M,
)
0.13
" The method is simple, rapid and rugged, and has been applied successfully to sample analysis in support of a drug-drug interaction study."( Simultaneous determination of tolbutamide, omeprazole, midazolam and dextromethorphan in human plasma by LC-MS/MS--a high throughput approach to evaluate drug-drug interactions.
Bertelsen, K; Guo, P; Han, F; Huang, MQ; Lin, ZJ; Weng, N; Zhang, W; Zhao, H, 2010
)
0.6
"25] for all analytes, indicating a drug-drug interaction."( Randomized open-label drug-drug interaction trial of dextromethorphan/quinidine and paroxetine in healthy volunteers.
Pope, LE; Schoedel, KA; Sellers, EM, 2012
)
0.63
"In summary, results from this study suggest that maintenance on dextromethorphan in combination with quinidine has a limited role in the treatment of opioid dependence."( Evaluation of the reinforcing and subjective effects of heroin in combination with dextromethorphan and quinidine.
Comer, SD; Sullivan, MA; Vosburg, SK,
)
0.6
"To investigate potential drug-drug interactions between clobazam and cytochrome P450 (CYP) isoenzyme substrates, inhibitors, and inducers."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
"Two, prospective, open-label, single-center, drug-drug interaction (DDI) studies and a population pharmacokinetics analysis of seven multicenter phase II-III trials."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
"Fifty-four healthy adult volunteers were enrolled in the two drug-drug interaction studies; 53 completed the studies."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
"In the first drug-drug interaction study, 36 participants received a single oral dose of clobazam 10 mg on day 1, followed by either ketoconazole 400 mg once/day or omeprazole 40 mg once/day on days 17-22, with a single dose of clobazam 10 mg coadministered on day 22, to study the effects of CYP3A4 or CYP2C19 inhibition, respectively, on clobazam and its active metabolite N-desmethylclobazam (N-CLB)."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
" In the second DDI study, no clinically significant drug-drug interactions were observed between clobazam 40 mg and the CYP probe substrates caffeine or tolbutamide."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
"These findings suggest no clinically meaningful drug-drug interactions between clobazam and drugs metabolized by CYP3A4, CYP2C19, CYP1A2, or CYP2C9."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
"The hepatic organic anion transporting polypeptides (OATPs) influence the pharmacokinetics of several drug classes and are involved in many clinical drug-drug interactions."( Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
Artursson, P; Haglund, U; Karlgren, M; Kimoto, E; Lai, Y; Norinder, U; Vildhede, A; Wisniewski, JR, 2012
)
0.38
" Because of instability in plasma, further characterization of the DM1 and DM4 intramolecular and intermolecular disulfide conjugates observed in vivo is required before an accurate drug-drug interaction (DDI) prediction can be made."( In vitro characterization of the drug-drug interaction potential of catabolites of antibody-maytansinoid conjugates.
Davis, JA; Pearson, JT; Rock, DA; Wienkers, LC, 2012
)
0.38
" This observed discrepancy between in vitro risk assessment and in vivo drug-drug interaction (DDI) profile was rationalized by time-varying dynamic pharmacokinetic models that incorporated circulating concentrations of fluoxetine and norfluoxetine enantiomers, mutual inhibitor-inhibitor interactions, and CYP3A4 induction."( Fluoxetine- and norfluoxetine-mediated complex drug-drug interactions: in vitro to in vivo correlation of effects on CYP2D6, CYP2C19, and CYP3A4.
Davis, C; Foti, RS; Isoherranen, N; Kunze, KL; Lutz, JD; Sager, JE, 2014
)
0.4
"The objective is to explore the clinical curative effects of methylprednisolone combined with MTX and DXM intrathecal injection in treating neuropsychiatric systemic lupus erythematosus (NPSLE) and its effects on autoantibody level and anti-N-methyl-D-aspartate receptor subtype NR2a/2b antibody (anti-NR2 antibody) level."( Impact analysis of autoantibody level and NR2 antibody level in neuropsychiatric SLE treated by methylprednisolone combined with MTX and DXM intrathecal injection.
Fu, B; Lei, H; Wang, J; Zhang, J; Zhao, Y; Zhu, G, 2014
)
0.4
"To evaluate felodipine as a potential perpetrator of pharmacokinetic drug-drug interactions (PK-DDIs) involving cytochrome P450 (CYP) enzymes and P-glycoprotein (P-gp)."( Evaluation of felodipine as a potential perpetrator of pharmacokinetic drug-drug interactions.
Doogue, MP; Miners, JO; Polasek, TM; Rowland, A; Snyder, BD, 2014
)
0.4
" To further validate this cocktail, in this study, we have verified whether probe drugs contained in the latter cause mutual drug-drug interactions."( Evaluation of Mutual Drug-Drug Interaction within Geneva Cocktail for Cytochrome P450 Phenotyping using Innovative Dried Blood Sampling Method.
Bosilkovska, M; Daali, Y; Déglon, J; Desmeules, J; Samer, C; Thomas, A; Walder, B, 2016
)
0.43
" The study aimed to evaluate the feasibility of different doses of DXM combined with standard AM treatment in clinical hypertension."( Combination With Low-dose Dextromethorphan Improves the Effect of Amlodipine Monotherapy in Clinical Hypertension: A First-in-human, Concept-proven, Prospective, Dose-escalation, Multicenter Study.
Chen, JW; Chen, P; Cheng, SM; Hung, YJ; Tseng, WK; Wang, KY; Wen, MS; Wu, CC; Wu, TC; Yeh, HI; Yin, WH, 2016
)
0.73
" In a clinical trial, a strong drug-drug interaction (DDI) was observed between dextromethorphan (DM, the object or victim drug) and GSK1034702 (the precipitant or perpetrator drug), following single and repeat doses."( Retrospective use of PBPK modelling to understand a clinical drug-drug interaction between dextromethorphan and GSK1034702.
Bloomer, J; Dear, G; Hobbs, MJ, 2017
)
0.9
" The magnitude of drug-drug interaction between dextromethorphan and paroxetine was higher in homozygous than in heterozygous subjects (14."( Impact of CYP2D6 Functional Allelic Variations on Phenoconversion and Drug-Drug Interactions.
Daali, Y; Desmeules, J; Lenglet, S; Matthey, A; Storelli, F; Thomas, A, 2018
)
0.74
"To evaluate the capacity for modafinil to be a perpetrator of metabolic drug-drug interactions by altering cytochrome P450 activity following a single dose and dosing to steady state."( Evaluation of modafinil as a perpetrator of metabolic drug-drug interactions using a model informed cocktail reaction phenotyping trial protocol.
Mangoni, AA; Rowland, A; Sorich, MJ; van Dyk, M; Warncken, D, 2018
)
0.48
"These data support consideration of the risk of clinically relevant metabolic drug-drug interactions perpetrated by modafinil when this drug is co-administered with drugs that are primarily cleared by CYP2C19 (single modafinil dose or steady state modafinil dosing) or CYP3A4 (steady state modafinil dosing only) catalysed metabolic pathways."( Evaluation of modafinil as a perpetrator of metabolic drug-drug interactions using a model informed cocktail reaction phenotyping trial protocol.
Mangoni, AA; Rowland, A; Sorich, MJ; van Dyk, M; Warncken, D, 2018
)
0.48
" Patients were enrolled into an inpatient study unit, and orally administered a 2 mg microdose of dextromethorphan (DM) to probe enzyme activity with and without CYP2D6-dependent drug-drug interactions."( CYP2D6 Genotype Phenotype Discordance Due to Drug-Drug Interaction.
Abbott, D; Abdelmawla, F; Anderson, PL; Bushman, LR; Flaten, HK; McDaniel, KT; Monte, AA; Saben, J; Shelton, S; West, K; Williamson, K, 2018
)
0.7
" The aim of this study was to determine whether all active retinoids downregulate CYP2D6 and whether in vitro downregulation translates to in vivo drug-drug interactions (DDIs)."( Does In Vitro Cytochrome P450 Downregulation Translate to In Vivo Drug-Drug Interactions? Preclinical and Clinical Studies With 13-cis-Retinoic Acid.
Amory, JK; Hogarth, C; Isoherranen, N; Kenny, JR; Kosaka, M; Stevison, F; Wong, S, 2019
)
0.51
" This study confirmed that the Inje cocktail approach was able to detect relevant drug-drug interactions impacting further development of ASP8477 and future therapeutic use."( A Cocktail Interaction Study Evaluating the Drug-Drug Interaction Potential of the Perpetrator Drug ASP8477 at Multiple Ascending Dose Levels.
Collins, C; Ernault, E; Fuhr, R; Gangaram-Panday, S; Passier, P; Treijtel, N; van Bruijnsvoort, M, 2019
)
0.51
"The aim of this work was to predict the extent of Cytochrome P450 2D6 (CYP2D6)-mediated drug-drug interactions (DDIs) in different CYP2D6 genotypes using physiologically-based pharmacokinetic (PBPK) modeling."( Physiologically-Based Pharmacokinetic Modeling for the Prediction of CYP2D6-Mediated Gene-Drug-Drug Interactions.
Daali, Y; Desmeules, J; Storelli, F, 2019
)
0.51

Bioavailability

The pharmacokinetic parameters and the relative bioavailability of dextromethorphan chewing gum preparation in rabbits were compared with those of the commercially available chewing dextrome tablets using 3P97 software.

ExcerptReferenceRelevance
" The bioavailability of dextromethorphan hydrobromide after 30 mg po, as measured by the concentration of total (free and conjugated) dextrorphan in the plasma, was determined in six human volunteers with this procedure."( Determination of dextrorphan in plasma and evaluation of bioavailability of dextromethorphan hydrobromide in humans.
Emele, JF; Ramachander, G; Williams, FD, 1977
)
0.79
"9% compared with the intravenous route, whereas the plasma bioavailability from the nasal route was 78."( Nasal delivery of [14C]dextromethorphan hydrochloride in rats: levels in plasma and brain.
Behl, CR; Char, H; Iqbal, K; Kumar, S; Malick, AW; Patel, S; Piemontese, D; Salvador, RA, 1992
)
0.59
"A large interindividual variability for clozapine bioavailability and plasma steady-state concentrations and clearance exists."( The antipsychotic clozapine is metabolized by the polymorphic human microsomal and recombinant cytochrome P450 2D6.
Fischer, V; Maurer, G; Tynes, RE; Vogels, B, 1992
)
0.28
"We compared in this double-blind crossover study the bioavailability of dextromethorphan from a dextromethorphan-salbutamol combination tablet (Redol comp) and from a plain dextromethorphan tablet (Extuson) by determining dextrorphan concentrations after single-dose oral administration in 10 healthy volunteers."( Pharmacokinetic comparison of a dextromethorphan-salbutamol combination tablet and a plain dextromethorphan tablet.
Happonen, P; Karttunen, P; Mykkänen, M; Romppanen, T; Silvasti, M; Tukiainen, H, 1990
)
0.79
"A randomized, two-way, steady-state crossover study was performed in 24 healthy male volunteers to evaluate the bioavailability of a controlled-release (CR) dextromethorphan (DM) suspension."( Bioavailability evaluation of a controlled-release dextromethorphan liquid.
Amsel, LP; Dennis, SR; Hinsvark, ON; Rotenberg, KS; Woodworth, JR, 1987
)
0.72
" Bioavailability of dextromethorphan tablets was comparable to syrup."( Dextromethorphan and codeine: comparison of plasma kinetics and antitussive effects.
Aylward, M; Davies, DE; Leideman, T; Maddock, J; Protheroe, DA, 1984
)
2.03
"A reverse-phase High Performance Liquid Chromatographic (HPLC) method was developed for the analysis of metoprolol in the large number of human plasma samples obtained in in vitro-in vivo correlations (IVIVC) and bioavailability studies of extended release formulations of metoprolol tartrate."( A sensitive assay of metoprolol and its major metabolite alpha-hydroxy metoprolol in human plasma and determination of dextromethorphan and its metabolite dextrorphan in urine with high performance liquid chromatography and fluorometric detection.
Eddington, NE; Leslie, J; Mistry, B, 1998
)
0.51
"A multiple dose bioavailability study with six healthy male human volunteers was conducted."( Bioavailability of dextromethorphan (as dextrorphan) from sustained release formulations in the presence of guaifenesin in human volunteers.
Demirbas, S; Reyderman, L; Stavchansky, S, 1998
)
0.63
" Bioavailability (F) of DM increased significantly with grapefruit and seville orange juice, but only returned to half the baseline value after three days of washout."( The effect of grapefruit juice and seville orange juice on the pharmacokinetics of dextromethorphan: the role of gut CYP3A and P-glycoprotein.
Di Marco, MP; Ducharme, MP; Edwards, DJ; Wainer, IW, 2002
)
0.54
" Oral bioavailability was 11% as calculated from naive pooled data."( Plasma profile and pharmacokinetics of dextromethorphan after intravenous and oral administration in healthy dogs.
Kukanich, B; Papich, MG, 2004
)
0.59
"The aim of this study was to compare the bioavailability of cyclosporine (CyA) from the generic dispersion formulation Cicloral (CIC) with the microemulsion formulation Neoral (NEO) and the original Sandimmune (SIM) capsules after single doses of 100, 300, or 600 mg of drug, respectively."( Comparative bioavailability of the microemulsion formulation of cyclosporine (Neoral) with a generic dispersion formulation (Cicloral) in young healthy male volunteers.
Bucher, M; Burhenne, J; Faerber, L; Gschaidmeier, H; Kees, F; Mikus, G; Schweda, F, 2006
)
0.33
"To assess the single-dose absolute oral bioavailability of traxoprodil in healthy male volunteers phenotyped as either CYP2D6 extensive or poor metabolisers."( Absolute oral bioavailability of traxoprodil in cytochrome P450 2D6 extensive and poor metabolisers.
Benincosa, LJ; Crownover, PH; Diringer, K; Gibbs, MA; Russell, T; Taylor, TJ; Venkatakrishnan, K; Wilner, K, 2006
)
0.33
"In poor metabolisers (n = 6), oral bioavailability was approximately 80% and was consistent with a liver extraction ratio of approximately 20% (plasma clearance of approximately 4 mL/min/kg) indicating near complete absorption."( Absolute oral bioavailability of traxoprodil in cytochrome P450 2D6 extensive and poor metabolisers.
Benincosa, LJ; Crownover, PH; Diringer, K; Gibbs, MA; Russell, T; Taylor, TJ; Venkatakrishnan, K; Wilner, K, 2006
)
0.33
" In extensive metabolisers, the oral bioavailability was nonlinear and dose-dependent, while in poor metabolisers, oral bioavailability appeared to be linear and dose-independent."( Absolute oral bioavailability of traxoprodil in cytochrome P450 2D6 extensive and poor metabolisers.
Benincosa, LJ; Crownover, PH; Diringer, K; Gibbs, MA; Russell, T; Taylor, TJ; Venkatakrishnan, K; Wilner, K, 2006
)
0.33
" DM's limited clinical benefit is proposed to be associated with its rapid metabolism to dextrorphan, which restricts its central bioavailability and therapeutic utility."( Dextromethorphan as a potential neuroprotective agent with unique mechanisms of action.
Calef, U; Lauterbach, EC; Werling, LL, 2007
)
1.78
"The data support an alteration in bioavailability due to an AMD070-mediated inhibition of presystemic metabolism, though an intestinal P-glycoprotein effect could also be contributing."( Pharmacokinetic effect of AMD070, an Oral CXCR4 antagonist, on CYP3A4 and CYP2D6 substrates midazolam and dextromethorphan in healthy volunteers.
Becker, S; Calandra, GB; Chee, P; Everts, S; Hendrix, CW; MacFarland, RT; Nyunt, MM; Scarborough, R, 2008
)
0.56
" A complete cross-over bioavailability study of the optimized formulation of the developed sustained tablets and marketed immediate release tablets was performed on six healthy male volunteers."( Formulation and evaluation of dextromethorphan hydrobromide sustained release tablets.
Krishnaraj, K; Meyyanathan, SN; Muralidaharan, S; Rajan, S; Siddaiah, MK; Suresh, B, 2008
)
0.63
" DM has been under investigation for several years as a neuroprotective agent in stroke, neurosurgery and amyotrophic lateral sclerosis (ALS); however, it is rapidly metabolized by CYP2D6, reducing the drug's bioavailability at neuronal targets."( AVP-923, a combination of dextromethorphan hydrobromide and quinidine sulfate for the treatment of pseudobulbar affect and neuropathic pain.
Olney, N; Rosen, H, 2010
)
0.66
"To establish an high-performance liquid chromatography (HPLC)-based method for analysis of the pharmacokinetics and relative bioavailability of dextromethorphan chewing gum tablets in rabbits."( [Pharmacokinetic study of a new chewing gum dextromethorphan delivery system].
Liu, BL; Liu, J; Tan, QY; Xu, ML; Zhang, JQ; Zhao, CJ, 2011
)
0.83
"The pharmacokinetic parameters and the relative bioavailability of dextromethorphan chewing gum preparation in rabbits were compared with those of the commercially available chewing dextromethorphan tablets using 3P97 software."( [Pharmacokinetic study of a new chewing gum dextromethorphan delivery system].
Liu, BL; Liu, J; Tan, QY; Xu, ML; Zhang, JQ; Zhao, CJ, 2011
)
0.87
" The relative bioavailability of the new chewing gum medicine system was (140."( [Pharmacokinetic study of a new chewing gum dextromethorphan delivery system].
Liu, BL; Liu, J; Tan, QY; Xu, ML; Zhang, JQ; Zhao, CJ, 2011
)
0.63
" However, maximum plasma concentration, half-life, area under the curve, and oral bioavailability were not different."( Pharmacokinetic and pharmacodynamic alterations in the Roux-en-Y gastric bypass recipients.
Chalasani, N; Hall, SD; Jones, DR; Mattar, S; Tandra, S; Vuppalanchi, R, 2013
)
0.39
" Quinidine, a cytochrome P450 (CYP) 2D6 inhibitor, was also evaluated in conjunction with dextromethorphan to increase the bioavailability of dextromethorphan and reduce exposure to additional metabolites."( Involvement of sigma-1 receptors in the antidepressant-like effects of dextromethorphan.
Healy, JR; Matsumoto, RR; Nguyen, L; Robson, MJ; Scandinaro, AL, 2014
)
0.86
" In the past few years, many studies showed that borneol can improve the bioavailability of other drugs, promoting drugs to cross the blood-brain barrier, so the potential drug interactions between borneol and other medicines have attracted great attention, but the influence of borneol to CYP450 and its isoforms are rarely reported."( [Borneol is an inducer of rat hepatic CYP2D activity in vivo].
Chen, JY; Chen, Y; Meng, MR; Wang, JJ, 2015
)
0.42
" Coadministration of DM with a low dose of quinidine inhibits DM metabolism, yields greater bioavailability and enables more specific testing of the therapeutic properties of DM apart from its metabolites."( Pharmacology of dextromethorphan: Relevance to dextromethorphan/quinidine (Nuedexta®) clinical use.
Matsumoto, RR; Pope, LE; Siffert, J; Taylor, CP; Traynelis, SF, 2016
)
0.78
" This study shows that dissolution and the rate of absorption into the enterocytes are clinically irrelevant for the performance of DEX immediate release product."( The Irrelevance of In Vitro Dissolution in Setting Product Specifications for Drugs Like Dextromethorphan That are Subject to Lysosomal Trapping.
Almukainzi, M; Bolger, MB; Löbenberg, R; Macwan, JS; Sarfraz, M, 2019
)
0.74
" However, the bioavailability of 3-HM is very low due to the rapid first-pass metabolism of the phenolic moiety."( Design, Synthesis, and Biological Evaluation of Phenol Bioisosteric Analogues of 3-Hydroxymorphinan.
Bai, X; Bao, X; Li, Z; Shang, J; Sheng, C; Wang, J; Wang, Y; Zhang, D; Zhang, G; Zhu, M, 2019
)
0.51
"Common cold symptoms may be mitigated by products in caplet, nasal spray, and oral solution formulations, although variations exist in the bioavailability of the active ingredients contained within these products."( A single-dose, open-label, randomized, scintigraphic study to investigate the gastrointestinal behavior of 2 triple-combination cold products (acetaminophen, phenylephrine, and dextromethorphan) in healthy male volunteers.
Armogida, M; Doll, WJ; Mallefet, P; Page, RC; Sandefer, EP, 2022
)
0.91

Dosage Studied

Dextromethorphan and metabolites in serum could be advantageous to measure individual CYP2D6 activities in vivo and thus optimize the dosing of drugs metabolized by CYP1A2. Animals receiving dextromethmorphan had significantly smaller lesions, which was independent of dosing schedule.

ExcerptRelevanceReference
" Therapy with benzoate, 500 mg/kg per day, was started on day 5, and the dosage was increased to 750 mg/kg per day on day 8, with prompt normalization of the neurologic and electroencephalographic findings."( Dextromethorphan and high-dose benzoate therapy for nonketotic hyperglycinemia in an infant.
Francomano, CA; Hamosh, A; Johnston, MV; McDonald, JW; Niedermeyer, E; Valle, D, 1992
)
1.73
", the morphine dose-response curve was shifted to the right."( Potentiation of disruptive effects of dextromethorphan by naloxone on fixed-interval performance in rats.
Taşkin, T, 1986
)
0.54
" Blood samples were collected over a 12-hour dosing period on the last day of each treatment and analyzed for DM and its active metabolite, dextrorphan (DP)."( Bioavailability evaluation of a controlled-release dextromethorphan liquid.
Amsel, LP; Dennis, SR; Hinsvark, ON; Rotenberg, KS; Woodworth, JR, 1987
)
0.52
"A high-performance liquid chromatographic (HPLC) method has been developed for the quantitation of acetaminophen, chlorpheniramine maleate, dextromethorphan hydrobromide, and phenylpropanolamine hydrochloride in combination in pharmaceutical dosage forms using a single column and three different mobile phases."( Quantitation of acetaminophen, chlorpheniramine maleate, dextromethorphan hydrobromide, and phenylpropanolamine hydrochloride in combination using high-performance liquid chromatography.
Das Gupta, V; Heble, AR, 1984
)
0.71
"Plasma kinetics of dextromethorphan (as dextrorphan ) and codeine were investigated after acute oral doses in 8 patients with pathological cough; after which the patients participated in an acute dose-response study of the antitussive effects of each drug administered as syrups."( Dextromethorphan and codeine: comparison of plasma kinetics and antitussive effects.
Aylward, M; Davies, DE; Leideman, T; Maddock, J; Protheroe, DA, 1984
)
2.04
"8 years), all urine EMIT assays six hours after ingestion of dextromethorphan, at both dosage levels, were negative for opioids and all other drugs."( The dextromethorphan defense: dextromethorphan and the opioid screen.
Magoon, MR; Norton, J; Storrow, AB, 1995
)
1.09
" Interpopulation differences in the prevalence of deficiencies of drug-metabolizing enzymes may be clinically important in the selection and dosage of drug therapies for patients."( Comparison of the prevalence of the poor metabolizer phenotype for CYP2D6 between 203 Hmong subjects and 280 white subjects residing in Minnesota.
Hansen, SR; Straka, RJ; Walker, PF, 1995
)
0.29
"Tolerance to the antinociceptive (analgesic) effect of morphine, a mu-opioid agonist, was developed in male CD-1 mice as assessed by a shift to the right of the analgesic (tail-flick) dose-response curves and an increase in the ED50 values."( Dextromethorphan attenuates and reverses analgesic tolerance to morphine.
Elliott, K; Hynansky, A; Inturrisi, CE, 1994
)
1.73
"0 nm, respectively, have been developed for the specific determination of guaiphenesin in the presence of dextromethorphan, drugs with closely overlapping absorption spectra, in synthetic admixtures and in pharmaceutical dosage forms (tablets and syrups)."( Determination of guaiphenesin in anti-tussive pharmaceutical preparations containing dextromethorphan by first- and second-derivative ultraviolet spectrophotometry.
Hu, TM; Lee, AR, 1994
)
0.73
" In many instances, a knowledge of the drug metabolism status of a patient can be helpful in the selection of a drug and its dosage regimen, and in the prediction of possible drug/drug interactions when two or more drugs have to be administered concomitantly."( Polymorphism in the metabolism of drugs, including antidepressant drugs: comments on phenotyping.
Coutts, RT, 1994
)
0.29
" Concentrations in plasma were determined after 7 days of desipramine (50 mg/day) dosing alone, during the 21 days of desipramine and selective serotonin reuptake inhibitor (SSRI) coadministration (fluoxetine, 20 mg/day; sertraline, 50 mg/day), and for 21 days of continued desipramine administration after SSRI discontinuation."( Pharmacokinetics of desipramine coadministered with sertraline or fluoxetine.
Alderman, J; Chung, M; Harris, S; Harrison, W; Messig, M; Preskorn, SH, 1994
)
0.29
" Patients were sequentially dosed with DM 40 mg/6 h (8 weeks) and 50 mg/6 h (8 weeks) while concurrent antiepileptic drugs were kept stable."( Pharmacokinetics of dextromethorphan and dextrorphan in epileptic patients.
Kazis, A; Kimiskidis, V; Niopas, I,
)
0.45
" Serial plasma and urine samples were collected at specified time intervals after dosing on Days 1, 16, 18, 20 and 22."( Single and multiple dose pharmacokinetics of nefazodone in subjects classified as extensive and poor metabolizers of dextromethorphan.
Barbhaiya, RH; Buch, AB; Greene, DS, 1996
)
0.5
"After CYP2D6 phenotype determination with dextromethorphan, 31 patients were treated with oral DMI at a dosage of 100 mg per day for 3 weeks."( Relationship between plasma desipramine levels, CYP2D6 phenotype and clinical response to desipramine: a prospective study.
Avenoso, A; Campo, GM; Caputi, AP; Gitto, C; Perucca, E; Spina, E, 1997
)
0.56
" The two patients with the poor metabolizer phenotype showed the highest plasma concentrations of DMI and complained of severe adverse effects, requiring dosage reduction."( Relationship between plasma desipramine levels, CYP2D6 phenotype and clinical response to desipramine: a prospective study.
Avenoso, A; Campo, GM; Caputi, AP; Gitto, C; Perucca, E; Spina, E, 1997
)
0.3
" In the patient with the apparent poor metabolizer (PM) phenotype, a change in the DM preparation to a sustained-release form and increase in the dosing interval was required to lower DM plasma concentrations."( Dextromethorphan in nonketotic hyperglycinaemia: metabolic variation confounds the dose-response relationship.
Arnold, GL; Griebel, ML; Kearns, GL; Koroma, DM; Valentine, JL, 1997
)
1.74
"3 mm3 (mean +/- standard error of the mean), while animals receiving dextromethorphan had significantly smaller lesions, which was independent of dosing schedule (59."( Neuroprotection by dextromethorphan in acute experimental subdural hematoma in the rat.
Boardman, C; Duhaime, AC; Gennarelli, LM, 1996
)
0.86
" Dosage was titrated in each patient to the highest level reached without disrupting normal activities; mean doses were 381 mg/day in diabetics and 439 mg/day in postherpetic neuralgia patients."( High-dose oral dextromethorphan versus placebo in painful diabetic neuropathy and postherpetic neuralgia.
Max, MB; Nelson, KA; Park, KM; Robinovitz, E; Tsigos, C, 1997
)
0.65
" IB showed a dose-response relationship which appeared to plateau at doses of 50 and 100 mg/kg."( Effects of the combined oral administration of NSAIDs and dextromethorphan on behavioral symptoms indicative of arthritic pain in rats.
Caruso, FS; Frenk, H; Lu, J; Mao, J; Mayer, DJ; Price, DD, 1996
)
0.54
" Therefore the determination of dextromethorphan and metabolites in serum could be advantageous to measure individual CYP2D6 activities in vivo and thus optimize the dosing of drugs metabolized by CYP2D6."( CYP2D6 genotype and phenotyping by determination of dextromethorphan and metabolites in serum of healthy controls and of patients under psychotropic medication.
Fuchs, K; Härtter, S; Hiemke, C; Köhler, D; Sieghart, W, 1997
)
0.83
" The dosage of sertraline ranged from 25 to 150 mg/day (93."( The extent and determinants of changes in CYP2D6 and CYP1A2 activities with therapeutic doses of sertraline.
Herrmann, N; Kalow, W; Naranjo, CA; Ozdemir, V; Reed, K; Sellers, EM; Shulman, RW, 1998
)
0.3
"03 mg/kg/infusion), suggesting that DM shifted the dose-response curve for cocaine self-administration to the left."( Dextromethorphan alters the reinforcing effect of cocaine in the rat.
Hong, SY; Jhoo, WK; Kim, HC; Park, BK, 1997
)
1.74
" Levodopa dose-response curves for antiparkinsonian and dyskinetic effects were then compared for each study arm."( Dextromethorphan improves levodopa-induced dyskinesias in Parkinson's disease.
Chase, TN; Del Dotto, P; Natté, R; van den Munckhof, P; Verhagen Metman, L, 1998
)
1.74
"Intravenous and oral verapamil clearance values were significantly correlated, and cumulative dextromethorphan/3-methoxymorphinan urinary ratios correlated with both plasma AUC(norverapamil)/AUC(verapamil) after oral verapamil dosing and with oral and intravenous verapamil clearance."( In vivo comparison of putative probes of CYP3A4/5 activity: erythromycin, dextromethorphan, and verapamil.
Barnas, CR; Gorski, JC; Krecic-Shepard, ME; Schwartz, JB; Slimko, J; Wainer, IW, 1999
)
0.75
" We compared metabolic ratios from six different dextromethorphan phenotyping doses to ascertain whether linearity existed over a dosage range."( Dose dependency of dextromethorphan for cytochrome P450 2D6 (CYP2D6) phenotyping.
Bertino, JS; Ellis, RE; Gaedigk, A; Gotschall, R; Kearns, GL; Leeder, JS; Nafziger, AN; Streetman, DS, 1999
)
0.89
"9-fold rightward shift of the cumulative dose-response curves."( Clinically available NMDA receptor antagonists memantine and dextromethorphan reverse existing tolerance to the antinociceptive effects of morphine in mice.
Danysz, W; Kozela, E; Popik, P, 2000
)
0.55
"A high performance liquid chromatography procedure has been developed for the simultaneous determination of guaifenesin pseudoephedrine-dextromethorphan and guaifenesin-pseudoephedrine in commercially available capsule dosage forms and guaifenesin-codeine in a commercial cough syrup dosage form."( HPLC determination of guaifenesin with selected medications on underivatized silica with an aqueous-organic mobile phase.
Stewart, JT; Wilcox, ML, 2000
)
0.51
" Dosage was titrated in each patient to the highest level reached without disrupting normal activities."( A randomized, controlled trial of high-dose dextromethorphan in facial neuralgias.
Booher, SL; Gilron, I; Max, MB; Rowan, MS; Smoller, MS, 2000
)
0.57
" Knowledge of the percolation threshold -- one of the most important concepts in percolation theory -- results in a clear improvement of the solid dosage form design."( Estimation of the percolation thresholds in dextromethorphan hydrobromide matrices.
Caraballo, I; Melgoza, LM; Rabasco, AM; Sandoval, H, 2001
)
0.57
"The inhibition potential of drugs towards five major human hepatic cytochrome P450 (CYP) isozymes (CYP2A6, 3A4, 2C9, 2D6, and 2E1) was investigated via cassette dosing of the five probe substrates (coumarin, midazolam, tolbutamide, dextromethorphan, and chlorzoxazone) in human liver microsomes using a 96-well plate format."( High-throughput cytochrome P450 (CYP) inhibition screening via a cassette probe-dosing strategy. VI. Simultaneous evaluation of inhibition potential of drugs on human hepatic isozymes CYP2A6, 3A4, 2C9, 2D6 and 2E1.
Bu, HZ; Knuth, K; Magis, L; Teitelbaum, P, 2001
)
0.49
" Three days of validation data are provided along with study sample data from a patient dosed with commercially available Vicks 44."( Increasing bioanalytical throughput using pcSFC-MS/MS: 10 minutes per 96-well plate.
Bolden, RD; Hoke, SH; Morand, KL; Pinkston, JD; Tomlinson, JA; Wehmeyer, KR, 2001
)
0.31
" Tolbutamide (CYP2C9), caffeine (CYP1A2), dextromethorphan (CYP2D6), oral midazolam (intestinal wall and hepatic CYP3A), and intravenous midazolam (hepatic CYP3A) were administered before, with short-term St John's wort dosing (900 mg), and after 2 weeks of intake (300 mg 3 times a day) to determine CYP activities."( The effects of St John's wort (Hypericum perforatum) on human cytochrome P450 activity.
Gorski, JC; Hall, SD; Hamman, MA; Huang, SM; Lesko, LJ; Wang, Z, 2001
)
0.57
" active placebo [lorazepam]) and (2) dose-response trial of the preferred active drug in responders from the first study (0% vs."( Dextromethorphan and memantine in painful diabetic neuropathy and postherpetic neuralgia: efficacy and dose-response trials.
Booher, S; Gilron, I; Max, MB; Parada, S; Sang, CN, 2002
)
1.76
" Blood pressure and heart rate were also monitored before dosing and after the dosing regimen."( Assessment of the effect of dextromethorphan and ketamine on the acute nociceptive threshold and wind-up of the second pain response in healthy male volunteers.
Fisher, G; Growcott, JW; Hughes, AM; Rhodes, J; Sellers, M, 2002
)
0.61
"For paroxetine and fluoxetine, plasma concentrations and dosage strongly influence the magnitude of enzyme inhibition."( CYP2D6 inhibition by selective serotonin reuptake inhibitors: analysis of achievable steady-state plasma concentrations and the effect of ultrarapid metabolism at CYP2D6.
Alfaro, CL; Ereshefsky, L; Gaedigk, A; Lam, YW; Simpson, J, 2002
)
0.31
"Oral dextromethorphan (120-180 mg daily) was administered to three selected cancer patients during a 3-week study and an additional 1 month of treatment with the dosage subjectively judged to be best."( Dextromethorphan for phantom pain attenuation in cancer amputees: a double-blind crossover trial involving three patients.
Ben Abraham, R; Kollender, Y; Marouani, N; Meller, I; Weinbroum, AA,
)
2.09
"Further clinical trials are warranted to determine the optimal dosage and identify which patients with phantom pain would benefit the most from this therapeutic approach."( Dextromethorphan for phantom pain attenuation in cancer amputees: a double-blind crossover trial involving three patients.
Ben Abraham, R; Kollender, Y; Marouani, N; Meller, I; Weinbroum, AA,
)
1.57
" Using a mouse hot-plate test, dose-response relationships were first determined for all compounds individually and then for opioids co-administered with fixed doses of ketamine or dextromethorphan."( Dextromethorphan and ketamine potentiate the antinociceptive effects of mu- but not delta- or kappa-opioid agonists in a mouse model of acute pain.
Baker, AK; Hoffmann, VL; Meert, TF, 2002
)
1.95
" Using a mouse hot-plate test, dose-response relationships were first determined for all compounds individually and then for opioids co-administered with fixed doses of clonidine, ketamine or dextromethorphan."( Interactions of NMDA antagonists and an alpha 2 agonist with mu, delta and kappa opioids in an acute nociception assay.
Baker, AK; Hoffmann, VL; Meert, TF, 2002
)
0.5
"In this study, the metabolic ratios of dextromethorphan to dextrorphan (DM/DX) in plasma were calculated at steady state after administering 2 dosage forms (Medicon) and Detusiv) of DM with different release rates."( Analysis of pharmacokinetic parameters for assessment of dextromethorphan metabolic phenotypes.
Chen, JY; Ho, HO; Lee, YJ; Sheu, MT; Tao, PL; Yeh, GC,
)
0.65
" After 14 days of St John's wort administration, participants were given the probe drugs along with 1 St John's wort tablet to establish postadministration CYP activity; the St John's wort dosing regimen was continued for 48 hours."( Effect of St John's wort on drug metabolism by induction of cytochrome P450 3A4 enzyme.
Chavin, KD; DeVane, CL; Donovan, JL; Markowitz, JS; Ruan, Y; Taylor, RM; Wang, JS, 2003
)
0.32
" This suggests that long-term administration of St John's wort may result in diminished clinical effectiveness or increased dosage requirements for all CYP 3A4 substrates, which represent at least 50% of all marketed medications."( Effect of St John's wort on drug metabolism by induction of cytochrome P450 3A4 enzyme.
Chavin, KD; DeVane, CL; Donovan, JL; Markowitz, JS; Ruan, Y; Taylor, RM; Wang, JS, 2003
)
0.32
" Naloxone, which had no effect on the dextromethorphan dose-response relation, abolished the synergism."( Morphine potentiates dextromethorphan-induced vasodilation in rat superior mesenteric artery.
Inan, S; Tallarida, RJ, 2004
)
0.91
"The aim of this work was to determine whether a dose-response relationship existed among a group of children administered a single nocturnal dose of DM for cough due to an upper respiratory tract infection."( Dose-response relationship with increasing doses of dextromethorphan for children with cough.
Baker, MS; Berlin, CM; Paul, IM; Shaffer, ML; Sturgis, SA; Yoder, KE, 2004
)
0.57
" The direction of dosing adjustments during pregnancy will depend on the drug and the enzyme that is responsible for its metabolism."( Temporal changes in drug metabolism (CYP1A2, CYP2D6 and CYP3A Activity) during pregnancy.
Caritis, SN; Glover, DD; Tracy, TS; Venkataramanan, R, 2005
)
0.33
"Twenty-eight randomized, double-blind, clinical studies, with 40 comparisons, including a variety of dosing regimens comparing DM treatment with placebo, were included."( A qualitative systematic review of peri-operative dextromethorphan in post-operative pain.
Dahl, JB; Duedahl, TH; Møiniche, S; Rømsing, J, 2006
)
0.59
" Subsequently blood and urine samples were collected after drug dosing as before."( Effect of honey on CYP3A4, CYP2D6 and CYP2C19 enzyme activity in healthy human volunteers.
Adithan, C; Rajan, S; Shashindran, C; Tushar, T; Vinod, T, 2007
)
0.34
" The objective measure of cough reflex sensitivity was the citric acid, dose-response cough challenge."( Assessment of antitussive efficacy of dextromethorphan in smoking related cough: objective vs. subjective measures.
Hull, D; Morice, AH; Ramsay, J; Thompson, R; Wright, C, 2008
)
0.62
" The risk of overdose, incorrect dosing and adverse events is increased in young children due to the greater number of colds they acquire each year."( Over-the-counter cough and cold medication use in young children.
Brewer, M; Ryan, T; Small, L,
)
0.13
" In vivo assessment of CYP1A2 and CYP3A4 activities, perhaps by phenotyping approaches, could assist the optimization of CLZ dosage and minimize pharmacokinetic interactions with coadministered drugs."( Interindividual variation in relative CYP1A2/3A4 phenotype influences susceptibility of clozapine oxidation to cytochrome P450-specific inhibition in human hepatic microsomes.
D'Esposito, F; Edwards, RJ; Murray, M; Ramzan, I; Zhang, WV, 2008
)
0.35
" Arterial rings were mounted in isolated tissue chambers equipped with isometric tension transducers to obtain pharmacologic dose-response curves."( N-methyl-D-aspartate (NMDA) antagonists--S(+)-ketamine, dextrorphan, and dextromethorphan--act as calcium antagonists on bovine cerebral arteries.
Carlsson, C; Chen, D; Harakal, C; Kamel, IR; Wendling, KS; Wendling, WW, 2008
)
0.58
"0 x 10(-3) M DXM hydrobromide in pure as well as in dosage forms by direct potentiometry and standard addition methods."( Polymeric membrane sensors for the selective determination of dextromethorphan in pharmaceutical preparations.
El-Naby, EH, 2008
)
0.59
" Information on a respective dose-response relationship is not available."( Dose-response relationship for the pharmacokinetic interaction of grapefruit juice with dextromethorphan investigated by human urinary metabolite profiles.
Bittner, N; Lutz, U; Lutz, WK; Strauch, K, 2009
)
0.58
" In an acute (3 day) dose-response study, anesthetized male Sprague-Dawley rats were exposed to a unilateral frontal PBBI with DM (0."( Neuroprotective profile of dextromethorphan in an experimental model of penetrating ballistic-like brain injury.
Lu, XC; Sharrow, K; Shear, DA; Tortella, FC; Williams, AJ, 2009
)
0.65
" After cerebral infarction, intravenous dosing of saline had no effect on these parameters."( Ameliorating effects of cloperastine on dysfunction of the urinary bladder caused by cerebral infarction in conscious rats.
Shirasaki, T; Soeda, F; Takahama, K; Yamamoto, G, 2009
)
0.35
"Compared to the no quinidine condition, quinidine pre-treatment decreased the area under the dose-response curve on subjective measures of positively reinforcing effects (e."( Effect of metabolic blockade on the psychoactive effects of dextromethorphan.
Busto, U; Sellers, EM; Tyndale, RF; Zawertailo, LA, 2010
)
0.6
" Of particular concern to the FDA were increased QTc intervals reported in patients dosed with a 30-/30-mg dose of DM/Q."( AVP-923, a combination of dextromethorphan hydrobromide and quinidine sulfate for the treatment of pseudobulbar affect and neuropathic pain.
Olney, N; Rosen, H, 2010
)
0.66
" Other endpoints showing statistically significant DMq benefit included, for both dosage levels, the likelihood of PBA remission during the final 14 days and, for the higher dosage, improvement on measures of social functioning and mental health."( Dextromethorphan plus ultra low-dose quinidine reduces pseudobulbar affect.
Brooks, BR; Cummings, J; Hepner, A; Kaye, R; Pioro, EP; Schiffer, R; Thisted, RA; Wynn, D, 2010
)
1.8
"First, dose-response curves for dextromethorphan, dextrorphan, and bupivacaine (n = 8 at each testing point) were determined for cutaneous analgesia on the rat back, and equipotent doses were calculated."( Systemic dextromethorphan and dextrorphan are less toxic in rats than bupivacaine at equianesthetic doses.
Chen, YC; Chen, YW; Hung, CH; Liu, TY; Wang, JJ, 2011
)
1.07
" This test could contribute to the personalization and optimization of tamoxifen treatment, but it needs additional validation and simplification before being applicable in future dosing strategies."( Dextromethorphan as a phenotyping test to predict endoxifen exposure in patients on tamoxifen treatment.
Beijnen, JH; de Graan, AJ; de Jongh, FE; de Vos, AI; de Vos, FY; Loos, WJ; Mathijssen, RH; Seynaeve, C; Teunissen, SF; van Alphen, RJ; van der Holt, B; van Schaik, RH; Verweij, J, 2011
)
1.81
" The approved dosage of DMQ is DM 20 mg and Q 10 mg twice daily."( Randomized open-label drug-drug interaction trial of dextromethorphan/quinidine and paroxetine in healthy volunteers.
Pope, LE; Schoedel, KA; Sellers, EM, 2012
)
0.63
" Thus, patients should be monitored for AEs and dosage adjustment considered when combining these two agents."( Randomized open-label drug-drug interaction trial of dextromethorphan/quinidine and paroxetine in healthy volunteers.
Pope, LE; Schoedel, KA; Sellers, EM, 2012
)
0.63
" Concomitant use of drugs metabolized by CYP2D6 may require dosage adjustment."( Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes.
Bekersky, I; Blum, RA; Tolbert, D; Walzer, M, 2012
)
0.38
"Orally disintegrating tablets (ODTs), which disintegrate rapidly (<1 min) in the mouth and do not require water for administration, have become a very popular dosage form."( Development and optimization of dextromethorphan hydrobromide oral disintegrating tablets: effect of formulation and process variables.
Ibrahim, MA; Mostafa, HF; Sakr, A,
)
0.41
" Therefore, we asked if a physiologically based pharmacokinetic (PBPK) model could be used to evaluate different dosing regimens for pregnant women."( A physiologically based pharmacokinetic model to predict disposition of CYP2D6 and CYP1A2 metabolized drugs in pregnant women.
Isoherranen, N; Ke, AB; Nallani, SC; Rostami-Hodjegan, A; Unadkat, JD; Zhao, P, 2013
)
0.39
" However, a higher dosage of DXM antagonized the differentiation of osteoblasts via the inhibition of osteogenic signals and increase of adipogenic signals."( Dextromethorphan inhibits osteoclast differentiation by suppressing RANKL-induced nuclear factor-κB activation.
Chen, YR; Fu, WM; Lin, TH; Liou, HC; Lu, DH; Wu, K; Yang, RS, 2013
)
1.83
" The former, on sale without prescription, presents a varied symptomatology, dosage and dependent metabolic action, ranging from euphoria to hallucinations."( [Emergent drugs (II): the Pharming phenomenon].
Aldea-Perona, A; Burillo-Putze, G; Climent, B; Dueñas, A; García-Sáiz, MM; Hoffman, RS; Munné, P; Nogué, S; Rodríguez-Jiménez, C,
)
0.13
" However, C-10068 failed to improve PBBI-induced motor and cognitive functions with the dosing regimen used in this study."( Dual Therapeutic Effects of C-10068, a Dextromethorphan Derivative, Against Post-Traumatic Nonconvulsive Seizures and Neuroinflammation in a Rat Model of Penetrating Ballistic-Like Brain Injury.
Bridson, GW; Chen, Z; Graham, PB; Leung, LY; Lu, XC; Shear, DA; Tortella, FC; Uttamsingh, V, 2015
)
0.69
" NIC abolished anodal tDCS-induced motor cortex excitability enhancement, which was restituted under medium dosage of DMO."( Mechanisms of Nicotinic Modulation of Glutamatergic Neuroplasticity in Humans.
Batsikadze, G; Fresnoza, S; Grundey, J; Kuo, MF; Lugon, MDMV; Nakamura-Palacios, EM; Nitsche, MA; Paulus, W, 2017
)
0.46
" The outcome on developmental progress and seizures was compared between the first child diagnosed and treated late with the second child diagnosed at birth and treated aggressively from the newborn period using dextromethorphan and benzoate at dosing sufficient to normalize plasma glycine levels."( Neurodevelopmental Outcome and Treatment Efficacy of Benzoate and Dextromethorphan in Siblings with Attenuated Nonketotic Hyperglycinemia.
Ahmad, A; Bjoraker, KJ; Burns, C; Christodoulou, J; Coughlin, CR; Creadon-Swindell, G; Dyack, S; Fergeson, M; Friederich, MW; Gaughan, S; Hodge, MA; Spector, EB; Swanson, MA; Tan, ES; Van Hove, JL, 2016
)
0.86
" However, the optimal dose and safety of chronic dosing are not fully known."( Dextromethorphan: An update on its utility for neurological and neuropsychiatric disorders.
Cavendish, JZ; Crowe, MS; Lucke-Wold, BP; Matsumoto, RR; Nguyen, L; Thomas, KL, 2016
)
1.88
" Metabolic ratios (MRs) from AUC∞ after single dosing and from AUC0-12h at steady state were taken as the gold standard."( Alternative methods for CYP2D6 phenotyping: comparison of dextromethorphan metabolic ratios from AUC, single point plasma, and urine.
Chen, R; Hu, P; Shi, J; Wang, H, 2016
)
0.68
" Present investigation was carried out in rats to study influence of corticosteroids after repeated dosing with/without pre-treatment with CYP2D inhibitor quinidine on the CYP2D1 mRNA levels and CYP2D enzyme activity using dextromethorphan as probe substrate."( Consequences of daily corticosteroid dosing with or without pre-treatment with quinidine on the in vivo cytochrome P450 2D (CYP2D) enzyme in rats: effect on O-demethylation activity of dextromethorphan and expression levels of CYP2D1 mRNA.
Delvadia, P; Giri, P; Gupta, L; Lad, K; Patel, HM; Patel, N; Srinivas, NR; Trivedi, P, 2018
)
0.86
" High dosage of DXM can induce euphoria, dissociative effects and even hallucinations."( Protective effect of Lycium Barbarum polysaccharides on dextromethorphan-induced mood impairment and neurogenesis suppression.
Chan, JN; Fung, TK; Lau, BW; Leung, JW; Po, KK; Sánchez-Vidaña, DI; Sin, EL; Siu, AM; So, KF, 2017
)
0.7
" Following dosing and euthanasia, rats decomposed outdoors to skeleton in two different microclimate environments (n = 5 ACU and n = 5 REP at each site): Site A (shaded forest microenvironment) and Site B (rocky substrate exposed to direct sunlight, 600 m from Site A)."( Analysis of Dextromethorphan and Dextrorphan in Skeletal Remains Following Differential Microclimate Exposure: Comparison of Acute vs. Repeated Drug Exposure.
Morrison, LM; Unger, KA; Watterson, JH, 2017
)
0.83
"To evaluate the capacity for modafinil to be a perpetrator of metabolic drug-drug interactions by altering cytochrome P450 activity following a single dose and dosing to steady state."( Evaluation of modafinil as a perpetrator of metabolic drug-drug interactions using a model informed cocktail reaction phenotyping trial protocol.
Mangoni, AA; Rowland, A; Sorich, MJ; van Dyk, M; Warncken, D, 2018
)
0.48
"These data support consideration of the risk of clinically relevant metabolic drug-drug interactions perpetrated by modafinil when this drug is co-administered with drugs that are primarily cleared by CYP2C19 (single modafinil dose or steady state modafinil dosing) or CYP3A4 (steady state modafinil dosing only) catalysed metabolic pathways."( Evaluation of modafinil as a perpetrator of metabolic drug-drug interactions using a model informed cocktail reaction phenotyping trial protocol.
Mangoni, AA; Rowland, A; Sorich, MJ; van Dyk, M; Warncken, D, 2018
)
0.48
" The in vitro data predicted ~ 50% decrease in CYP2D6 activity in humans after dosing with 13cisRA."( Does In Vitro Cytochrome P450 Downregulation Translate to In Vivo Drug-Drug Interactions? Preclinical and Clinical Studies With 13-cis-Retinoic Acid.
Amory, JK; Hogarth, C; Isoherranen, N; Kenny, JR; Kosaka, M; Stevison, F; Wong, S, 2019
)
0.51
" He reported a subjective need to increase his dosing over time to achieve the same degree of intoxication."( Ten Years of Robotripping: Evidence of Tolerance to Dextromethorphan Hydrobromide in a Long-Term User.
Boley, SP; LeRoy, JM; Olives, TD; Stellpflug, SJ, 2019
)
0.76
" The population-based human study analyzed the data from the Taiwan Longitudinal Health Insurance Database 2005 between January 2005 and December 2013 and then used the DXM dose-response curve to investigate the trend of its protective effect against PD."( The effect of dextromethorphan use in Parkinson's disease: A 6-hydroxydopamine rat model and population-based study.
Cheng, CY; Chien, WC; Chiu, CH; Huang, YS; Kao, LT; Li, IH; Liu, CT; Ma, KH; Shih, JH; Shiue, CY, 2019
)
0.87
"Ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-QToF-MS) analysis of dextromethorphan (DXM) and its metabolites-dextrorphan, 3-methoxymorphinan (3-MEM) and 3-hydroxymorphinan-in skeletal remains of rats exposed to DXM under different dosing patterns is described."( Analysis of Dextromethorphan and Three Metabolites in Decomposed Skeletal Tissues by UPLC-QToF-MS: Comparison of Acute and Repeated Drug Exposures.
Cornthwaite, HM; McDonald, CS; Watterson, JH, 2019
)
1.1
" Diphenhydramine and dextromethorphan dosing errors were the most common cause of medication errors resulting from CCM use."( Medication Errors From Over-the-Counter Cough and Cold Medications in Children.
Banner, W; Bond, GR; Dart, RC; Green, JL; Kauffman, RE; Palmer, RB; Paul, IM; Rapp-Olsson, M; Reynolds, KM; Wang, GS, 2020
)
0.88
" Blood samples were collected 3 h after dosing and plasma separated."( Cytochrome P
Chatterjee, S; Dhuya, M; Gogtay, N; Hazra, A; Pal, MM,
)
0.13
" Incorporation of genotype-based dosing guidelines should be considered for CYP2D6 substrates given the prevalent use of these agents in this pediatric age group."( A longitudinal study of cytochrome P450 2D6 (CYP2D6) activity during adolescence.
Gaedigk, A; Leeder, JS; Lin, YS; Pearce, RE; Soden, SE; Staggs, VS; Wright, KJ, 2022
)
0.72
"This study has a number of strengths including adequate power, a randomized controlled design, and an evidence-based dosing schedule."( A double-blinded, placebo-controlled, randomized study to evaluate the efficacy of perioperative dextromethorphan compared to placebo for the treatment of postoperative pain: a study protocol.
Christ, AB; Gilbert, PK; Gucev, GV; Heckmann, ND; Jones, IA; Lieberman, JR; Longjohn, DB; Oakes, DA; Piple, AS; Ratto, CE; Yan, PY, 2023
)
1.13
" Eligible subjects met entry criteria and qualified by completing a run-in period where coughs were recorded with a cough monitor after they were dosed with sweet syrup."( Objective and self-reported evidence of dextromethorphan antitussive efficacy in children, aged 6-11 years, with acute cough due to the common cold.
Cruz-Rivera, M; Gelotte, CK; Jayawardena, S; Leyva, RA; Meeves, SG; Moreira, SA; Wilson, BL, 2023
)
1.18
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (7)

RoleDescription
NMDA receptor antagonistAny substance that inhibits the action of N-methyl-D-aspartate (NMDA) receptors. They tend to induce a state known as dissociative anesthesia, marked by catalepsy, amnesia, and analgesia, while side effects can include hallucinations, nightmares, and confusion. Due to their psychotomimetic effects, many NMDA receptor antagonists are used as recreational drugs.
neurotoxinA poison that interferes with the functions of the nervous system.
xenobioticA xenobiotic (Greek, xenos "foreign"; bios "life") is a compound that is foreign to a living organism. Principal xenobiotics include: drugs, carcinogens and various compounds that have been introduced into the environment by artificial means.
environmental contaminantAny minor or unwanted substance introduced into the environment that can have undesired effects.
antitussiveAn agent that suppresses cough. Antitussives have a central or a peripheral action on the cough reflex, or a combination of both. Compare with expectorants, which are considered to increase the volume of secretions in the respiratory tract, so facilitating their removal by ciliary action and coughing, and mucolytics, which decrease the viscosity of mucus, facilitating its removal by ciliary action and expectoration.
prodrugA compound that, on administration, must undergo chemical conversion by metabolic processes before becoming the pharmacologically active drug for which it is a prodrug.
oneirogenAny substance that produces or enhances dream-like states of consciousness.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (1)

ClassDescription
6-methoxy-11-methyl-1,3,4,9,10,10a-hexahydro-2H-10,4a-(epiminoethano)phenanthreneAn organic heterotetracyclic compound that is 1,3,4,9,10,10a-hexahydro-2H-10,4a-(epiminoethano)phenanthrene which is substituted by a methoxy group at position 6 and a methyl group at position 11.
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Pathways (10)

PathwayProteinsCompounds
Metabolism14961108
Biological oxidations150276
Phase I - Functionalization of compounds69175
Cytochrome P450 - arranged by substrate type30110
Xenobiotics450
Disease1278231
Infectious disease89579
SARS-CoV Infections28229
Potential therapeutics for SARS592
Viral Infection Pathways72739

Protein Targets (36)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
thyroid stimulating hormone receptorHomo sapiens (human)Potency0.00630.001318.074339.8107AID926; AID938
cytochrome P450 2D6 isoform 1Homo sapiens (human)Potency2.51190.00207.533739.8107AID891
muscarinic acetylcholine receptor M1Rattus norvegicus (Norway rat)Potency10.88200.00106.000935.4813AID943; AID944
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Solute carrier family 22 member 1 Homo sapiens (human)IC50 (µMol)12.72500.21005.553710.0000AID1442001; AID1526751
Glutamate receptor ionotropic, NMDA 2DHomo sapiens (human)Ki1.68000.00120.70256.0000AID156805
Glutamate receptor ionotropic, NMDA 3BHomo sapiens (human)Ki1.68000.00120.70256.0000AID156805
Neuronal acetylcholine receptor subunit alpha-3Rattus norvegicus (Norway rat)IC50 (µMol)11.05000.00300.77706.0000AID1174934
Sodium channel protein type 1 subunit alphaRattus norvegicus (Norway rat)IC50 (µMol)5.65000.01001.14052.9390AID179553; AID205275
Sodium channel protein type 2 subunit alphaRattus norvegicus (Norway rat)IC50 (µMol)5.65000.00401.14854.7300AID179553; AID205275
Sodium channel protein type 3 subunit alphaRattus norvegicus (Norway rat)IC50 (µMol)5.65000.00600.86052.9390AID179553; AID205275
Cytochrome P450 2D26Rattus norvegicus (Norway rat)IC50 (µMol)5.60000.09402.16285.6000AID54564
Cytochrome P450 2D6Homo sapiens (human)IC50 (µMol)2.17680.00002.015110.0000AID1862576; AID1884504; AID262947; AID54564; AID54570
Muscarinic acetylcholine receptor M2Rattus norvegicus (Norway rat)Ki10.00000.00010.58908.2600AID142870
Cytochrome P450 2C9 Homo sapiens (human)IC50 (µMol)50.00000.00002.800510.0000AID1210069
Neuronal acetylcholine receptor subunit beta-4Rattus norvegicus (Norway rat)IC50 (µMol)11.05000.00300.88696.0000AID1174934
Sodium-dependent serotonin transporterHomo sapiens (human)IC50 (µMol)0.00270.00010.86458.7096AID625222
Sodium-dependent serotonin transporterHomo sapiens (human)Ki0.00140.00000.70488.1930AID625222
Glutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)Ki2.24600.00030.86666.6900AID157456
Cytochrome P450 2J2Homo sapiens (human)IC50 (µMol)50.00000.01202.53129.4700AID1210069
Glutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)Ki2.24600.00030.68056.6900AID157456
Glutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)Ki2.24600.00030.70716.6900AID157456
Glutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)Ki2.24600.00030.81966.6900AID157456
Glutamate receptor ionotropic, NMDA 1Homo sapiens (human)Ki1.68000.00120.48246.0000AID156805
Glutamate receptor ionotropic, NMDA 2AHomo sapiens (human)Ki1.68000.00120.62056.0000AID156805
Glutamate receptor ionotropic, NMDA 2BHomo sapiens (human)Ki1.68000.00120.32686.0000AID156805
Glutamate receptor ionotropic, NMDA 2CHomo sapiens (human)Ki1.68000.00120.70256.0000AID156805
Sigma non-opioid intracellular receptor 1Cavia porcellus (domestic guinea pig)IC50 (µMol)0.12360.00202.123310.0000AID317399; AID317400
Sigma non-opioid intracellular receptor 1Cavia porcellus (domestic guinea pig)Ki0.03650.00000.338510.0000AID1744232; AID1744242
Glutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)Ki2.24600.00030.70726.6900AID157456
Cytochrome P450 2D4Rattus norvegicus (Norway rat)IC50 (µMol)136.00001.70001.70001.7000AID54566
Glutamate receptor ionotropic, NMDA 3AHomo sapiens (human)Ki1.68000.00120.70256.0000AID156805
Glutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)Ki2.24600.00030.70726.6900AID157456
Sigma non-opioid intracellular receptor 1Homo sapiens (human)Ki0.34800.00000.490110.0000AID204634
Sigma non-opioid intracellular receptor 1Rattus norvegicus (Norway rat)Ki5.07000.00030.26715.0700AID203818
Glutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)Ki2.24600.00030.70726.6900AID157456
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Taste receptor type 2 member 46Homo sapiens (human)EC50 (µMol)31.00000.00493.09838.6000AID1622651
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Cytochrome P450 2D6Homo sapiens (human)Km1.40001.10003.72868.2000AID1209237
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (150)

Processvia Protein(s)Taxonomy
xenobiotic metabolic processSolute carrier family 22 member 1 Homo sapiens (human)
neurotransmitter transportSolute carrier family 22 member 1 Homo sapiens (human)
serotonin transportSolute carrier family 22 member 1 Homo sapiens (human)
establishment or maintenance of transmembrane electrochemical gradientSolute carrier family 22 member 1 Homo sapiens (human)
organic cation transportSolute carrier family 22 member 1 Homo sapiens (human)
quaternary ammonium group transportSolute carrier family 22 member 1 Homo sapiens (human)
prostaglandin transportSolute carrier family 22 member 1 Homo sapiens (human)
monoamine transportSolute carrier family 22 member 1 Homo sapiens (human)
putrescine transportSolute carrier family 22 member 1 Homo sapiens (human)
spermidine transportSolute carrier family 22 member 1 Homo sapiens (human)
acetylcholine transportSolute carrier family 22 member 1 Homo sapiens (human)
dopamine transportSolute carrier family 22 member 1 Homo sapiens (human)
norepinephrine transportSolute carrier family 22 member 1 Homo sapiens (human)
thiamine transportSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic transportSolute carrier family 22 member 1 Homo sapiens (human)
epinephrine transportSolute carrier family 22 member 1 Homo sapiens (human)
serotonin uptakeSolute carrier family 22 member 1 Homo sapiens (human)
norepinephrine uptakeSolute carrier family 22 member 1 Homo sapiens (human)
thiamine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
metanephric proximal tubule developmentSolute carrier family 22 member 1 Homo sapiens (human)
purine-containing compound transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
dopamine uptakeSolute carrier family 22 member 1 Homo sapiens (human)
monoatomic cation transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
transport across blood-brain barrierSolute carrier family 22 member 1 Homo sapiens (human)
(R)-carnitine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
acyl carnitine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
spermidine transmembrane transportSolute carrier family 22 member 1 Homo sapiens (human)
cellular detoxificationSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic transport across blood-brain barrierSolute carrier family 22 member 1 Homo sapiens (human)
startle responseGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
adult locomotory behaviorGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of sensory perception of painGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
cellular response to L-glutamateGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
protein insertion into membraneGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
regulation of calcium ion transportGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
calcium ion transmembrane transportGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
modulation of chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
xenobiotic metabolic processCytochrome P450 2D6Homo sapiens (human)
steroid metabolic processCytochrome P450 2D6Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2D6Homo sapiens (human)
estrogen metabolic processCytochrome P450 2D6Homo sapiens (human)
coumarin metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
alkaloid catabolic processCytochrome P450 2D6Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2D6Homo sapiens (human)
isoquinoline alkaloid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2D6Homo sapiens (human)
retinol metabolic processCytochrome P450 2D6Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2D6Homo sapiens (human)
negative regulation of bindingCytochrome P450 2D6Homo sapiens (human)
oxidative demethylationCytochrome P450 2D6Homo sapiens (human)
negative regulation of cellular organofluorine metabolic processCytochrome P450 2D6Homo sapiens (human)
arachidonic acid metabolic processCytochrome P450 2D6Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2C9 Homo sapiens (human)
steroid metabolic processCytochrome P450 2C9 Homo sapiens (human)
cholesterol metabolic processCytochrome P450 2C9 Homo sapiens (human)
estrogen metabolic processCytochrome P450 2C9 Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 2C9 Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
urea metabolic processCytochrome P450 2C9 Homo sapiens (human)
monocarboxylic acid metabolic processCytochrome P450 2C9 Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 2C9 Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
amide metabolic processCytochrome P450 2C9 Homo sapiens (human)
icosanoid biosynthetic processCytochrome P450 2C9 Homo sapiens (human)
oxidative demethylationCytochrome P450 2C9 Homo sapiens (human)
omega-hydroxylase P450 pathwayCytochrome P450 2C9 Homo sapiens (human)
monoamine transportSodium-dependent serotonin transporterHomo sapiens (human)
response to hypoxiaSodium-dependent serotonin transporterHomo sapiens (human)
neurotransmitter transportSodium-dependent serotonin transporterHomo sapiens (human)
response to nutrientSodium-dependent serotonin transporterHomo sapiens (human)
memorySodium-dependent serotonin transporterHomo sapiens (human)
circadian rhythmSodium-dependent serotonin transporterHomo sapiens (human)
response to xenobiotic stimulusSodium-dependent serotonin transporterHomo sapiens (human)
response to toxic substanceSodium-dependent serotonin transporterHomo sapiens (human)
positive regulation of gene expressionSodium-dependent serotonin transporterHomo sapiens (human)
positive regulation of serotonin secretionSodium-dependent serotonin transporterHomo sapiens (human)
negative regulation of cerebellar granule cell precursor proliferationSodium-dependent serotonin transporterHomo sapiens (human)
negative regulation of synaptic transmission, dopaminergicSodium-dependent serotonin transporterHomo sapiens (human)
response to estradiolSodium-dependent serotonin transporterHomo sapiens (human)
social behaviorSodium-dependent serotonin transporterHomo sapiens (human)
vasoconstrictionSodium-dependent serotonin transporterHomo sapiens (human)
sperm ejaculationSodium-dependent serotonin transporterHomo sapiens (human)
negative regulation of neuron differentiationSodium-dependent serotonin transporterHomo sapiens (human)
positive regulation of cell cycleSodium-dependent serotonin transporterHomo sapiens (human)
negative regulation of organ growthSodium-dependent serotonin transporterHomo sapiens (human)
behavioral response to cocaineSodium-dependent serotonin transporterHomo sapiens (human)
enteric nervous system developmentSodium-dependent serotonin transporterHomo sapiens (human)
brain morphogenesisSodium-dependent serotonin transporterHomo sapiens (human)
serotonin uptakeSodium-dependent serotonin transporterHomo sapiens (human)
membrane depolarizationSodium-dependent serotonin transporterHomo sapiens (human)
platelet aggregationSodium-dependent serotonin transporterHomo sapiens (human)
cellular response to retinoic acidSodium-dependent serotonin transporterHomo sapiens (human)
cellular response to cGMPSodium-dependent serotonin transporterHomo sapiens (human)
regulation of thalamus sizeSodium-dependent serotonin transporterHomo sapiens (human)
conditioned place preferenceSodium-dependent serotonin transporterHomo sapiens (human)
sodium ion transmembrane transportSodium-dependent serotonin transporterHomo sapiens (human)
amino acid transportSodium-dependent serotonin transporterHomo sapiens (human)
fatty acid metabolic processCytochrome P450 2J2Homo sapiens (human)
icosanoid metabolic processCytochrome P450 2J2Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 2J2Homo sapiens (human)
regulation of heart contractionCytochrome P450 2J2Homo sapiens (human)
epoxygenase P450 pathwayCytochrome P450 2J2Homo sapiens (human)
linoleic acid metabolic processCytochrome P450 2J2Homo sapiens (human)
organic acid metabolic processCytochrome P450 2J2Homo sapiens (human)
detection of chemical stimulus involved in sensory perception of bitter tasteTaste receptor type 2 member 46Homo sapiens (human)
G protein-coupled receptor signaling pathwayTaste receptor type 2 member 46Homo sapiens (human)
cellular response to amyloid-betaGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
monoatomic cation transportGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
visual learningGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of calcium ion transport into cytosolGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
propylene metabolic processGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of membrane potentialGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
protein heterotetramerizationGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium ion homeostasisGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of reactive oxygen species biosynthetic processGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
response to glycineGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
cellular response to amyloid-betaGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
startle responseGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to amphetamineGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
learning or memoryGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
memoryGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
visual learningGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to xenobiotic stimulusGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to woundingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
sensory perception of painGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
neurogenesisGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
protein catabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
sleepGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
directional locomotionGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
negative regulation of protein catabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
dopamine metabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
serotonin metabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
positive regulation of apoptotic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
activation of cysteine-type endopeptidase activityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
protein localization to postsynaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
learning or memoryGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
protein heterotetramerizationGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
negative regulation of dendritic spine maintenanceGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
positive regulation of cysteine-type endopeptidase activityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
response to woundingGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
directional locomotionGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
negative regulation of protein catabolic processGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
neuromuscular process controlling balanceGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
protein localization to postsynaptic membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
calcium ion transportGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
dendrite developmentGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
rhythmic processGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
prepulse inhibitionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
negative regulation of dendritic spine developmentGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
calcium ion transmembrane transportGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
modulation of chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
lipid transportSigma non-opioid intracellular receptor 1Homo sapiens (human)
nervous system developmentSigma non-opioid intracellular receptor 1Homo sapiens (human)
G protein-coupled opioid receptor signaling pathwaySigma non-opioid intracellular receptor 1Homo sapiens (human)
regulation of neuron apoptotic processSigma non-opioid intracellular receptor 1Homo sapiens (human)
protein homotrimerizationSigma non-opioid intracellular receptor 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (72)

Processvia Protein(s)Taxonomy
acetylcholine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
neurotransmitter transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
dopamine:sodium symporter activitySolute carrier family 22 member 1 Homo sapiens (human)
norepinephrine:sodium symporter activitySolute carrier family 22 member 1 Homo sapiens (human)
protein bindingSolute carrier family 22 member 1 Homo sapiens (human)
monoamine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
secondary active organic cation transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
organic anion transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
organic cation transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
prostaglandin transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
pyrimidine nucleoside transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
thiamine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
putrescine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
spermidine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
quaternary ammonium group transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
toxin transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
identical protein bindingSolute carrier family 22 member 1 Homo sapiens (human)
xenobiotic transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
(R)-carnitine transmembrane transporter activitySolute carrier family 22 member 1 Homo sapiens (human)
glutamate-gated receptor activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
glutamate bindingGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
voltage-gated monoatomic cation channel activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
calcium channel activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
monoatomic cation channel activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
neurotransmitter receptor activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
glutamate receptor activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
monooxygenase activityCytochrome P450 2D6Homo sapiens (human)
iron ion bindingCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activityCytochrome P450 2D6Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2D6Homo sapiens (human)
heme bindingCytochrome P450 2D6Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 2D6Homo sapiens (human)
monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
iron ion bindingCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 14,15-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
arachidonic acid 11,12-epoxygenase activityCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
(S)-limonene 7-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
caffeine oxidase activityCytochrome P450 2C9 Homo sapiens (human)
(R)-limonene 6-monooxygenase activityCytochrome P450 2C9 Homo sapiens (human)
aromatase activityCytochrome P450 2C9 Homo sapiens (human)
heme bindingCytochrome P450 2C9 Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2C9 Homo sapiens (human)
integrin bindingSodium-dependent serotonin transporterHomo sapiens (human)
monoatomic cation channel activitySodium-dependent serotonin transporterHomo sapiens (human)
neurotransmitter transmembrane transporter activitySodium-dependent serotonin transporterHomo sapiens (human)
serotonin:sodium:chloride symporter activitySodium-dependent serotonin transporterHomo sapiens (human)
protein bindingSodium-dependent serotonin transporterHomo sapiens (human)
monoamine transmembrane transporter activitySodium-dependent serotonin transporterHomo sapiens (human)
antiporter activitySodium-dependent serotonin transporterHomo sapiens (human)
syntaxin-1 bindingSodium-dependent serotonin transporterHomo sapiens (human)
cocaine bindingSodium-dependent serotonin transporterHomo sapiens (human)
sodium ion bindingSodium-dependent serotonin transporterHomo sapiens (human)
identical protein bindingSodium-dependent serotonin transporterHomo sapiens (human)
nitric-oxide synthase bindingSodium-dependent serotonin transporterHomo sapiens (human)
actin filament bindingSodium-dependent serotonin transporterHomo sapiens (human)
serotonin bindingSodium-dependent serotonin transporterHomo sapiens (human)
monooxygenase activityCytochrome P450 2J2Homo sapiens (human)
iron ion bindingCytochrome P450 2J2Homo sapiens (human)
arachidonic acid epoxygenase activityCytochrome P450 2J2Homo sapiens (human)
arachidonic acid 14,15-epoxygenase activityCytochrome P450 2J2Homo sapiens (human)
arachidonic acid 11,12-epoxygenase activityCytochrome P450 2J2Homo sapiens (human)
isomerase activityCytochrome P450 2J2Homo sapiens (human)
linoleic acid epoxygenase activityCytochrome P450 2J2Homo sapiens (human)
hydroperoxy icosatetraenoate isomerase activityCytochrome P450 2J2Homo sapiens (human)
arachidonic acid 5,6-epoxygenase activityCytochrome P450 2J2Homo sapiens (human)
heme bindingCytochrome P450 2J2Homo sapiens (human)
oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen, reduced flavin or flavoprotein as one donor, and incorporation of one atom of oxygenCytochrome P450 2J2Homo sapiens (human)
G protein-coupled receptor activityTaste receptor type 2 member 46Homo sapiens (human)
bitter taste receptor activityTaste receptor type 2 member 46Homo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium channel activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
amyloid-beta bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium ion bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calmodulin bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
glutamate bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
protein-containing complex bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
signaling receptor activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
ligand-gated monoatomic ion channel activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
amyloid-beta bindingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
zinc ion bindingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
amyloid-beta bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
zinc ion bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glutamate bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
calcium channel activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
identical protein bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
protein phosphatase 2A bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
glutamate receptor activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
G protein-coupled opioid receptor activitySigma non-opioid intracellular receptor 1Homo sapiens (human)
protein bindingSigma non-opioid intracellular receptor 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (50)

Processvia Protein(s)Taxonomy
plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
basal plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
membraneSolute carrier family 22 member 1 Homo sapiens (human)
basolateral plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
apical plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
lateral plasma membraneSolute carrier family 22 member 1 Homo sapiens (human)
presynapseSolute carrier family 22 member 1 Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
presynaptic active zone membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
hippocampal mossy fiber to CA3 synapseGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
neuronal cell bodyGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
mitochondrionCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulumCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2D6Homo sapiens (human)
cytoplasmCytochrome P450 2D6Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2D6Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 2C9 Homo sapiens (human)
plasma membraneCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
cytoplasmCytochrome P450 2C9 Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2C9 Homo sapiens (human)
plasma membraneSodium-dependent serotonin transporterHomo sapiens (human)
focal adhesionSodium-dependent serotonin transporterHomo sapiens (human)
endosome membraneSodium-dependent serotonin transporterHomo sapiens (human)
endomembrane systemSodium-dependent serotonin transporterHomo sapiens (human)
presynaptic membraneSodium-dependent serotonin transporterHomo sapiens (human)
membrane raftSodium-dependent serotonin transporterHomo sapiens (human)
synapseSodium-dependent serotonin transporterHomo sapiens (human)
postsynaptic membraneSodium-dependent serotonin transporterHomo sapiens (human)
serotonergic synapseSodium-dependent serotonin transporterHomo sapiens (human)
synapseSodium-dependent serotonin transporterHomo sapiens (human)
plasma membraneSodium-dependent serotonin transporterHomo sapiens (human)
neuron projectionSodium-dependent serotonin transporterHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)
endoplasmic reticulum membraneCytochrome P450 2J2Homo sapiens (human)
extracellular exosomeCytochrome P450 2J2Homo sapiens (human)
cytoplasmCytochrome P450 2J2Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 2J2Homo sapiens (human)
plasma membraneTaste receptor type 2 member 46Homo sapiens (human)
membraneTaste receptor type 2 member 46Homo sapiens (human)
ciliary membraneTaste receptor type 2 member 46Homo sapiens (human)
membraneTaste receptor type 2 member 46Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)
cytoplasmGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synaptic vesicleGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
cell surfaceGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
postsynaptic densityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
dendriteGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synaptic cleftGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
terminal boutonGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
dendritic spineGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synapseGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
excitatory synapseGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synaptic membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synapseGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
synaptic vesicleGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
cell surfaceGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
postsynaptic densityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
cytoplasmic vesicle membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
presynaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
dendritic spineGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
synaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
cytoplasmGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
lysosomeGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
late endosomeGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
cytoskeletonGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
cell surfaceGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
postsynaptic densityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
synaptic membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
neuronal cell bodyGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
synapseGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
presynapseGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)
nuclear envelopeSigma non-opioid intracellular receptor 1Homo sapiens (human)
nuclear inner membraneSigma non-opioid intracellular receptor 1Homo sapiens (human)
nuclear outer membraneSigma non-opioid intracellular receptor 1Homo sapiens (human)
endoplasmic reticulumSigma non-opioid intracellular receptor 1Homo sapiens (human)
endoplasmic reticulum membraneSigma non-opioid intracellular receptor 1Homo sapiens (human)
lipid dropletSigma non-opioid intracellular receptor 1Homo sapiens (human)
cytosolSigma non-opioid intracellular receptor 1Homo sapiens (human)
postsynaptic densitySigma non-opioid intracellular receptor 1Homo sapiens (human)
membraneSigma non-opioid intracellular receptor 1Homo sapiens (human)
growth coneSigma non-opioid intracellular receptor 1Homo sapiens (human)
cytoplasmic vesicleSigma non-opioid intracellular receptor 1Homo sapiens (human)
anchoring junctionSigma non-opioid intracellular receptor 1Homo sapiens (human)
postsynaptic density membraneSigma non-opioid intracellular receptor 1Homo sapiens (human)
endoplasmic reticulumSigma non-opioid intracellular receptor 1Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (151)

Assay IDTitleYearJournalArticle
AID1210360Drug metabolism in human liver microsomes assessed as dextrorphan formation measured as microsomal fraction unbound2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
Prediction of relative in vivo metabolite exposure from in vitro data using two model drugs: dextromethorphan and omeprazole.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1220887Inhibition of catalytic activity of recombinant human CYP2D6 expressed in Escherichia coli co-expressing P450 reductase assessed as hydroxybufuralol activity at 16 uM after <15 mins by HPLC analysis in presence of NADPH2011Drug metabolism and disposition: the biological fate of chemicals, Jun, Volume: 39, Issue:6
Substituted imidazole of 5-fluoro-2-[4-[(2-phenyl-1H-imidazol-5-yl)methyl]-1-piperazinyl]pyrimidine Inactivates cytochrome P450 2D6 by protein adduction.
AID1245598Half life in human microsomes at 1 uM in presence of NADPH2015Bioorganic & medicinal chemistry, Oct-01, Volume: 23, Issue:19
Discovery of potent nitrotriazole-based antitrypanosomal agents: In vitro and in vivo evaluation.
AID54565Inhibition of MAMC O-dealkylation mediated by rat Cytochrome P450 2D3 expressed in Saccharomyces cerevisiae2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
Homology modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational rationalization of experimental ligand-binding specificities.
AID1702155n-Octanol/water distribution coefficient, logD of the compound at pH 7.42018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1773897Half life in mouse liver microsomes preincubated for 5 mins followed by NADPH addition and measured after 5 to 60 mins by LC-MS/MS analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Discovery of Novel Pterostilbene-Based Derivatives as Potent and Orally Active NLRP3 Inflammasome Inhibitors with Inflammatory Activity for Colitis.
AID1884504Inhibition of CYP2D6 in human liver microsomes incubated for 15 to 40 mins in presence of NADPH2022European journal of medicinal chemistry, Aug-05, Volume: 238Expansion of the S-CN-DABO scaffold to exploit the impact on inhibitory activities against the non-nucleoside HIV-1 reverse transcriptase.
AID1756359Metabolic stability in human hepatocytes assessed as half life measured upto 120 mins by LC-MS/MS analysis2021European journal of medicinal chemistry, Mar-05, Volume: 213Discovery and development of novel pyrimidine and pyrazolo/thieno-fused pyrimidine derivatives as potent and orally active inducible nitric oxide synthase dimerization inhibitor with efficacy for arthritis.
AID1611376Half life in human liver microsomes at 1 uM in presence of NADPH generating system measured for 10 to 90 mins by LC-MS/MS analysis
AID1862576Inhibition of CYP2D6 (unknown origin)
AID194779Percent inhibition of veratridine-induced glutamate release in rat brain slices at 10000 nmol/L2002Journal of medicinal chemistry, Aug-15, Volume: 45, Issue:17
Synthesis and structure-activity relationships of 6,7-benzomorphan derivatives as use-dependent sodium channel blockers for the treatment of stroke.
AID1247111Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of 3 to 50 mM Na+ to IC50 for NMDA receptor in Wistar rat brain membranes in absence of 3 to 50 mM Na+2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID1313189Half life in human liver microsomes at 3 uM in presence of NADPH by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 08-01, Volume: 26, Issue:15
Rational design and synthesis of novel anti-prostate cancer agents bearing a 3,5-bis-trifluoromethylphenyl moiety.
AID1635245Half life in human liver microsomes at 3 uM preincubated with compound followed by NADPH addition by LC-MS/MS analysis2016European journal of medicinal chemistry, Aug-08, Volume: 118Design and synthesis of novel bicalutamide and enzalutamide derivatives as antiproliferative agents for the treatment of prostate cancer.
AID1756362Metabolic stability in rat hepatocytes assessed as half life measured upto 120 mins by LC-MS/MS analysis2021European journal of medicinal chemistry, Mar-05, Volume: 213Discovery and development of novel pyrimidine and pyrazolo/thieno-fused pyrimidine derivatives as potent and orally active inducible nitric oxide synthase dimerization inhibitor with efficacy for arthritis.
AID1209250Drug metabolism in human liver microsomes by multiple depletion curves method2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID1526773Ratio of unbound maximal portal vein concentration in human at 60 mg, po to inhibition of human OCT1 expressed in HEK293 cells assessed as reduction in ASP+ substrate uptake by microplate reader based analysis2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1526752Passive membrane permeability by LC-MS/MS analysis based PAMPA2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID179824Tested for the anticonvulsant activity administered 60 min orally before MES induction1999Bioorganic & medicinal chemistry letters, Sep-06, Volume: 9, Issue:17
Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants.
AID1210362Drug metabolism in human liver microsomes assessed as dextrorphan formation measured as intrinsic unbound clearance2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
Prediction of relative in vivo metabolite exposure from in vitro data using two model drugs: dextromethorphan and omeprazole.
AID1209247Drug metabolism in human liver microsomes assessed as intrinsic clearance per mg protein by multiple depletion curves method2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID536851Half life in human liver microsomes at 3 uM by LC-MS/MS analysis2010Journal of medicinal chemistry, Nov-25, Volume: 53, Issue:22
Bicyclic substituted hydroxyphenylmethanones as novel inhibitors of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) for the treatment of estrogen-dependent diseases.
AID1773898Intrinsic clearance in mouse liver microsomes preincubated for 5 mins followed by NADPH addition and measured after 5 to 60 mins by LC-MS/MS analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Discovery of Novel Pterostilbene-Based Derivatives as Potent and Orally Active NLRP3 Inflammasome Inhibitors with Inflammatory Activity for Colitis.
AID681153TP_TRANSPORTER: inhibition of Daunorubicin efflux in NIH-3T3-G185 cells2001Chemical research in toxicology, Dec, Volume: 14, Issue:12
Quantitative distinctions of active site molecular recognition by P-glycoprotein and cytochrome P450 3A4.
AID203818Binding affinity was measured as selectivity for sigma-1 site over muscarinic (M1) receptor in rat1994Journal of medicinal chemistry, Jun-24, Volume: 37, Issue:13
Novel (4-phenylpiperidinyl)- and (4-phenylpiperazinyl)alkyl-spaced esters of 1-phenylcyclopentanecarboxylic acids as potent sigma-selective compounds.
AID1174935Behavioral effect in Wistar rat assessed as inhibition of nicotine-induced conditioned place preference at 10 mg/kg, ip administered during post-conditioning test phase on day 5 of nicotine treatment2014Bioorganic & medicinal chemistry, Dec-15, Volume: 22, Issue:24
Synthesis, in vitro and in vivo studies, and molecular modeling of N-alkylated dextromethorphan derivatives as non-competitive inhibitors of α3β4 nicotinic acetylcholine receptor.
AID179553Inhibitory effect against veratridine-induced glutamate release from rat brain slices2002Journal of medicinal chemistry, Aug-15, Volume: 45, Issue:17
Synthesis and structure-activity relationships of 6,7-benzomorphan derivatives as use-dependent sodium channel blockers for the treatment of stroke.
AID1702152Metabolic stability in human liver pooled microsomes assessed as half-life at 5 uM incubated upto 60 mins in presence of beta-NADPH by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1247112Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of 50 mM K+ to IC50 for NMDA receptor in Wistar rat brain membranes in absence of 50 mM K+2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID1254927Half life in human liver S9 microsomes in presence of NADPH by LC-MS/MS method2015European journal of medicinal chemistry, Oct-20, Volume: 1033-Nitrotriazole-based piperazides as potent antitrypanosomal agents.
AID190134Toxic dose causing 50% treated mice fall from the rotarod was evaluated; not calculated1999Bioorganic & medicinal chemistry letters, Sep-06, Volume: 9, Issue:17
Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants.
AID1409611AUC (cytotoxicity %) of compound against Vero E6 cells by MTT assay.2020Nature, 07, Volume: 583, Issue:7816
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
AID1254933Half life in human liver S9 microsomes in absence of NADPH by LC-MS/MS method2015European journal of medicinal chemistry, Oct-20, Volume: 1033-Nitrotriazole-based piperazides as potent antitrypanosomal agents.
AID1526731Substrate activity at human OCT1 expressed in HEK293 cells assessed as increase in compound uptake at 0.05 uM incubated for 2 mins by LC-MS/MS analysis relative to control empty vector transfected cells2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1496417Apparent half life in human liver microsomes at 3 uM2018Bioorganic & medicinal chemistry, 07-15, Volume: 26, Issue:11
BET bromodomain ligands: Probing the WPF shelf to improve BRD4 bromodomain affinity and metabolic stability.
AID74203Antitussive activity was evaluated by inhibition of irritant electrically-induced coughing by (po) administration in guinea pig1995Journal of medicinal chemistry, Feb-03, Volume: 38, Issue:3
N-acyl-2-substituted-1,3-thiazolidines, a new class of non-narcotic antitussive agents: studies leading to the discovery of ethyl 2-[(2-methoxyphenoxy)methyl]-beta-oxothiazolidine-3-propanoate.
AID1526732Substrate activity at human OCT1 expressed in HEK293 cells assessed as increase in compound uptake at 0.1 uM incubated for 2 mins by LC-MS/MS analysis relative to control empty vector transfected cells2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1129361Unbound fraction in HEK293 cell homogenate at 0.1 uM by equilibrium dialysis based UPLC-MS/MS analysis2014Journal of medicinal chemistry, Apr-10, Volume: 57, Issue:7
A high-throughput cell-based method to predict the unbound drug fraction in the brain.
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID205276Inhibition of [3H]saxitoxin binding to rat brain sodium channel1999Bioorganic & medicinal chemistry letters, Sep-06, Volume: 9, Issue:17
Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants.
AID353103Displacement of [3H]MK801 from NMDA receptor in rat brain neuronal membrane2009Bioorganic & medicinal chemistry, May-01, Volume: 17, Issue:9
NMDA receptor affinities of 1,2-diphenylethylamine and 1-(1,2-diphenylethyl)piperidine enantiomers and of related compounds.
AID1247110Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of H+ at pH 6.4 to 8.2 to IC50 for NMDA receptor in Wistar rat brain membranes in absence of H+ at pH 6.4 to 8.22015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID1702159Clearance in mouse liver microsomes at pH 7.4 at 5 uM incubated upto 60 mins in presence of NADPH measured per mg of protein by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1365724Intrinsic clearance in human liver S9 microsomes at 1 uM in presence of NADPH by LC-MS/MS analysis2017Bioorganic & medicinal chemistry, 11-01, Volume: 25, Issue:21
The antitubercular activity of various nitro(triazole/imidazole)-based compounds.
AID1702153Metabolic stability in mouse liver microsomes assessed as half-life at 5 uM incubated upto 60 mins in presence of beta-NADPH by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID205275Inhibition of [3H]batrachotoxin binding to rat brain sodium channel1999Bioorganic & medicinal chemistry letters, Sep-06, Volume: 9, Issue:17
Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants.
AID1409608AUC (viral infection %) for SARS-CoV-2 in the Vero E6 cell line at 48 h by immunofluorescence-based assay (detecting the viral NP protein in the nucleus of the Vero E6 cells).2020Nature, 07, Volume: 583, Issue:7816
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
AID1365725Intrinsic clearance in human liver S9 microsomes at 1 uM in absence of NADPH by LC-MS/MS analysis2017Bioorganic & medicinal chemistry, 11-01, Volume: 25, Issue:21
The antitubercular activity of various nitro(triazole/imidazole)-based compounds.
AID1744242Displacement of [3H]-(+)-pentazocine from sigma-1 receptor in guinea pig brain membranes incubated for 1 hr in presence of 250 uM phenytoin liquid scintillation counting method2021Journal of medicinal chemistry, 01-14, Volume: 64, Issue:1
Novel Sigma 1 Receptor Antagonists as Potential Therapeutics for Pain Management.
AID1652314Inhibition of human CYP2D6 at 10 uM2020Journal of medicinal chemistry, 07-09, Volume: 63, Issue:13
Novel Pyrrolopyridone Bromodomain and Extra-Terminal Motif (BET) Inhibitors Effective in Endocrine-Resistant ER+ Breast Cancer with Acquired Resistance to Fulvestrant and Palbociclib.
AID54570Inhibition of MAMC O-dealkylation mediated by human Cytochrome P450 2D6 expressed in human lymphoblastoid cell line2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
Homology modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational rationalization of experimental ligand-binding specificities.
AID1759550Metabolic stability in mouse liver microsomes assessed as half life2021Bioorganic & medicinal chemistry letters, 06-01, Volume: 41Inhibitors of heat shock protein 70 (Hsp70) with enhanced metabolic stability reduce tau levels.
AID1210361Drug metabolism in human liver microsomes assessed as dextrorphan formation measured as intrinsic clearance2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
Prediction of relative in vivo metabolite exposure from in vitro data using two model drugs: dextromethorphan and omeprazole.
AID1773894Half life in human liver microsomes preincubated for 5 mins followed by NADPH addition and measured after 5 to 60 mins by LC-MS/MS analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Discovery of Novel Pterostilbene-Based Derivatives as Potent and Orally Active NLRP3 Inflammasome Inhibitors with Inflammatory Activity for Colitis.
AID1174942Effect on basal locomotor activity in Wistar rat at 5 mg/kg, ip2014Bioorganic & medicinal chemistry, Dec-15, Volume: 22, Issue:24
Synthesis, in vitro and in vivo studies, and molecular modeling of N-alkylated dextromethorphan derivatives as non-competitive inhibitors of α3β4 nicotinic acetylcholine receptor.
AID1247114Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of 1.3 mM Mg2+ to IC50 for NMDA receptor in Wistar rat brain membranes in absence of 1.3 mM Mg2+2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID699540Inhibition of human liver OATP1B3 expressed in HEK293 Flp-In cells assessed as reduction in [3H]E17-betaG uptake at 20 uM incubated for 5 mins by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID1526759Cmax in human at 60 mg, po2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID311933Inhibition of ASM in rat PC12 cells assessed as residual activity at 10 uM2008Journal of medicinal chemistry, Jan-24, Volume: 51, Issue:2
Identification of new functional inhibitors of acid sphingomyelinase using a structure-property-activity relation model.
AID1212341Cytotoxicity against human Fa2N-4 cells by lactate dehydrogenase assay2013Drug metabolism and disposition: the biological fate of chemicals, Apr, Volume: 41, Issue:4
Lysosomal sequestration (trapping) of lipophilic amine (cationic amphiphilic) drugs in immortalized human hepatocytes (Fa2N-4 cells).
AID317399Displacement of [3H]pentazocine from sigma1 receptor in guinea pig membrane2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Synthesis and structure-activity relationships of N-(3-phenylpropyl)-N'-benzylpiperazines: Potent ligands for sigma1 and sigma2 receptors.
AID317400Displacement of [3H]pentazocine from sigma1 receptor in guinea pig membrane in presence of phenytoin2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Synthesis and structure-activity relationships of N-(3-phenylpropyl)-N'-benzylpiperazines: Potent ligands for sigma1 and sigma2 receptors.
AID1702160Clearance in rat liver microsomes at pH 7.4 at 5 uM incubated upto 60 mins in presence of NADPH measured per mg of protein by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1174939Behavioral effect in Wistar rat assessed as inhibition of expression of nicotine-induced locomotor sensitization at 5 to 10 mg/kg, ip administered on day 16 of pairing phase followed by nicotine treatment measured for 60 mins after compound treatment2014Bioorganic & medicinal chemistry, Dec-15, Volume: 22, Issue:24
Synthesis, in vitro and in vivo studies, and molecular modeling of N-alkylated dextromethorphan derivatives as non-competitive inhibitors of α3β4 nicotinic acetylcholine receptor.
AID54564Inhibition of MAMC O-dealkylation mediated by rat Cytochrome P450 2D2 expressed in Saccharomyces cerevisiae2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
Homology modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational rationalization of experimental ligand-binding specificities.
AID1426330Half life in human liver microsomes at 1.5 uM in presence of NADPH by liquid chromatography/tandem mass spectrometric analysis2017Journal of medicinal chemistry, 02-09, Volume: 60, Issue:3
Urea Derivatives of 2-Aryl-benzothiazol-5-amines: A New Class of Potential Drugs for Human African Trypanosomiasis.
AID1526771Unbound maximal portal vein concentration of in human at 60 mg, po2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID311367Permeability coefficient in human skin2007Bioorganic & medicinal chemistry, Nov-15, Volume: 15, Issue:22
Transdermal penetration behaviour of drugs: CART-clustering, QSPR and selection of model compounds.
AID1247113Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of 20 mM NH4+ to IC50 for NMDA receptor in Wistar rat brain membranes in absence of 20 mM NH4+2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID1526760Unbound Cmax in human at 60 mg, po2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1526751Inhibition of human OCT1 expressed in HEK293 cells assessed as reduction in ASP+ substrate uptake by microplate reader based analysis2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1247109Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of 10 to 50 mM Tris to IC50 for NMDA receptor in Wistar rat brain membranes in absence of 10 to 50 mM Tris2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID404304Effect on human MRP2-mediated estradiol-17-beta-glucuronide transport in Sf9 cells inverted membrane vesicles relative to control2008Journal of medicinal chemistry, Jun-12, Volume: 51, Issue:11
Prediction and identification of drug interactions with the human ATP-binding cassette transporter multidrug-resistance associated protein 2 (MRP2; ABCC2).
AID1313195Intrinsic clearance in human liver microsomes assessed per mg of protein at 3 uM in presence of NADPH by LC-MS/MS analysis2016Bioorganic & medicinal chemistry letters, 08-01, Volume: 26, Issue:15
Rational design and synthesis of novel anti-prostate cancer agents bearing a 3,5-bis-trifluoromethylphenyl moiety.
AID536762Intrinsic clearance in human liver microsomes at 3 uM measured per mg of protein by LC-MS/MS analysis2010Journal of medicinal chemistry, Nov-25, Volume: 53, Issue:22
Bicyclic substituted hydroxyphenylmethanones as novel inhibitors of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) for the treatment of estrogen-dependent diseases.
AID1209248Drug metabolism in human liver microsomes assessed as intrinsic clearance per mg protein by optimal design approach2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID54566Inhibition of MAMC O-dealkylation mediated by rat Cytochrome P450 2D4 expressed in Saccharomyces cerevisiae2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
Homology modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational rationalization of experimental ligand-binding specificities.
AID1212315Drug uptake in lysosomes of human Fa2N-4 cells assessed as inhibition of LysoTracker Red fluorescence at 500 uM after 30 mins relative to control2013Drug metabolism and disposition: the biological fate of chemicals, Apr, Volume: 41, Issue:4
Lysosomal sequestration (trapping) of lipophilic amine (cationic amphiphilic) drugs in immortalized human hepatocytes (Fa2N-4 cells).
AID1610344Clearance in human liver microsomes at 1 uM in presence of NADPH generating system measured for 10 to 90 mins by LC-MS/MS analysis
AID1454611Half life in mouse liver microsomes by LC-MS/MS method2017ACS medicinal chemistry letters, Jul-13, Volume: 8, Issue:7
New Class of Antitrypanosomal Agents Based on Imidazopyridines.
AID1744243Ratio of Ki for displacement of [3H]-(+)-pentazocine from sigma-1 receptor to Ki for displacement of [3H]-(+)-pentazocine from sigma-1 receptor in guinea pig brain membranes in presence of phenytoin2021Journal of medicinal chemistry, 01-14, Volume: 64, Issue:1
Novel Sigma 1 Receptor Antagonists as Potential Therapeutics for Pain Management.
AID1365718Half life in human liver S9 microsomes at 1 uM in absence of NADPH by LC-MS/MS analysis2017Bioorganic & medicinal chemistry, 11-01, Volume: 25, Issue:21
The antitubercular activity of various nitro(triazole/imidazole)-based compounds.
AID1222793Dissociation constant, pKa of the compound2013Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 41, Issue:5
Which metabolites circulate?
AID1409613Selectivity ratio: ratio of AUC (viral infection %) of SARS-CoV-2 in the Vero E6 cell line compared to AUC (cytotoxicity %) of compound against Vero E6 cells by MTT assay.2020Nature, 07, Volume: 583, Issue:7816
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
AID1773895Intrinsic clearance in human liver microsomes preincubated for 5 mins followed by NADPH addition and measured after 5 to 60 mins by LC-MS/MS analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Discovery of Novel Pterostilbene-Based Derivatives as Potent and Orally Active NLRP3 Inflammasome Inhibitors with Inflammatory Activity for Colitis.
AID1756360Metabolic stability in human hepatocytes assessed as clearance incubated up to 120 mins measured per million cells by LC-MS/MS analysis2021European journal of medicinal chemistry, Mar-05, Volume: 213Discovery and development of novel pyrimidine and pyrazolo/thieno-fused pyrimidine derivatives as potent and orally active inducible nitric oxide synthase dimerization inhibitor with efficacy for arthritis.
AID1548950Unbound hepatic clearance in rat liver microsomes2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Discovery of
AID1215672Drug metabolism in pooled human hepatocytes assessed as aldehyde oxidase-mediated drug metabolism at 10 uM up to 120 mins by HPLC analysis in presence of 50 uM of hydralazine2012Drug metabolism and disposition: the biological fate of chemicals, Jul, Volume: 40, Issue:7
Hydralazine as a selective probe inactivator of aldehyde oxidase in human hepatocytes: estimation of the contribution of aldehyde oxidase to metabolic clearance.
AID1174934Non-competitive inhibition of rat alpha3beta4 nAChR expressed in KX cells assessed as reduction in current by whole-cell patch clamp assay2014Bioorganic & medicinal chemistry, Dec-15, Volume: 22, Issue:24
Synthesis, in vitro and in vivo studies, and molecular modeling of N-alkylated dextromethorphan derivatives as non-competitive inhibitors of α3β4 nicotinic acetylcholine receptor.
AID157456Binding affinity was measured against phencyclidine (PCP) receptor in rat using [3H]TCP as radioligand1994Journal of medicinal chemistry, Jun-24, Volume: 37, Issue:13
Novel (4-phenylpiperidinyl)- and (4-phenylpiperazinyl)alkyl-spaced esters of 1-phenylcyclopentanecarboxylic acids as potent sigma-selective compounds.
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1442001Inhibition of human OCT1 expressed in HEK293 cells assessed as decrease in uptake of ASP+ after 2 mins by fluorescence assay2017Journal of medicinal chemistry, 04-13, Volume: 60, Issue:7
Discovery of Competitive and Noncompetitive Ligands of the Organic Cation Transporter 1 (OCT1; SLC22A1).
AID1409614Overall antiviral activity against SARS-CoV-2 (isolate France/IDF0372/2020) in the Vero E6 cell line at 48 h based on three assays 1) detection of viral RNA by qRT-PCR (targeting the N-gene), 2) plaque assay using lysate 3 days after addition of compound 2020Nature, 07, Volume: 583, Issue:7816
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
AID311934Dissociation constant, pKa of the compound2008Journal of medicinal chemistry, Jan-24, Volume: 51, Issue:2
Identification of new functional inhibitors of acid sphingomyelinase using a structure-property-activity relation model.
AID1426327Half life in mouse liver microsomes at 1.5 uM in presence of NADPH by liquid chromatography/tandem mass spectrometric analysis2017Journal of medicinal chemistry, 02-09, Volume: 60, Issue:3
Urea Derivatives of 2-Aryl-benzothiazol-5-amines: A New Class of Potential Drugs for Human African Trypanosomiasis.
AID1496418Intrinsic clearance in human liver microsomes at 3 uM2018Bioorganic & medicinal chemistry, 07-15, Volume: 26, Issue:11
BET bromodomain ligands: Probing the WPF shelf to improve BRD4 bromodomain affinity and metabolic stability.
AID1409609Cytotoxicity of compound against Vero E6 cells by MTT assay.2020Nature, 07, Volume: 583, Issue:7816
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
AID1465450Unbound intrinsic clearance in human liver microsomes at 10 uM preincubated for 5 mins followed by NADPH addition measured after 60 mins by HPLC-UV-MS analysis2017Journal of medicinal chemistry, 10-26, Volume: 60, Issue:20
Discovery of N-(4-(2,4-Difluorophenoxy)-3-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)ethanesulfonamide (ABBV-075/Mivebresib), a Potent and Orally Available Bromodomain and Extraterminal Domain (BET) Family Bromodomain Inhibitor.
AID699539Inhibition of human liver OATP1B1 expressed in HEK293 Flp-In cells assessed as reduction in E17-betaG uptake at 20 uM by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID1245597Half life in human microsomes at 1 uM in absence of NADPH2015Bioorganic & medicinal chemistry, Oct-01, Volume: 23, Issue:19
Discovery of potent nitrotriazole-based antitrypanosomal agents: In vitro and in vivo evaluation.
AID1210359Drug metabolism in human liver microsomes assessed as dextrorphan formation2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
Prediction of relative in vivo metabolite exposure from in vitro data using two model drugs: dextromethorphan and omeprazole.
AID1526733Substrate activity at human OCT1 expressed in HEK293 cells assessed as increase in compound uptake at 0.5 uM incubated for 2 mins by LC-MS/MS analysis relative to control empty vector transfected cells2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1682815Metabolic stability in human liver microsomes assessed as half life
AID1409607IC50 for antiviral activity against SARS-CoV-2 in the Vero E6 cell line at 48 h by immunofluorescence-based assay (detecting the viral NP protein in the nucleus of the Vero E6 cells).2020Nature, 07, Volume: 583, Issue:7816
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing.
AID1210069Inhibition of human recombinant CYP2J2 assessed as reduction in astemizole O-demethylation by LC-MS/MS method2013Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 41, Issue:1
Discovery and characterization of novel, potent, and selective cytochrome P450 2J2 inhibitors.
AID1610343Half life in human liver microsomes at 1 uM in presence of NADPH generating system measured for 10 to 90 mins by LC-MS/MS analysis
AID1247108Ratio of IC50 for NMDA receptor in Wistar rat brain membranes in presence of 30 uM spermine to IC50 for NMDA receptor in Wistar rat brain membranes in absence of 30 uM spermine2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID1209258Drug metabolism in human liver microsomes assessed as maximum rate of reaction per mg protein by multiple depletion curves method2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID212532Lethal dose in rats1999Bioorganic & medicinal chemistry letters, Sep-06, Volume: 9, Issue:17
Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants.
AID262947Inhibition of human CYP2D6 expressed in Escherichia coli JM1092006Journal of medicinal chemistry, Apr-20, Volume: 49, Issue:8
Catalytic site prediction and virtual screening of cytochrome P450 2D6 substrates by consideration of water and rescoring in automated docking.
AID1756363Metabolic stability in rat hepatocytes assessed as intrinsic clearance incubated up to 120 mins measured per million cells by LC-MS/MS analysis2021European journal of medicinal chemistry, Mar-05, Volume: 213Discovery and development of novel pyrimidine and pyrazolo/thieno-fused pyrimidine derivatives as potent and orally active inducible nitric oxide synthase dimerization inhibitor with efficacy for arthritis.
AID226359Displacement of [3H]BTX from sodium channel of rat cerebral cortex synaptosomes2002Journal of medicinal chemistry, Aug-15, Volume: 45, Issue:17
Synthesis and structure-activity relationships of 6,7-benzomorphan derivatives as use-dependent sodium channel blockers for the treatment of stroke.
AID699541Inhibition of human liver OATP2B1 expressed in HEK293 Flp-In cells assessed as reduction in [3H]E3S uptake at 20 uM incubated for 5 mins by scintillation counting2012Journal of medicinal chemistry, May-24, Volume: 55, Issue:10
Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions.
AID204634Inhibition of [3H](+)-pentazocine binding to Sigma receptor type 12004Bioorganic & medicinal chemistry letters, Apr-05, Volume: 14, Issue:7
Synthesis of potent sigma-1 receptor ligands via fragmentation of dextromethorphan.
AID1247107Displacement of [3H]MK-801 from NMDA receptor in Wistar rat brain membranes by scintillation counting analysis2015Bioorganic & medicinal chemistry letters, Oct-01, Volume: 25, Issue:19
Differential influence of 7 cations on 16 non-competitive NMDA receptor blockers.
AID1526772Ratio of unbound Cmax in human at 60 mg, po to inhibition of human OCT1 expressed in HEK293 cells assessed as reduction in ASP+ substrate uptake by microplate reader based analysis2019Journal of medicinal chemistry, 11-14, Volume: 62, Issue:21
Opioids as Substrates and Inhibitors of the Genetically Highly Variable Organic Cation Transporter OCT1.
AID1209259Drug metabolism in human liver microsomes assessed as maximum rate of reaction per mg protein by optimal design approach2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID142870Binding affinity was measured against muscarinic (M2) receptor at 10 uM in rat using [3H]QN as radioligand1994Journal of medicinal chemistry, Jun-24, Volume: 37, Issue:13
Novel (4-phenylpiperidinyl)- and (4-phenylpiperazinyl)alkyl-spaced esters of 1-phenylcyclopentanecarboxylic acids as potent sigma-selective compounds.
AID1209251Drug metabolism in human liver microsomes by optimal design approach2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID54562Inhibition of MAMC O-dealkylation mediated by rat Cytochrome P450 2D1 expressed in Saccharomyces cerevisiae2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
Homology modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational rationalization of experimental ligand-binding specificities.
AID1611377Clearance in human liver microsomes at 1 uM in presence of NADPH generating system measured for 10 to 90 mins by LC-MS/MS analysis
AID1365717Half life in human liver S9 microsomes at 1 uM in presence of NADPH by LC-MS/MS analysis2017Bioorganic & medicinal chemistry, 11-01, Volume: 25, Issue:21
The antitubercular activity of various nitro(triazole/imidazole)-based compounds.
AID313147Reduction in pain intensity in diabetic neuropathy patient2007Journal of medicinal chemistry, May-31, Volume: 50, Issue:11
N-methyl-D-aspartate antagonists and neuropathic pain: the search for relief.
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1702154Metabolic stability in rat liver microsomes assessed as half-life at 5 uM incubated upto 60 mins in presence of beta-NADPH by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1210363Drug metabolism in human liver microsomes assessed as dextrorphan formation measured as plasma fraction unbound2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
Prediction of relative in vivo metabolite exposure from in vitro data using two model drugs: dextromethorphan and omeprazole.
AID1212314Drug uptake in lysosomes of human Fa2N-4 cells assessed as inhibition of LysoTracker Red fluorescence after 30 mins2013Drug metabolism and disposition: the biological fate of chemicals, Apr, Volume: 41, Issue:4
Lysosomal sequestration (trapping) of lipophilic amine (cationic amphiphilic) drugs in immortalized human hepatocytes (Fa2N-4 cells).
AID353104Ratio of Ki for rat brain NMDA receptor in presence of 100 uM spermine to Ki for rat brain NMDA receptor in absence of spermine2009Bioorganic & medicinal chemistry, May-01, Volume: 17, Issue:9
NMDA receptor affinities of 1,2-diphenylethylamine and 1-(1,2-diphenylethyl)piperidine enantiomers and of related compounds.
AID74201Antitussive activity was evaluated by inhibition of irritant aerosol-induced coughing by (po) administration in guinea pig1995Journal of medicinal chemistry, Feb-03, Volume: 38, Issue:3
N-acyl-2-substituted-1,3-thiazolidines, a new class of non-narcotic antitussive agents: studies leading to the discovery of ethyl 2-[(2-methoxyphenoxy)methyl]-beta-oxothiazolidine-3-propanoate.
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1210358Drug metabolism in human liver microsomes assessed as dextrorphan formation measured per mg of microsomal protein2012Drug metabolism and disposition: the biological fate of chemicals, Jan, Volume: 40, Issue:1
Prediction of relative in vivo metabolite exposure from in vitro data using two model drugs: dextromethorphan and omeprazole.
AID1702156Inhibition of human ERG by fluorescence polarization assay2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID19006Calculated membrane partition coefficient (Kmemb)2004Journal of medicinal chemistry, Mar-25, Volume: 47, Issue:7
Surface activity profiling of drugs applied to the prediction of blood-brain barrier permeability.
AID1209236Drug metabolism in assessed as human CYP2D6-mediated maximum rate of reaction per mg protein2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID1209262Drug metabolism in assessed as human CYP2D6-mediated intrinsic clearance per mg protein2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID1702158Clearance in human liver pooled microsomes at pH 7.4 at 5 uM incubated upto 60 mins in presence of NADPH measured per mg of protein by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1174937Toxicity in Wistar rat assessed as inhibition of conditioned place preference at 10 mg/kg, ip in absence of nicotine preconditioning2014Bioorganic & medicinal chemistry, Dec-15, Volume: 22, Issue:24
Synthesis, in vitro and in vivo studies, and molecular modeling of N-alkylated dextromethorphan derivatives as non-competitive inhibitors of α3β4 nicotinic acetylcholine receptor.
AID1209237Drug metabolism in assessed as human CYP2D6-mediated metabolism2012Drug metabolism and disposition: the biological fate of chemicals, Dec, Volume: 40, Issue:12
Optimized experimental design for the estimation of enzyme kinetic parameters: an experimental evaluation.
AID1442006Ratio of Kii for human OCT1-ASP+-inhibitor complex to Kis for human OCT1-inhibitor complex by Lineweaver-Burk plot analysis2017Journal of medicinal chemistry, 04-13, Volume: 60, Issue:7
Discovery of Competitive and Noncompetitive Ligands of the Organic Cation Transporter 1 (OCT1; SLC22A1).
AID156805The compound was tested for its ability to block PCP N-methyl-D-aspartate glutamate receptor at the PCP (phencyclidine) binding site in postmortem human frontal cortex.1998Journal of medicinal chemistry, Jan-29, Volume: 41, Issue:3
Quantitative analysis of the structural requirements for blockade of the N-methyl-D-aspartate receptor at the phencyclidine binding site.
AID1744232Displacement of [3H]-(+)-pentazocine from sigma-1 receptor in guinea pig brain membranes incubated for 1 hr by liquid scintillation counting method2021Journal of medicinal chemistry, 01-14, Volume: 64, Issue:1
Novel Sigma 1 Receptor Antagonists as Potential Therapeutics for Pain Management.
AID145105Binding affinity was measured against N-methyl-D-aspartate (NMDA) receptor in rat using [3H]CGS-19,755 as radioligand at 10 uM1994Journal of medicinal chemistry, Jun-24, Volume: 37, Issue:13
Novel (4-phenylpiperidinyl)- and (4-phenylpiperazinyl)alkyl-spaced esters of 1-phenylcyclopentanecarboxylic acids as potent sigma-selective compounds.
AID1773899Hepatic clearance in mouse liver microsomes preincubated for 5 mins followed by NADPH addition and measured after 5 to 60 mins by LC-MS/MS analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Discovery of Novel Pterostilbene-Based Derivatives as Potent and Orally Active NLRP3 Inflammasome Inhibitors with Inflammatory Activity for Colitis.
AID1773896Hepatic clearance in human liver microsomes preincubated for 5 mins followed by NADPH addition and measured after 5 to 60 mins by LC-MS/MS analysis2021Journal of medicinal chemistry, 09-23, Volume: 64, Issue:18
Discovery of Novel Pterostilbene-Based Derivatives as Potent and Orally Active NLRP3 Inflammasome Inhibitors with Inflammatory Activity for Colitis.
AID1635243Intrinsic clearance in human liver microsomes assessed per mg protein at 3 uM preincubated with compound followed by NADPH addition measured after 5 to 45 mins by LC-MS/MS analysis2016European journal of medicinal chemistry, Aug-08, Volume: 118Design and synthesis of novel bicalutamide and enzalutamide derivatives as antiproliferative agents for the treatment of prostate cancer.
AID1682813Metabolic stability in human liver microsomes assessed as intrinsic clearance
AID226516Protective index (PI) is the ratio of TD50 in the rotarod test over the ED50 in the anticonvulsant test;not calculated1999Bioorganic & medicinal chemistry letters, Sep-06, Volume: 9, Issue:17
Sodium channel activity and sigma binding of 2-aminopropanamide anticonvulsants.
AID1702161Drug metabolism assessed as human recombinant CYP3A4-mediated clearance at pH 7.4 at 5 uM incubated upto 60 mins in presence of NADPH by LC-MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID1159607Screen for inhibitors of RMI FANCM (MM2) intereaction2016Journal of biomolecular screening, Jul, Volume: 21, Issue:6
A High-Throughput Screening Strategy to Identify Protein-Protein Interaction Inhibitors That Block the Fanconi Anemia DNA Repair Pathway.
AID1346601Human GluN2C (Ionotropic glutamate receptors)2007The Journal of physiology, May-15, Volume: 581, Issue:Pt 1
Subunit-specific mechanisms and proton sensitivity of NMDA receptor channel block.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (1,908)

TimeframeStudies, This Drug (%)All Drugs %
pre-1990209 (10.95)18.7374
1990's468 (24.53)18.2507
2000's594 (31.13)29.6817
2010's498 (26.10)24.3611
2020's139 (7.29)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 94.02

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index94.02 (24.57)
Research Supply Index7.77 (2.92)
Research Growth Index4.77 (4.65)
Search Engine Demand Index236.74 (26.88)
Search Engine Supply Index2.73 (0.95)

This Compound (94.02)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials342 (16.93%)5.53%
Reviews122 (6.04%)6.00%
Case Studies179 (8.86%)4.05%
Observational3 (0.15%)0.25%
Other1,374 (68.02%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (136)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Randomized Drug Interaction Study of RO4929097 for Advanced Solid Tumors [NCT01218620]Phase 117 participants (Actual)Interventional2010-09-30Completed
Endobiotics for Phenotyping of Human Cytochrome P450 Enzymes: Use of Metabolomics for the Identification of New CYP2D6 Endogenous Biomarkers in Healthy Volunteers [NCT04188028]40 participants (Actual)Interventional2019-01-01Completed
Licorice Botanical Dietary Supplements - Metabolism and Safety in Women [NCT03948243]Phase 119 participants (Actual)Interventional2019-04-01Completed
The Potential Therapeutic Effects of add-on Low Dose Dextromethorphan and Memantine in Patients With Amphetamine-type Stimulants Use Disorder [NCT03729128]Phase 1/Phase 285 participants (Actual)Interventional2018-07-24Completed
Evaluation of the Effect of LY3871801 on the Pharmacokinetics of CYP450 Substrates and an OAT1/3 Substrate in Healthy Participants [NCT05602675]Phase 139 participants (Actual)Interventional2022-11-02Completed
A Phase 1, Open-label, Drug Interaction Study to Evaluate the Effect of Guselkumab (CNTO 1959) on Cytochrome P450 Enzyme Activities Following a Single Subcutaneous Administration in Subjects With Moderate to Severe Plaque-type Psoriasis [NCT02397382]Phase 116 participants (Actual)Interventional2015-06-18Completed
Effects of Multiple Doses of Abemaciclib on the Pharmacokinetics of Cytochrome P450 (CYP) 1A2, CYP2C9, CYP2D6, and CYP3A Substrates (Caffeine, Warfarin, Dextromethorphan, and Midazolam) in Cancer Patients [NCT02688088]Phase 148 participants (Actual)Interventional2016-03-08Completed
Psilocybin-facilitated Treatment for Chronic Pain [NCT05068791]Early Phase 130 participants (Anticipated)Interventional2023-11-01Recruiting
[NCT02391688]Phase 130 participants (Actual)Interventional2014-11-30Completed
Impact of Nuedexta on Bulbar Physiology and Function in ALS [NCT03883581]Phase 1/Phase 228 participants (Actual)Interventional2019-07-25Completed
An Open-Label Drug Interaction Study in Healthy Subjects to Evaluate the Effects of Multiple Doses of JNJ55308942 on the Cytochrome P450 CYP3A4, CYP2D6 and CYP2C19 Activity and on the Pharmacokinetics of Levonorgestrel/Ethinyl Estradiol [NCT03547024]Phase 114 participants (Actual)Interventional2018-06-08Completed
Fluoxetine/Dextromethorphan in Obsessive-Compulsive and Related Disorders: an Open-Label Crossover Pilot Study [NCT04899687]Phase 260 participants (Anticipated)Interventional2022-01-20Recruiting
An Open-Label Study to Assess the Long-term Safety and Efficacy of AXS-05 in Subjects With Major Depressive Disorder [NCT04039022]Phase 3876 participants (Actual)Interventional2019-07-08Completed
A Double-blind, Placebo-Controlled, Randomized Study of the Efficacy of Dextromethorphan as Add-On Therapy to Risperidone Versus Risperidone Alone in Patients With Schizophrenia [NCT01189006]Phase 2/Phase 3161 participants (Actual)Interventional2005-01-31Completed
The Reno-protective and Cardiovascular Effect of Dextromethorphan and Silymarin in Patients With Chronic Kidney Disease [NCT01091324]Phase 345 participants (Anticipated)Interventional2010-01-31Active, not recruiting
Dextromethorphan in Fibromyalgia [NCT03538054]Phase 227 participants (Actual)Interventional2018-06-26Completed
Effect of Acute Ethanol Consumption on The Activity of Major Cytochrome P450 Enzymes, NAT2 and P-glycoprotein [NCT02515526]16 participants (Actual)Interventional2015-06-30Completed
A Study to Evaluate the Effect of Multiple Doses of 500 mg of BIRT 2584 XX Tablets on the Pharmacokinetic Parameters of Warfarin, Omeprazole, Caffeine, and Dextromethorphan Dosed Orally and Midazolam Dosed IV, in Healthy Male Volunteers [NCT02256813]Phase 120 participants (Actual)Interventional2005-09-30Completed
Cocktail Phenotypic Approach to Explore Antidepressant Pharmacokinetic Variability: a Pilot Study [NCT02438072]100 participants (Anticipated)Interventional2014-12-31Recruiting
Evaluation of the Effect of Ixekizumab on the Pharmacokinetics of Cytochrome P450 Substrates in Patients With Moderate-to-Severe Plaque Psoriasis [NCT02993471]Phase 128 participants (Actual)Interventional2016-12-22Completed
Single Centre Open-label, Non-randomised, 3-treatment, 2-period, Pharmacokinetic Drug Interaction Study of Single Oral Dose of Acoziborole With Sequential Co-administration of Midazolam and Dextromethorphan in Healthy Male Participants [NCT05947604]Phase 120 participants (Actual)Interventional2023-02-09Completed
Trial of Naltrexone and Dextromethorphan for Gulf War Veterans Illnesses [NCT02206490]Phase 260 participants (Actual)Interventional2012-01-31Completed
A PLACEBO-CONTROLLED, DOUBLE-BLIND, RANDOMIZED, PARALLEL GROUP PILOT STUDY TO EVALUATE THE EFFICACY OF DEXTROMETHORPHAN HYDROBROMIDE ON ACUTE COUGH IN A PEDIATRIC POPULATION [NCT02651116]Phase 4131 participants (Actual)Interventional2016-02-25Terminated(stopped due to The study was prematurely discontinued due to slow enrollment during the 2019/2020 cold season. No safety or efficacy concerns led to the decision to terminate)
Effects of Type 2 Diabetes on CYP450s Activities; Intersubject Variability in Drug Metabolism. [NCT02291666]Phase 473 participants (Actual)Interventional2015-04-30Completed
A Phase I Multiple Dose Trial to Investigate Safety With Special Emphasis on ECG Effects and Tolerability After Oral Doses of 30 mg q.i.d. and 90 mg q.i.d. Dextromethorphan Hydrobromide Monohydrate (2 mg/mL Syrup) in Healthy Male and Female Subjects for 2 [NCT02191176]Phase 148 participants (Actual)Interventional2009-06-30Completed
Risk of Phenoconversion in Genetic Extensive Metabolizers Healthy Volunteers Carriers of One Fully-Functional and One Non-Functional Allele Versus Carriers of Two Fully-Functional Alleles [NCT03054220]34 participants (Actual)Interventional2016-07-31Completed
Clinical Trials on Evaluate the Red Ginseng and Fermented-Red Ginseng Affect to Drug Metabolizing Enzyme and Transporter in Healthy Volunteers; Open-label, Parallel Group [NCT02056743]Phase 130 participants (Actual)Interventional2013-09-30Completed
A Phase 2a, Double-blind, Randomized, Placebo-controlled, Crossover Study to Evaluate the Safety and Efficacy of AVP-923 (Dextromethorphan/Quinidine) in the Treatment of Levodopa-induced Dyskinesia in Parkinson's Disease Patients. [NCT01767129]Phase 214 participants (Actual)Interventional2013-10-16Completed
Evaluation of the Effects of Single Oral Dose and Multiple Oral Doses of BI 201335 NA on Cytochrome P450 and P-glycoprotein Activity Using a Probe Drug Cocktail. An Open-label, Single-arm Phase I Study in Healthy Human Volunteers [NCT02182336]Phase 123 participants (Actual)Interventional2008-06-30Completed
An Open-label, 2-cycle Clinical Study to Evaluate the Drug Interaction Between Itraconazole or Dextromethorphan and IBI351 in Healthy Subjects [NCT05699993]Phase 124 participants (Actual)Interventional2023-03-10Completed
Dextromethorphan as a Novel Non-opioid Adjunctive Agent for Pain Control in Medication Abortion: a Randomized Controlled Trial [NCT03480009]Phase 3156 participants (Actual)Interventional2018-07-28Completed
Dextromethorphan Enhances the Therapeutic Efficacy of Valoproate in Bipolar Disorder Patients [NCT01188265]Phase 3300 participants (Actual)Interventional2007-06-30Completed
A Phase 1, Randomized, Single-Dose, 3-Way, Crossover Study to Compare the Relative Bioavailability, Pharmacokinetics, Safety and Tolerability of AVP-923 (Dextromethorphan Hydrobromide and Quinidine Sulfate Capsules) Administered in Applesauce or Via a Nas [NCT03381664]Phase 117 participants (Actual)Interventional2017-11-28Completed
Combined Therapy of Methadone and Dextromethrophan: A Novel Strategy for the Treatment of Opioid Dependence [NCT01189097]Phase 31,500 participants (Anticipated)Interventional2008-04-30Recruiting
Phase 1, Open-label, One-arm, Fixed-sequence Study to Evaluate the Effects of 90 mg Intravenous Infusions of GC4419 on the Single Dose Pharmacokinetics of Dextromethorphan in Healthy Adult Subjects [NCT03792971]Phase 113 participants (Actual)Interventional2019-02-10Completed
A Phase 1, Randomized, Open-Label, Two-Way Crossover Study To Evaluate The Steady-State Effect Of Dimebon [PF-01913539] On The Single-Dose Pharmacokinetics Of The CYP2D6 Substrate Dextromethorphan In Healthy Subjects [NCT00788047]Phase 114 participants (Actual)Interventional2008-11-30Completed
A Phase I, Single Dose, Controlled Two-way Crossover Study to Evaluate the Relative Bioavailability of Orally Administered Dextromethorphan Syrup (21 mg Dextromethorphan Hydrobromide Monohydrate) in Comparison to Dextromethorphan Soft Pastilles (21 mg Dex [NCT02191709]Phase 118 participants (Actual)Interventional2009-01-31Completed
A Fixed-Sequence Trial to Examine the Effect of Multiple-Dose CT1812 Administration on Standard Probes of CYP2C19 (Omeprazole), CYP2C9 (Tolbutamide), CYP2D6 (Dextromethorphan), and CYP3A4/5 (Midazolam) Activity in Healthy Adult Volunteers [NCT03716427]Phase 116 participants (Actual)Interventional2016-11-10Completed
Combination of Dextromethorphan and Memantine in Treating Bipolar Disorder [NCT03039842]Phase 3300 participants (Anticipated)Interventional2013-01-01Enrolling by invitation
A Placebo-controlled, Single-blind, Randomised Study to Investigate the Safety, Tolerability, Pharmacokinetics and Drug Interaction of GSK1034702 After Repeat Doses in Healthy Subjects [NCT00950586]Phase 148 participants (Actual)Interventional2009-08-24Completed
A Randomized Clinical Trial to Assess the Effects of Single Doses of MK2637 and Dextromethorphan on Cerebral Cortex Excitability in Healthy Subjects [NCT00934466]Phase 115 participants (Actual)Interventional2009-07-31Completed
An Open-label Study to Evaluate the Safety, Tolerability and Pharmacokinetics of Cytochrome P450 Probe Drugs in Healthy Adult Subjects [NCT00964106]Phase 187 participants (Actual)Interventional2009-08-26Completed
An Abiraterone Acetate Plus Prednisone Drug-Drug Interaction Study With Dextromethorphan and Theophylline in Patients With Metastatic Castration-Resistant Prostate Cancer [NCT01017939]Phase 134 participants (Actual)Interventional2010-01-31Completed
A Phase 1 Open-label Study to Evaluate the Effect of Multiple Doses of Enzalutamide on the Pharmacokinetics of Substrates for CYP1A2 and CYP2D6 in Male Subjects With Prostate Cancer [NCT02225093]Phase 112 participants (Actual)Interventional2013-10-02Completed
A Two-cohort, Two-part, Phase 1, Multicenter, Open-label, Fixed-sequence, Drug-Drug Interaction and QTc Assessments of Sitravatinib Followed by Combination Treatment With Nivolumab in Patients With Advanced Solid Malignancies [NCT04887194]Phase 140 participants (Actual)Interventional2021-03-26Completed
A Double-Blind, Randomized, Placebo-Controlled, Multicenter Study to Assess the Safety and Efficacy and to Determine the Pharmacokinetics of Two Doses of AVP-923 (Dextromethorphan/Quinidine) in the Treatment of Pseudobulbar Affect (PBA) in Patients With A [NCT00573443]Phase 3326 participants (Actual)Interventional2007-12-31Completed
Randomized Clinical Trial to Evaluate Guidelines for Acute Rhinosinusitis (Phase IV Study) [NCT00377403]Phase 4172 participants (Actual)Interventional2006-10-31Completed
A Phase 1, Open-label, Drug Interaction Study to Evaluate the Effect of Ustekinumab on Cytochrome P450 Enzyme Activities Following Induction and Maintenance Dosing in Participants With Active Crohn's Disease or Ulcerative Colitis. [NCT03358706]Phase 157 participants (Anticipated)Interventional2018-02-02Suspended(stopped due to unavailability of probe substrates)
Trial of Dextromethorphan in Rett Syndrome [NCT00593957]Phase 238 participants (Actual)Interventional2004-08-31Terminated(stopped due to Study changed to a placebo controlled trial of dextromethorphan)
A Phase I Study to Evaluate the Effect of Darunavir/Ritonavir and Lopinavir/Ritonavir on GSK2248761 Pharmacokinetics and to Assess the Effect of GSK2248761 on CYP450 Probe Drugs in Healthy Adult Subjects [NCT00920088]Phase 124 participants (Actual)Interventional2009-06-30Completed
A Phase 1 Dose-Escalation Study of the Safety and Pharmacokinetics of GDC-0973/XL518 Administered Orally Daily to Subjects With Solid Tumors [NCT00467779]Phase 1119 participants (Actual)Interventional2007-05-31Completed
Effect of Concomitant Administration of BMS-708163 on the Pharmacokinetics of Midazolam, Warfarin, Caffeine, Omeprazole and Dextromethorphan in Healthy Male Subjects by Administration of a Modified Cooperstown Cocktail [NCT00726726]Phase 122 participants (Actual)Interventional2008-08-31Completed
A Phase II, Fixed-sequenced, Open- Label, Research Study to Assess Pharmacokinetic Drug Interactions of AEGR-733 on Lipid-lowering Therapies in Healthy Volunteers [NCT00359281]Phase 2125 participants (Actual)Interventional2006-03-31Completed
An Open-Label, 2-Part, Multicenter, Post-marketing Study to Evaluate the Effect of Moderately or Severely Active Ulcerative Colitis or Crohn's Disease on Cytochrome P-450 Enzyme Substrates Compared to Healthy Subjects and the Effect of Vedolizumab Treatme [NCT02760615]Phase 40 participants (Actual)Interventional2016-11-01Withdrawn(stopped due to No enrollment)
Evaluating the Efficacy of Dextromethorphan/Quinidine in Treating Irritability in Huntington's Disease [NCT03854019]Phase 320 participants (Actual)Interventional2019-08-05Completed
A Phase IIa, Dose-finding, Double-blind, Placebo-controlled, Double-dummy, Randomized, Eightfold Cross-over Study to Investigate the Glucose Lowering Effects of Dextromethorphan Alone or in Combination With Sitagliptin in Subjects With Type 2 Diabetes Mel [NCT01936025]Phase 234 participants (Actual)Interventional2013-10-31Completed
NMDA Receptor Antagonist Treatment of Neurodegenerative Disease [NCT00001365]Phase 242 participants Interventional1993-07-31Completed
Clinical Trials in Orofacial Neuralgias [NCT00001725]Phase 2100 participants Interventional1997-12-31Completed
Assessing the Pharmacokinetics and Drug Interaction Liability of Kratom, an Opioid-like Natural Product [NCT04392011]Early Phase 115 participants (Actual)Interventional2019-10-09Completed
Dextromethorphan as an Augmentation Agent in Treatment-resistant Schizophrenia: A Randomized, Group Sequential Adaptive Design, Controlled Clinical Trial [NCT05944510]Phase 472 participants (Anticipated)Interventional2023-08-31Recruiting
Antagonists NMDA in Relay to Ketamine in Neuropathic Pain [NCT01602185]Phase 27 participants (Actual)Interventional2012-05-31Completed
A Phase 2, Double-blind, Randomized, Placebo-controlled, Four-arm, Multicenter, Dose-finding Study to Assess the Safety and Efficacy of Three Dose Levels of AVP-923 (Dextromethorphan/Quinidine) in the Treatment of Central Neuropathic Pain in Patients With [NCT01324232]Phase 2209 participants (Actual)Interventional2011-09-08Completed
The Study of Risk Factors and Intervention for Children With Chronic Irritability - a Preliminary Approach With Epidemiological, Neuropsychological and Neuroinflammation Studies [NCT05043805]Phase 4120 participants (Anticipated)Interventional2021-05-09Enrolling by invitation
A Prospective Randomized Double-blind Placebo Controlled Trial; Does Pre and Postoperative Dextromethorphan Reduce Post-tonsillectomy Pain in Children? [NCT02727491]Phase 485 participants (Actual)Interventional2012-03-31Completed
¹³C - Dextromethorphan (DM) Breath Test for Determination of CYP2D6 Enzyme Activity in Patients Receiving Tamoxifen [NCT00873366]92 participants (Actual)Observational2009-05-31Terminated(stopped due to Funding issues)
An Open Label Phase 1, Randomized Cross Over Trial To Estimate The Effect Of PF- 00299804 On The Pharmacokinetics Of Dextromethorphan In Healthy Volunteers [NCT00999817]Phase 114 participants (Actual)Interventional2009-11-30Completed
A Randomized, Double-Blind 4-Week Study to Evaluate the Impact of AXS-05 on Smoking Behavior [NCT03471767]Phase 258 participants (Actual)Interventional2018-03-25Completed
Repurposing of Dextromethorphan as an Adjunct Therapy in Patients With Major Depressive Disorder: A Randomized, Group Sequential, Adaptive Design, Controlled Clinical Trial [NCT05181527]Phase 460 participants (Actual)Interventional2022-02-10Completed
A Randomized, Parallel-Group, Double-Blind, Placebo-Controlled, Single-Dose, Pilot Study To Evaluate Efficacy Of Dextromethorphan Hydrobromide On Acute Cough In A Pediatric Population [NCT01257542]Phase 4140 participants (Actual)Interventional2010-12-31Terminated(stopped due to See termination reason in detailed description.)
A Double-Blind Placebo-Controlled Trial of Dextromethorphan for Treatment of Major Depressive Disorder [NCT02860962]Phase 14 participants (Actual)Interventional2016-08-31Completed
[NCT00333242]Phase 140 participants (Actual)Interventional2002-09-30Active, not recruiting
A Pilot Study of Dextromethorphan for the Prevention and Treatment of Methotrexate Neurotoxicity [NCT00176553]20 participants (Anticipated)Interventional2003-03-31Terminated(stopped due to slow accrual)
An Open-Label Study to Assess the Long-term Safety and Efficacy of AXS-05 in Subjects With Treatment Resistant Depression [NCT04634669]Phase 2186 participants (Actual)Interventional2020-09-23Completed
An Open-Label Study to Assess the Long-term Safety and Efficacy of AXS-05 in Subjects With Dementia of the Alzheimer's Type [NCT04947553]Phase 3260 participants (Anticipated)Interventional2021-06-17Enrolling by invitation
A Phase 2, Multicenter, Randomized, Double-blind, Placebo-controlled Study to Assess the Efficacy, Safety, and Tolerability of AVP-786 (Deuterium Modified Dextromethorphan Hydrobromide/Quinidine Sulfate) as an Adjunctive Therapy in Patients With Major Dep [NCT02153502]Phase 2206 participants (Actual)Interventional2014-07-31Completed
Recombinant Human Growth Hormone Therapy and Drug Metabolism [NCT00458991]9 participants (Actual)Observational2001-06-30Completed
Non-Invasive Assessment of Opioid Analgesia in Children With Sickle Cell Disease [NCT00513864]Phase 40 participants (Actual)Interventional2006-11-30Withdrawn(stopped due to lack of funding)
Treatment Efficacy in Dextromethorphan, Memantine Monotherapy, or Combined Use of Dextromethorphan and Memantine in Patients With Amphetamine-type Stimulants Use Disorder [NCT04687566]Phase 2/Phase 3120 participants (Anticipated)Interventional2020-08-11Recruiting
Evaluating the Effects of Tipranavir (With Ritonavir) Capsule and Liquid Formulation on Cytochrome P450 and P-glycoprotein Activity Using a Biomarker Cocktail in Healthy Human Volunteers [NCT02243553]Phase 134 participants (Actual)Interventional2006-01-31Completed
Effectiveness of Antitussives, Anticholinergics and Honey Versus Usual Care in Adults With Uncomplicated Acute Bronchitis. [NCT03738917]Phase 4668 participants (Actual)Interventional2019-02-01Completed
The Effect of Imlunestrant on CYP2C8, CYP2C19, CYP2D6, P-gp, and BCRP Activity and the Effect of P-gp Inhibition on Imlunestrant Pharmacokinetics in Healthy Women of Non-childbearing Potential [NCT05444556]Phase 1113 participants (Actual)Interventional2022-07-07Completed
Effect of Honey and Dextromethorphan on Nocturnal Cough and Sleep Quality for Coughing Children and Their Parents [NCT00127686]Phase 1105 participants (Actual)Interventional2005-09-30Completed
Characterization of Pain Processing Mechanisms in Irritable Bowel Syndrome [NCT00108446]Phase 20 participants Interventional2003-10-31Completed
An Open-Label, Multiple-Dose, Non-Randomized Study to Assess the Drug-Drug Interactions of Proellex® (CDB-4124) With Cytochrome P450 Isoenzymes CYP1A2, 2C9, 2C19, 2D6, and 3A4 in Healthy Female Subjects [NCT00741468]Phase 118 participants (Actual)Interventional2008-07-31Completed
Dextromethorphan Versus Placebo for Neuropathic Pain [NCT00001344]Phase 2129 participants Interventional1993-03-31Completed
A Pilot Trial to Evaluate the Effects of Dextromethorphan in Patients Suffering From Cancer-Related Fatigue [NCT00176540]30 participants (Anticipated)Interventional2003-10-31Terminated(stopped due to slow accrual)
Anti-Inflammation & Vascular Endothelial Protection Effects of Dextromethorphan on Heavy Smoker [NCT00605605]Phase 440 participants (Actual)Interventional2005-03-31Completed
Effect of N-methyl D-aspartate (NMDA) Receptor Antagonist Dextromethorphan on Opiods Analgesia and Tolerance in Pediatric Intensive Care Unit Patients [NCT01553435]36 participants (Anticipated)Interventional2011-01-31Active, not recruiting
The Effects of an NMDA-Receptor Antagonist in Idiopathic Voice Disorders [NCT00055549]Phase 165 participants Interventional2003-03-04Completed
Pathogenesis of Rett Syndrome: Natural History and Treatment [NCT00069550]Phase 390 participants Interventional2004-09-30Recruiting
Dextromethorphan in Chemotherapy-induced Peripheral Neuropathy Management [NCT02271893]Phase 219 participants (Actual)Interventional2014-11-25Terminated(stopped due to Recruitment difficulties)
The Experimental Treatment of Bulbar Dysfunction in Amyotrophic Lateral Sclerosis (ALS) [NCT01806857]Phase 290 participants (Actual)Interventional2013-04-30Completed
Placebo Controlled Trial of Dextromethorphan in Rett Syndrome [NCT01520363]Phase 257 participants (Actual)Interventional2012-03-31Completed
Antitussive Effect of a Naturally Flavored, Multi-Component Syrup Containing Diphenhydramine, Compared With Dextromethorphan and Placebo [NCT02062710]Phase 422 participants (Actual)Interventional2014-01-31Completed
A Phase 1, Open-label, 2-period Fixed-sequence Study to Evaluate the Effect of Multiple Doses of SHP620 (Maribavir) on the Pharmacokinetics of Digoxin and Dextromethorphan in Healthy Adult Subjects [NCT02775240]Phase 118 participants (Actual)Interventional2016-07-21Completed
A Study to Assess the Safety, Tolerability and Effectiveness of Nuedexta (Dextromethorphan 20 mg/Quinidine 10 mg) in the Treatment of Pseudobulbar Affect (PBA) [NCT01799941]Phase 4367 participants (Actual)Interventional2013-02-28Completed
Effect of Chronic Intermittent Nocturnal Hypoxia on Hepatic Drug Biotransformation in Children With Obstructive Sleep Apnea [NCT00310323]69 participants (Actual)Interventional2003-01-31Completed
A Randomized, Placebo-Controlled Study to Evaluate the Efficacy of Perioperative Dextromethorphan Compared to Placebo for the Treatment of Postoperative Pain [NCT05278494]160 participants (Anticipated)Interventional2022-09-15Recruiting
A Phase 1, Open-Label, Sequential Study of the Effect of Multiple Doses of Isavuconazole on the Pharmacokinetics of a Single Dose of Dextromethorphan in Healthy Adult Subjects [NCT01651325]Phase 124 participants (Actual)Interventional2012-05-31Completed
[NCT01731067]Phase 110 participants (Anticipated)Interventional2012-11-30Completed
Dextromethorphan Added on Methylphenidate in the Treatment of the Patients With ADHD [NCT01787136]0 participants Expanded AccessNo longer available
Clinical Protocol of a Prospective, Open-label Study to Assess the Safety and Efficacy of Nuedexta (Dextromethorphan/Quinidine) in the Treatment of Pseudobulbar Affect (PBA) in Patients With Alzheimer's Disease [NCT01832350]Phase 434 participants (Actual)Interventional2012-08-28Terminated(stopped due to Sponsor decided to stop study early)
A Phase IIa, Double-blind, Placebo-controlled, Randomised, Fourfold Crossover Study to Investigate the Glucose Lowering Effects of Dextromethorphan and Amantadine in Subjects With Type 2 Diabetes Mellitus (T2DM) After an Oral Glucose Tolerance Test [NCT01441986]Phase 220 participants (Actual)Interventional2011-09-30Completed
A Phase 2, Multicenter, Randomized, Double-blind, Placebo-controlled Study to Assess the Safety, Tolerability, and Efficacy of AVP-786 for the Treatment of Disinhibition in Patients With Neurodegenerative Disorders [NCT02534038]Phase 21 participants (Actual)Interventional2015-12-31Terminated(stopped due to The study was terminated prematurely because of difficulty with recruiting.)
Effects of Dextromethorphan on Opioid Tolerance in Methadone Patients [NCT00000352]Phase 20 participants Interventional1996-12-31Completed
Nuedexta for Neurobehavioral Symptoms of Adults With Autism Spectrum Disorder [NCT01630811]Phase 213 participants (Actual)Interventional2012-01-24Completed
The Effects of Multiple Dose Fluoxetine and Metabolites on CYP1A2, CYP2C19, CYP2D6 and CYP3A4 Activity [NCT01361217]10 participants (Actual)Interventional2011-09-30Completed
Clinical Neuropharmacology of Pain in Spinal Cord Injury- Dextromethorphan/Lidocaine Combination (Factorial Design) Clinical Trial [NCT02218203]Phase 226 participants (Actual)Interventional2003-04-30Completed
Developmental Regulation of CYPs 1A2, 2D6, 3A4 [NCT00117715]121 participants (Actual)Observational2000-10-31Completed
A Study to Assess Effectiveness, Safety, and Health-related Outcomes of NUEDEXTA® (Dextromethorphan Hydrobromide and Quinidine Sulfate) for the Treatment of Pseudobulbar Affect (PBA) in Nursing Home Patients [NCT02496039]Phase 45 participants (Actual)Interventional2015-09-30Terminated(stopped due to The study was terminated prematurely because of difficulty with recruiting.)
Development of High-throughput Minidose Inje Cocktail Method for Simultaneous Evaluating Five Cytochrome P450 Isoforms in Human [NCT01570569]Phase 126 participants (Actual)Interventional2011-01-31Completed
An Open-Label Tolerance Study of Three Dosing Regimens of Dextromethorphan (DXM) Reportedly Used in Major Depressive Disorder [NCT04226352]Phase 15 participants (Actual)Interventional2020-03-15Completed
A Pilot Study: a Non-opioid Technique for Postoperative Adenoidectomy Pain Relief in Pediatric Patients [NCT03714919]Phase 210 participants (Actual)Interventional2019-08-02Completed
[NCT00004807]120 participants Interventional1995-01-31Completed
"An Open-label, Single-sequence Study of the Effect of Belatacept on the Pharmacokinetics of Caffeine, Losartan, Omeprazole, Dextromethorphan, and Midazolam Administered as Inje Cocktail in Healthy Subjects" [NCT01766050]Phase 445 participants (Actual)Interventional2013-01-31Completed
A Double-blind, Placebo-controlled, Therapeutic Trial With Antitussive Drug-Dextromethorphan: Aimed to Determine Its Therapeutic Effect in Patients With Rheumatoid Arthritis [NCT02368093]48 participants (Actual)Interventional2010-01-31Completed
A Phase III Double-Blind Equivalence Study of Two Different Formulations of Slow-Release Morphine Followed by a Randomization Between Dextromethorphan or Placebo Plus Statex SR for Chronic Cancer Pain Relief in Terminally Ill Patients [NCT00003687]Phase 387 participants (Actual)Interventional1998-06-11Completed
A Single Dose, Open Label, Randomized Scintigraphic Study to Investigate the Gastrointestinal Behavior of 2 Triple Combination Products (Acetaminophen, Phenylephrine and Dextromethorphan) in Healthy Male Volunteers [NCT03415243]Phase 128 participants (Actual)Interventional2018-03-01Completed
A Phase 2, Randomized, Double-dummy, Double-blind, Placebo-controlled Study to Assess the Efficacy, Safety and Tolerability of AVP-923 (Dextromethorphan/Quinidine) for the Treatment of Symptoms of Agitation in Patients With Alzheimer's Disease. [NCT01584440]Phase 2220 participants (Actual)Interventional2012-08-13Completed
An Open-label Drug Interaction Study in Healthy Subjects to Evaluate the Effect of Oral Doses of JNJ-54175446 on the Inhibition of Cytochrome P450 CYP3A4, CYP2C9, CYP1A2 and CYP2D6 Activity and the Induction of CYP2B6 and CYP2C19 Activity Using a Multiple [NCT03058419]Phase 116 participants (Actual)Interventional2017-03-14Completed
A Randomized Pilot Study of Nuedexta for the Prevention and Modification of Disease Progression in Episodic Migraine [NCT02176018]Phase 276 participants (Actual)Interventional2014-08-31Completed
Glutaminergic and Histaminergic Pathway Modulation in Acute Ischemic Stroke as an Effective Neuroprotection Strategy. [NCT02142712]Phase 23 participants (Actual)Interventional2014-12-31Completed
A Pilot Study Using an NMDA Antagonist to Modulate Transcranial Direct Current Stimulation (tDCS) Effects on Auditory Sensory Memory Processing [NCT02426983]Phase 1/Phase 212 participants (Anticipated)Interventional2015-04-30Not yet recruiting
A Phase I, Open-label, Fixed-sequence, Crossover, Drug-drug Interaction Study to Investigate the Inhibition Potential of KL1333 on CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 in Healthy Subjects [NCT04643249]Phase 114 participants (Actual)Interventional2020-11-10Completed
A Phase 1, Single-center, Randomized, Double-blind, Double-dummy, 2-way Crossover Study Comparing AVP-786 With AVP-923 [NCT02336347]Phase 162 participants (Actual)Interventional2014-05-31Completed
Targeting the NMDA Glutamate Receptor as Novel Antidepressant Strategy: A Pilot Clinical Trial of Nuedexta in Treatment-Resistant Major Depression [NCT01882829]Phase 220 participants (Actual)Interventional2013-07-31Completed
An Open-label Phase 1 Study to Evaluate Drug-Drug Interactions of Agents Co-Administered With Encorafenib and Binimetinib in Patients With BRAF V600-mutant Unresectable or Metastatic Melanoma or Other Advanced Solid Tumors [NCT03864042]Phase 156 participants (Actual)Interventional2018-01-02Active, not recruiting
A Phase I, Open-label, Fixed-sequence, Two-period, Crossover, Drug-drug Interaction Study to Evaluate the Effect of Multiple Doses of Ganaplacide and Lumefantrine Combination on the Pharmacokinetics of Midazolam, Repaglinide, Dextromethorphan, Metformin, [NCT05236530]Phase 148 participants (Actual)Interventional2022-03-09Completed
Clinical Neuropharmacology of Pain in Spinal Cord Injury- Dextromethorphan Dose Response Clinical Trial [NCT01435798]Phase 226 participants (Actual)Interventional2003-04-30Completed
A Pilot Phase I/II Study for the Evaluation of Dextromethorphan as a Microglia Inhibitor in the Treatment of Diabetic Macular Edema (MiDME2) [NCT01441102]Phase 1/Phase 27 participants (Actual)Interventional2011-08-31Completed
Pharmacological Mechanisms of Low-intensity Focused Ultrasound for Motor Cortex Neuroplasticity [NCT04923659]Early Phase 120 participants (Anticipated)Interventional2021-05-25Recruiting
A Randomized Double-blinded Study to Evaluate Preincisional Dextromethorphan in Patients Undergoing Total Knee Arthroplasty and Its Effect on Postoperative Opioid Use [NCT02987920]Phase 423 participants (Actual)Interventional2017-01-31Terminated(stopped due to The surgeon changed pain control protocol for all patients. Continued enrollment impossible under approved protocol.)
Pharmacogenetic Prediction of Metoprolol Effectiveness [NCT02293096]462 participants (Actual)Interventional2014-09-30Terminated(stopped due to Study was terminated due to the change in funding.)
A Multicenter, Single-Arm, Open-Label Study to Evaluate the Immunogenicity and Pharmacokinetics of BIIB019, Daclizumab High Yield Process (DAC HYP), Prefilled Syringe Administered by Subcutaneous Injection in Subjects With Relapsing-Remitting Multiple Scl [NCT01462318]Phase 3133 participants (Actual)Interventional2011-11-30Completed
Effects of Isotretinoin on CYP2D6 Activity [NCT03076021]Phase 436 participants (Actual)Interventional2016-07-26Completed
The Effect of High Dose Rifampicin on the Activity of Cytochrome P450 Enzymes and P-glycoprotein in Patients With Pulmonary Tuberculosis: a Cocktail Phenotyping Study [NCT04525235]Phase 125 participants (Actual)Interventional2021-01-07Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00117715 (3) [back to overview]Change in CYP1A2 Drug Metabolism Phenotype With Age
NCT00117715 (3) [back to overview]Change in CYP3A4 Drug Metabolism Phenotype With Age
NCT00117715 (3) [back to overview]Change in CYP2D6 Drug Metabolism Phenotype With Age
NCT00359281 (11) [back to overview]AUC0-t Simvastatin Acid
NCT00359281 (11) [back to overview]AUC0-t Nicotinic Acid
NCT00359281 (11) [back to overview]AUC0-t Nicotinuric Acid
NCT00359281 (11) [back to overview]AUC0-t Rosuvastatin (Lomitapide 10 mg)
NCT00359281 (11) [back to overview]AUC0-t Rosuvastatin (Lomitapide 60 mg)
NCT00359281 (11) [back to overview]Percent Change From Baseline in Low-density Lipoprotein Cholesterol (LDL-C)
NCT00359281 (11) [back to overview]AUC0-t Total Ezetimibe
NCT00359281 (11) [back to overview]AUC0-t Atorvastatin Acid (Lomitapide 60 mg)
NCT00359281 (11) [back to overview]AUC0-t Fenofibric Acid
NCT00359281 (11) [back to overview]AUC0-t Simvastatin
NCT00359281 (11) [back to overview]Area Under Concentration-time Curve From 0 to Last Measureable Concentration (AUC0-t) Atorvastatin Acid (Lomitapide 10 mg)
NCT00377403 (1) [back to overview]SNOT-16 Score (Sino-Nasal Outcomes Test) at Day 3
NCT00467779 (44) [back to overview]Stage 1A: MTD of Cobimetinib in 14/14 Schedule
NCT00467779 (44) [back to overview]Stage 1A: Cmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1A: Cmax of Cobimetinib at Cycle 1 Day 1
NCT00467779 (44) [back to overview]Stage 1A: AUC 0-24 of Cobimetinib at Cycle 1 Day 1
NCT00467779 (44) [back to overview]Stage 1A: Apparent Clearance of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1A: AUC 0-24/D of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1A: AUC 0-24 of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1A: Accumulation Ratio of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: Tmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: Time to Maximum Concentration (Tmax) of Cobimetinib at Day 1, Cycle 1
NCT00467779 (44) [back to overview]Stage 1: Maximum Tolerated Dose (MTD) of Cobimetinib in 21/7 Schedule
NCT00467779 (44) [back to overview]Stage 1: Maximum Observed Concentration (Cmax) of Cobimetinib at Day 1, Cycle 1
NCT00467779 (44) [back to overview]Stage 1: Half-Life (t1/2) of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: Cmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: AUC 0-24/D of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: AUC 0-24 of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: Area Under the Plasma Cobimetinib Concentration Curve From Time 0 to 24 Hours (AUC 0-24) Day 1, Cycle 1
NCT00467779 (44) [back to overview]Stage 1: Apparent Clearance of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1: Accumulation Ratio of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1 and 1A: Number of Participants With Dose Limiting Toxicities (DLTs)
NCT00467779 (44) [back to overview]Stage 2A: Accumulation Ratio of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2A: Tmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2: Accumulation Ratio of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2: Tmax of Cobimetinib at Cycle 1 Day 1
NCT00467779 (44) [back to overview]Stage 2: Tmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2:AUC 0-24 of Cobimetinib at Cycle 1 Day 1
NCT00467779 (44) [back to overview]Stage 2:Half-Life of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage III: AUC 0-24 of Dextromethorphan
NCT00467779 (44) [back to overview]Stage 2A: Apparent Clearance of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2A: AUC 0-24/D of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2A: Cmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 1A: Tmax of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2A: Half-Life of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage III: Cmax of Dextromethorphan
NCT00467779 (44) [back to overview]Stage III: AUC0-inf of Midazolam
NCT00467779 (44) [back to overview]Stage III: AUC0-24 of Midazolam
NCT00467779 (44) [back to overview]Stage III: AUC 0-inf of Dextromethorphan
NCT00467779 (44) [back to overview]Stage III: Cmax of Midazolam
NCT00467779 (44) [back to overview]Stage 2: Apparent Clearance of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2: AUC 0-24 of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2: AUC 0-24/D of Cobimetinib at Steady State
NCT00467779 (44) [back to overview]Stage 2: Cmax of Cobimetinib at Cycle 1 Day 1
NCT00467779 (44) [back to overview]Stage 1A: Tmax of Cobimetinib at Cycle 1 Day 1
NCT00467779 (44) [back to overview]Stage 1A: t1/2 of Cobimetinib at Steady State
NCT00573443 (7) [back to overview]Mean Change From Baseline at Day 84 in Beck Depression Inventory (BDI-II) Total Score
NCT00573443 (7) [back to overview]Mean Change From Baseline to Day 84 in Pain Rating Scale (PRS) of MS Subjects
NCT00573443 (7) [back to overview]Mean Change From Baseline to Day 84 in Neuropsychiatric Inventory (NPI-Q) Frequency and Severity Score (ITT Population)
NCT00573443 (7) [back to overview]Mean Change From Baseline to Day 84 in Neuropsychiatric Inventory (NPI-Q) Frequency and Severity Score (EE Population)
NCT00573443 (7) [back to overview]Mean Change From Baseline in CNS-LS Total Score by Visit
NCT00573443 (7) [back to overview]Mean Change From Baseline at Day 84 in SF-36 (Short-Form) Health Survey Medical Outcome Score by Category
NCT00573443 (7) [back to overview]PBA Episode Rate Ratio (Post/Pre), Regression Adjusted
NCT00593957 (4) [back to overview]Difference in EEG Spike Counts at Six Months Compared to Baseline for Each Treatment Arm.
NCT00593957 (4) [back to overview]Mean SSI Score for Total Subjects at Baseline and 6 Months
NCT00593957 (4) [back to overview]Improvement in Receptive Language as Measured by the Mullen Scale.
NCT00593957 (4) [back to overview]Difference in SSI Mean Score at Six Months Compared to Baseline for Each Treatment Arm.
NCT00741468 (1) [back to overview]Plasma AUC Ratio of Day 1 and Day 8
NCT00873366 (16) [back to overview]Median of 3 Month Tamoxifen Steady State Plasma Concentrations According to CYP2D6 Phenotype Group
NCT00873366 (16) [back to overview]Median of 3 Month N-desmethyl-tamoxifen (NDMT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group
NCT00873366 (16) [back to overview]Median of 3 Month Endoxifen Steady State Concentrations (Endx Css) According to CYP2D6 Phenotype Group and Activity Score
NCT00873366 (16) [back to overview]Median of 3 Month 4-hydroxy-tamoxifen (4HT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group
NCT00873366 (16) [back to overview]Median of 6 Month 4-hydroxy-tamoxifen (4HT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group
NCT00873366 (16) [back to overview]Median of 6 Month Endoxifen Steady State Concentrations (Endx Css) According to CYP2D6 Phenotype Group and Activity Score
NCT00873366 (16) [back to overview]Median of 6 Month N-desmethyl-tamoxifen (NDMT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group
NCT00873366 (16) [back to overview]Median of 6 Month Tamoxifen Steady State Plasma Concentrations According to CYP2D6 Phenotype Group
NCT00873366 (16) [back to overview]Operating Characteristics of the ¹³C-dextromethorphan (13C-DM) Breath Test in Identifying Those Who Are CYP2D6 Genotypic Poor Metabolizers
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between CYP2D6 Activity Score and Endoxifen Steady State Concentrations (Endx Css)
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between CYP2D6 Activity Score and Endoxifen/N-desmethyl-tamoxifen (Endx/NDMT) Ratio
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between CYP2D6 Genotype and ¹³Cdextromethorphan Breath Test (DM-BT)
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between Baseline DM-BT and 3 Month Endoxifen/N-desmethyl-tamoxifen (Endx/NDMT) Ratio
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between 6 Month DM-BT and 6 Month Endoxifen Steady State Concentrations
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between 3 Month DM-BT and 3 Month Endoxifen Steady State Concentrations
NCT00873366 (16) [back to overview]Spearman's Rank Correlation Coefficient Between Baseline DM-BT and 3 Month Endoxifen Steady State Concentrations
NCT01257542 (7) [back to overview]Change From Baseline in Perceived Numerical Cough Severity Scale for 6 Hour Post-Dose Period
NCT01257542 (7) [back to overview]Change From Baseline in Perceived Verbal Cough Severity Scale for 6 Hour Post-Dose Period
NCT01257542 (7) [back to overview]Participants' Global Assessment of Cough: Cough Severity
NCT01257542 (7) [back to overview]Participants' Global Assessment of Cough: Relief From Cough
NCT01257542 (7) [back to overview]Total Cough Count
NCT01257542 (7) [back to overview]Change From Baseline in Verbal Cough Severity Scale at Hours 1, 2, 3, 4, 5 and 6
NCT01257542 (7) [back to overview]Change From Baseline in Numerical Cough Severity Scale at Hours 1, 2, 3, 4, 5 and 6
NCT01324232 (12) [back to overview]Average Logarithms of Dextromethorphan (DM) Plasma Concentrations (Cmax) on Days 22 and 50
NCT01324232 (12) [back to overview]Mean Change From Baseline in Pittsburgh Sleep Quality Index (PSQI) Scores at Day 85
NCT01324232 (12) [back to overview]Mean Overall Patient Global Impression of Change (PGIC) Scores at Day 85
NCT01324232 (12) [back to overview]Mean Change From Baseline in Expanded Disability Status Scale (EDSS) Scores at Days 22 and 85
NCT01324232 (12) [back to overview]Association Between the Dextromethorphan Plasma Concentration and the Change From Baseline Pain Rating Scale (PRS) Score to the Average Pain Rating Scale Score During Days 57 Through 84
NCT01324232 (12) [back to overview]Comparison of the Adjusted Mean Change From Baseline PRS Score to the Average PRS Score During Days 57 Through 84
NCT01324232 (12) [back to overview]Mean Change From Baseline in Beck Depression Inventory (BDI-II) Scores at Day 85
NCT01324232 (12) [back to overview]Mean Change From Baseline in Fatigue Severity Scale (FSS) Scores at Days 57 Through 84
NCT01324232 (12) [back to overview]Mean Change From Baseline in MS Neuropsychological Screening Questionnaire (MSNQ) Scores at Day 85
NCT01324232 (12) [back to overview]Mean Change From Baseline in Multiple Sclerosis Impact Scale-29 (MSIS-29) Scores at Day 85
NCT01324232 (12) [back to overview]Mean Change From Baseline in Symbol Digit Modalities Test (SDMT) Scores at Day 85
NCT01324232 (12) [back to overview]Mean Numerical Rating Scale (NRS) Scores at Days 22, 50, and 85
NCT01361217 (2) [back to overview]AUC of Dextromethorphan, Midazolam and Omeprazole in the Presence of Fluoxetine
NCT01361217 (2) [back to overview]Lovastatin AUC in the Presence of Fluoxetine
NCT01435798 (1) [back to overview]Mean Pain Intensity (Percent Change From Baseline)
NCT01441102 (17) [back to overview]Changes in Mean Macular Sensitivity in the Study Eye at 24 Months Compared to Baseline
NCT01441102 (17) [back to overview]Number of Participants Withdrawn From the Study Therapy Due to Vision Loss or Adverse Events
NCT01441102 (17) [back to overview]Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 12 Months Compared to Baseline
NCT01441102 (17) [back to overview]Changes in Mean Macular Sensitivity in the Study Eye at 6 Months Compared to Baseline
NCT01441102 (17) [back to overview]Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 18 Months Compared to Baseline
NCT01441102 (17) [back to overview]Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 24 Months Compared to Baseline
NCT01441102 (17) [back to overview]Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 6 Months Compared to Baseline
NCT01441102 (17) [back to overview]Changes in Mean Macular Sensitivity in the Study Eye at 12 Months Compared to Baseline
NCT01441102 (17) [back to overview]Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 12 Months Compared to Baseline
NCT01441102 (17) [back to overview]Changes in Mean Macular Sensitivity in the Study Eye at 18 Months Compared to Baseline
NCT01441102 (17) [back to overview]Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 18 Months Compared to Baseline
NCT01441102 (17) [back to overview]Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 24 Months Compared to Baseline
NCT01441102 (17) [back to overview]Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 6 Months Compared to Baseline
NCT01441102 (17) [back to overview]Percentage Change in Retinal Thickness in the Study Eye at 12 Months Compared to Baseline
NCT01441102 (17) [back to overview]Percentage Change in Retinal Thickness in the Study Eye at 18 Months Compared to Baseline
NCT01441102 (17) [back to overview]Percentage Change in Retinal Thickness in the Study Eye at 24 Months Compared to Baseline
NCT01441102 (17) [back to overview]Percentage Change in Retinal Thickness in the Study Eye at 6 Months Compared to Baseline
NCT01462318 (14) [back to overview]TP-DI Sub-study: Dextromethorphan to Dextrorphan Urine Concentration Ratio
NCT01462318 (14) [back to overview]TP-DI Sub-study: Omeprazole/Hydroxyomeprazole Concentration Ratio at 2 Hours Post-omeprazole Dosing
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Apparent Clearance (CL/F) of DAC HYP
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Apparent Volume of Distribution (V/F) of DAC HYP
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Elimination Half-life (t½) of DAC HYP
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Minimum Concentrations (Cmin) of DAC HYP
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Area-Under-the-Curve From Start to End of the Dosing Interval (AUCtau) of DAC HYP
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Cmax of DAC HYP
NCT01462318 (14) [back to overview]Intensive PK Sub-study: Time to Reach Maximum Concentration (Tmax) of DAC HYP
NCT01462318 (14) [back to overview]Number of Participants With Anti-DAC HYP Binding Antibodies (ADAbs): Electrochemiluminescent (ECL) Anti-Drug Antibody (ADA) Assay
NCT01462318 (14) [back to overview]Number of Participants With Anti-DAC HYP Neutralizing Antibodies (NAbs): ECL ADA Assay
NCT01462318 (14) [back to overview]TP-DI Sub-study: Area-Under-the-Curve From Zero to Infinity (AUCinf) of Each Probe Drug
NCT01462318 (14) [back to overview]TP-DI Sub-study: CL/F of Each Probe Drug
NCT01462318 (14) [back to overview]TP-DI Sub-study: Cmax of Each Probe Drug
NCT01520363 (17) [back to overview]Change in PedsQL Social Functioning Subscale Score, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in PedsQL Total Score, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in Rett Syndrome Behavior Questionnaire Score, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in Seizure Frequency, Pre- and Post-Intervention, 0-4 Year Age Group
NCT01520363 (17) [back to overview]Change in Seizure Frequency, Pre-and Post-Intervention, 5-10 Year Age Group
NCT01520363 (17) [back to overview]Change in VABS: Socialization Domain Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in VABS: Motor Skills Domain Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in VABS:Communication Domain Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in PedsQL Emotional Functioning Subscale Score, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in VABS:Daily Living Skills Domain Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in Ghuman-Folstein Screen for Social Interaction (SSI) Score, Pre- and Post-Intervention.
NCT01520363 (17) [back to overview]Change in Mullen, Expressive Language Sub-scale Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in Mullen; Fine Motor Sub-scale Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in Mullen; Receptive Language Subscale Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in Mullen; Visual Reception Sub-scale Scores, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in PedsQL Physical Functioning Subscale Score, Pre- and Post-Intervention
NCT01520363 (17) [back to overview]Change in PedsQL School Functioning Subscale Score, Pre- and Post-Intervention
NCT01584440 (22) [back to overview]Number of Participants With the Indicated Change in the Concomitant Use of Allowed Psychotropic Drugs Compared to Their Baseline Use
NCT01584440 (22) [back to overview]Change in the Caregiver Strain Index (CSI) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the NPI Agitation/Aggression Domain Score From Day 1 (Stage 1 Baseline) to Day 8 and Day 22 and From Day 36 (Stage 2 Baseline) to Day 43 and Day 57
NCT01584440 (22) [back to overview]Change in the Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory (ADCS-ADL) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Cornell Scale for Depression in Dementia (CSDD) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Individual NPI Domain Scores From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Mini-Mental State Examination (MMSE) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline), as Analyzed by the Specified SPCD Methodology
NCT01584440 (22) [back to overview]Change in the Neuropsychiatric Inventory (NPI) Agitation/Aggression Domain Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the NPI-CDS for the Agitation/Aggression Domain From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the NPI-CDS NPI4A Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the NPI-CDS NPI4D Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Quality of Life-Alzheimer's Disease (QoL-AD) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Sum of the Agitation/Aggression, Irritability/Lability, Anxiety, and Aberrant Motor Behavior NPI Domain (NPI4A) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Sum of the Agitation/Aggression, Irritability/Lability, Disinhibition, and Aberrant Motor Behavior NPI Domain (NPI4D) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Total Neuropsychiatric Inventory-Caregiver Distress Score (NPI-CDS) From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Change in the Total NPI Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70
NCT01584440 (22) [back to overview]Number of Participants Using Rescue Medications
NCT01584440 (22) [back to overview]Number of Participants With the Indicated Categorical Response on the Patient Global Impression of Change (PGI-C) for the Caregiver Domain at Day 36 (Stage 2 Baseline) and Day 70 Compared to Their Response at Day 1 (Stage 1 Baseline)
NCT01584440 (22) [back to overview]Number of Participants With the Indicated Categorical Response on the PGI-C for the Caregiver Domain at Day 70 Compared to Their Response at Day 1 (Stage 1 Baseline)
NCT01584440 (22) [back to overview]Number of Participants With the Indicated Response on the Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change Rating (mADCS-CGIC) Scale Agitation Domain at Day 36 and Day 70 Compared to Their Response at Day 1 (Stage 1 Baseline)
NCT01584440 (22) [back to overview]Number of Participants With the Indicated Response on the mADCS-CGIC Scale Agitation Domain at Day 70 Compared to Their Response at Day 36 (Stage 2 Baseline)
NCT01584440 (22) [back to overview]Number of Participants With the Indicated Type of Adverse Event
NCT01630811 (3) [back to overview]Primary Safety Endpoints
NCT01630811 (3) [back to overview]Change in Maladaptive Behaviors
NCT01630811 (3) [back to overview]Change in Aggressive Behavior
NCT01766050 (36) [back to overview]Adjusted Geometric Mean Cmax of Dextromethorphan With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Adjusted Geometric Mean Cmax of Caffeine With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Mean Change From Baseline in Sitting Heart Rate - All Treated Participants
NCT01766050 (36) [back to overview]Cmax of Inje Cocktail Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]AUC(INF) of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Adjusted Geometric Mean Area Under the Concentration Time Curve (AUC) From Zero to Last Concentration (0-T) and AUC Extrapolated to Infinity (INF) of Midazolam With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population
NCT01766050 (36) [back to overview]Apparent Total Body Clearance (CLT/F) of the Inje Cocktail Components (Midazolam, Losartan, Omeprazole, Dextromethorphan and Caffeine) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Number of Participants With Marked Hematology and Urinalysis Laboratory Abnormalities - All Treated Participants
NCT01766050 (36) [back to overview]Number of Participants With Marked Serum Chemistry Laboratory Abnormalities - All Treated Participants
NCT01766050 (36) [back to overview]Number of Participants With Out-of-Range Electrocardiogram Intervals - All Treated Participants
NCT01766050 (36) [back to overview]Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Omeprazole With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population
NCT01766050 (36) [back to overview]Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Losartan With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population
NCT01766050 (36) [back to overview]AUC(0-T) of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Plasma Half-Life (T-HALF) of the Inje Cocktail Components (Midazolam, Losartan, Omeprazole, Dextromethorphan, and Caffeine) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Dextromethorphan With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of 1'-Hydroxy-Midazolam AUC(0-T) to Midazolam AUC(0-T) and 1'-Hydroxy-Midazolam AUC(INF) to Midazolam AUC(INF), Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC (INF)] With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of Paraxanthine (Cmax) to Caffeine (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of E-3174 (Cmax) to Losartan (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of 5-Hydroxyomeprazole (Cmax) to Omeprazole (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of 5-Dextrorphan (Cmax) to Dextromethorphan (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of 5-Dextrorphan AUC(0-T) to Dextromethorphan AUC(0-T) and 5-Dextrorphan AUC(INF) to Dextromethorphan AUC(INF), Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC (INF)] With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of 5-Hydroxyomeprazole AUC(0-T) to Omeprazole AUC(0-T) and 5-Hydroxyomeprazole AUC(INF) to Omeprazole AUC(INF) , Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC(INF)] With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of E-3174 AUC(0-T) to Losartan AUC(0-T) and E3174 AUC (INF) to Losartan AUC (INF) Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC(INF)] With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of Paraxanthine AUC(0-T) to Caffeine AUC(0-T) and Paraxanthine AUC (INF) to Caffeine AUC (INF), Corrected for Molecular Weight [MR_AUC(0-T) and MR_AUC (INF)] With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]T-HALF of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Time of Maximum Observed Plasma Concentration (Tmax) of the Inje Cocktail Components (Midazolam, Losartan, Omeprazole, Dextromethorphan, and Caffeine) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Tmax of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Ratio of 1'-Hydroxy-Midazolam (Cmax) to Midazolam (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Adjusted Geometric Mean Cmax of Omeprazole With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population
NCT01766050 (36) [back to overview]Mean Change From Baseline in Heart Rate at Study Discharge (Day 46±2 Days)
NCT01766050 (36) [back to overview]Adjusted Geometric Mean Maximum Drug Concentration (Cmax) of Midazolam With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population
NCT01766050 (36) [back to overview]Adjusted Geometric Mean Cmax of Losartan With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01766050 (36) [back to overview]Mean Change From Baseline in Sitting Systolic and Diastolic Blood Pressure - All Treated Participants
NCT01766050 (36) [back to overview]Mean Change From Baseline in Systolic and Diastolic Blood Pressure at Study Discharge (Day 46±2 Days)
NCT01766050 (36) [back to overview]Number of Participants With Adverse Events (AEs), Serious AEs (SAEs), Deaths, and AEs Leading to Discontinuation - All Treated Participants
NCT01766050 (36) [back to overview]Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Caffeine With and Without the Coadministration of Belatacept - PK Evaluable Population
NCT01767129 (11) [back to overview]Change From Baseline in the Dyskinesia and Other PD Symptoms Score As Assessed by Patient Global Impression of Change (PGIC)
NCT01767129 (11) [back to overview]Change From Baseline in MDS-UDysRS Scores for Part 1 and 2
NCT01767129 (11) [back to overview]"Change From Baseline in PD Motor Diary Ratings Of Duration Of On-time Without Bothersome Dyskinesia"
NCT01767129 (11) [back to overview]Change From Baseline in PDQ-39 Single Index (PDQ-39-SI) Scores at the End of Each Treatment Period
NCT01767129 (11) [back to overview]Change From Screening in the Montreal Cognitive Assessment (MoCA) Calculated Score at the End of Each Treatment Period
NCT01767129 (11) [back to overview]Least Squares Mean Disability Area Under the Curve (AUC) Score As Assessed By Modified Movement Disorder Society-Unified Dyskinesia Rating Scale (MDS-UDysRS) Part 4
NCT01767129 (11) [back to overview]Least Squares Mean Dyskinesia Severity Area Under the Curve (AUC) Score As Assessed By Modified Movement Disorder Society-Unified Dyskinesia Rating Scale (MDS-UDysRS) Part 3
NCT01767129 (11) [back to overview]Least Squares Mean Motor Movement Area Under the Curve Score As Assessed by Modified Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS), Part III
NCT01767129 (11) [back to overview]Least Squares Mean Timed Finger Tapping Area Under the Curve (AUC) Score
NCT01767129 (11) [back to overview]Change From Baseline in MDS-UPDRS Scores for Part I, II, and IV
NCT01767129 (11) [back to overview]Change From Baseline in Parkinson's Disease Questionnaire-39 (PDQ-39) Domain Scores at the End of Each Treatment Period
NCT01799941 (12) [back to overview]Percentage of Participants With Clinical Global Impression-Change (CGI-C) Score at Day 90
NCT01799941 (12) [back to overview]Percentage of Participants With Patient Global Impression-Change (PGI-C) Score at Day 90
NCT01799941 (12) [back to overview]Percentage of Participants With PBA Remission
NCT01799941 (12) [back to overview]Percentage of Participants With Treatment Satisfaction Survey
NCT01799941 (12) [back to overview]Mean Change From Baseline in Center for Neurologic Study-Lability Scale (CNS-LS) Score at Day 30
NCT01799941 (12) [back to overview]Mean Change From Baseline in Center for Neurologic Study-Lability Scale (CNS-LS) Score at Day 90
NCT01799941 (12) [back to overview]Mean Change From Baseline in Quality of Life Visual Analog Scale (QOL-VAS) Score at Day 90
NCT01799941 (12) [back to overview]Mean Pseudobulbar Affect (PBA) Episode Count Per Week by Visit
NCT01799941 (12) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT01799941 (12) [back to overview]Percentage Change From Baseline in PBA Episode Count Per Week
NCT01799941 (12) [back to overview]Percentage of Participants With ≥ 50% Reduction in PBA Episode Count Per Week
NCT01799941 (12) [back to overview]Percentage of Participants With ≥ 75% Reduction in PBA Episode Count Per Week
NCT01806857 (16) [back to overview]Visual Analog Scale - Swallowing Score
NCT01806857 (16) [back to overview]Average Water Swallowing Test (WST)
NCT01806857 (16) [back to overview]Average Solids Swallowing Test
NCT01806857 (16) [back to overview]Ashworth Spasticity Scale Score - Right Leg
NCT01806857 (16) [back to overview]Ashworth Spasticity Scale Score - Right Arm
NCT01806857 (16) [back to overview]Ashworth Spasticity Scale Score - Left Leg
NCT01806857 (16) [back to overview]ALS Functional Rating Scale- Revised (ALSFRS-R) Total Score
NCT01806857 (16) [back to overview]Ashworth Spasticity Scale Score - Left Arm
NCT01806857 (16) [back to overview]Visual Analog Scale - Speech Scores
NCT01806857 (16) [back to overview]Visual Analog Scale - Salivation (Sialorrhea) Score
NCT01806857 (16) [back to overview]Timed Reading of Test Paragraph Result
NCT01806857 (16) [back to overview]Center for Neurologic Study - Lability Scale (CNS-LS) Total Score
NCT01806857 (16) [back to overview]Bulbar Function Scale (CNS-BFS) Total Score
NCT01806857 (16) [back to overview]Bulbar Function Scale (CNS-BFS) Swallowing Score
NCT01806857 (16) [back to overview]Bulbar Function Scale (CNS-BFS) Speech Score
NCT01806857 (16) [back to overview]Bulbar Function Scale (CNS-BFS) Sialorrhea Score
NCT01882829 (11) [back to overview]Beck Scale for Suicidal Ideation (BSI)
NCT01882829 (11) [back to overview]Sheehan Disability Scale
NCT01882829 (11) [back to overview]Range of Impaired Functioning Tool
NCT01882829 (11) [back to overview]Quick Inventory of Depressive Symptomatology, Self Report (QIDS-SR)
NCT01882829 (11) [back to overview]Quality of Life Enjoyment and Satisfaction Questionnaire Short Form
NCT01882829 (11) [back to overview]Patient Rated Inventory of Side Effects (PRISE)
NCT01882829 (11) [back to overview]Montgomery-Asberg Depression Rating Scale
NCT01882829 (11) [back to overview]Massachusetts General Hospital Cognitive and Physical Functioning Questionnaire (CPFQ)
NCT01882829 (11) [back to overview]HAM-A
NCT01882829 (11) [back to overview]Columbia-Suicide Severity Rating Scale (C-SSRS)
NCT01882829 (11) [back to overview]Clinical Global Impression (CGI) Scale
NCT02062710 (1) [back to overview]Change in Cough Reflex Sensitivity to Capsaicin
NCT02142712 (2) [back to overview]Glasgow Coma Scale (GCS)
NCT02142712 (2) [back to overview]National Institutes of Health Stroke Severity (NIHSS) Scale
NCT02176018 (10) [back to overview]Adverse Events
NCT02176018 (10) [back to overview]Headache Days in Treatment Period Month 3
NCT02176018 (10) [back to overview]50% Headache Reduction
NCT02176018 (10) [back to overview]Headache Days in Each Treatment Period Month
NCT02176018 (10) [back to overview]Headache Duration in Each Treatment Period Month
NCT02176018 (10) [back to overview]Headache Health Score
NCT02176018 (10) [back to overview]Headache Severity in Each Treatment Period Month
NCT02176018 (10) [back to overview]Migraine Days in Each Treatment Period Month
NCT02176018 (10) [back to overview]Acute Medication Use in Each Treatment Period Month
NCT02176018 (10) [back to overview]Migraine Disability Assessment Scale (MIDAS)
NCT02218203 (1) [back to overview]Percent Change in Peak Pain Intensity
NCT02293096 (4) [back to overview]Adverse Drug Events: CYP2D6 Metabolizer Status
NCT02293096 (4) [back to overview]Adverse Drug Events: ADRB1 Genotype
NCT02293096 (4) [back to overview]Heart Rate Decline
NCT02293096 (4) [back to overview]Blood Pressure Decline
NCT02368093 (1) [back to overview]Good European League Against Rheumatism (EULAR) Therapeutic Response Rate
NCT02651116 (13) [back to overview]Mean of Total Cough Counts: Between Dose 1 to Dose 2 on Day 1
NCT02651116 (13) [back to overview]Mean of Total Cough Counts: Between Dose 2 on Day 1 to Dose 3 on Day 2
NCT02651116 (13) [back to overview]Mean of Total Cough Counts: Between Dose 3 to Dose 4 on Day 2
NCT02651116 (13) [back to overview]Mean of Total Cough Time Accumulated Over a 24-Hour Period Post-First Dose on Day 1
NCT02651116 (13) [back to overview]Change From Baseline in Afternoon Cough Frequency Assessed at Afternoon on Day 2, 3, and 4
NCT02651116 (13) [back to overview]Change From Baseline in Afternoon Cough Severity Assessed at Afternoon on Day 2, 3, and 4
NCT02651116 (13) [back to overview]Change From Baseline in Child Global Question Assessed at Afternoon on Day 2, 3, and 4
NCT02651116 (13) [back to overview]Change From Baseline in Impact of Cough on Sleep Assessed in Morning at Day 2, 3, and 4
NCT02651116 (13) [back to overview]Change From Baseline in Morning Cough Frequency Assessed in Morning at Day 2, 3, and 4
NCT02651116 (13) [back to overview]Change From Baseline in Morning Cough Severity Assessed in Morning at Day 2, 3, and 4
NCT02651116 (13) [back to overview]Pediatric Global Assessment of Satisfaction With Study Medication: By Participant, and Caregiver
NCT02651116 (13) [back to overview]Mean of Total Cough Counts: Over 24 Hours Post-First Dose on Day 1
NCT02651116 (13) [back to overview]Mean of Total Cough Counts: Between Dose 1 to Dose 2 on Day 1, and Between Dose 3 to Dose 4 on Day 2
NCT02688088 (14) [back to overview]Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of Caffeine
NCT02688088 (14) [back to overview]Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of Dextromethorphan
NCT02688088 (14) [back to overview]Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of Midazolam
NCT02688088 (14) [back to overview]Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of S-Warfarin
NCT02688088 (14) [back to overview]Mean Change From Baseline at 24 Hours in Pulse Rate in Period 1
NCT02688088 (14) [back to overview]Pharmacokinetics: Maximum Concentration (Cmax) of Caffeine
NCT02688088 (14) [back to overview]Pharmacokinetics: Maximum Concentration (Cmax) of Dextromethorphan
NCT02688088 (14) [back to overview]Pharmacokinetics: Maximum Concentration (Cmax) of Midazolam
NCT02688088 (14) [back to overview]Mean Change From Baseline at 24 Hours in Pulse Rate in Period 2
NCT02688088 (14) [back to overview]Pharmacokinetics: Maximum Concentration (Cmax) S-Warfarin
NCT02688088 (14) [back to overview]Mean Change From Baseline at 24 Hours in Systolic and Diastolic Blood Pressure in Period 1
NCT02688088 (14) [back to overview]Mean Change From Baseline at 24 Hours in Systolic and Diastolic Blood Pressure in Period 2
NCT02688088 (14) [back to overview]Mean Change From Baseline at 24 Hours in Systolic and Diastolic Blood Pressure in Period 2
NCT02688088 (14) [back to overview]Mean Change From Baseline at 24 Hours in Pulse Rate in Period 2
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) of Dextrorphan
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) of Dextromethorphan
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) of Digoxin
NCT02775240 (33) [back to overview]Apparent Oral Clearance (CL/F) of Digoxin
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve From Time Zero to the End of the Dosing Interval at Steady-State (AUCtau) of Maribavir
NCT02775240 (33) [back to overview]Concentration at the End of Dosing Interval (Ctau) of Maribavir
NCT02775240 (33) [back to overview]Apparent Oral Clearance (CL/F) of Maribavir
NCT02775240 (33) [back to overview]Volume of Distribution Divided by the Fraction of Dose Absorbed (Vz/F) of Dextromethorphan
NCT02775240 (33) [back to overview]Terminal Half-life (t1/2) of Dextromethorphan
NCT02775240 (33) [back to overview]Parent/Metabolite Ratio of Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) for Dextromethorphan Over AUC0-infinity for Dextrorphan (AUC0-infinity Parent/Metabolite Ratio)
NCT02775240 (33) [back to overview]Number of Participants With Study-related Adverse Events (AEs), Serious Adverse Events (SAEs) and Treatment-emergent Adverse Events (TEAEs)
NCT02775240 (33) [back to overview]First-order Rate Constant (Lambda z) Associated With the Terminal (Log-linear) Portion of the Curve of Dextromethorphan
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) of Dextromethorphan
NCT02775240 (33) [back to overview]Apparent Oral Clearance (CL/F) of Dextromethorphan
NCT02775240 (33) [back to overview]Volume of Distribution Divided by the Fraction of Dose Absorbed (Vz/F) of Digoxin
NCT02775240 (33) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax) of Maribavir
NCT02775240 (33) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax) of Digoxin
NCT02775240 (33) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax) of Dextrorphan
NCT02775240 (33) [back to overview]Time to Reach Maximum Plasma Concentration (Tmax) of Dextromethorphan
NCT02775240 (33) [back to overview]Terminal Half-life (t1/2) of Maribavir
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) of Dextrorphan
NCT02775240 (33) [back to overview]Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) of Digoxin
NCT02775240 (33) [back to overview]Terminal Half-life (t1/2) of Digoxin
NCT02775240 (33) [back to overview]Number of Participants With Clinically Significant Changes Reported as TEAE in Physical Examination, Vital Signs, 12-lead ECGs, Hematology, Blood Chemistry and Urinalysis
NCT02775240 (33) [back to overview]First-order Rate Constant (Lambda z) Associated With the Terminal (Log-linear) Portion of the Curve of Dextrorphan
NCT02775240 (33) [back to overview]First-order Rate Constant (Lambda z) Associated With the Terminal (Log-linear) Portion of the Curve of Digoxin
NCT02775240 (33) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Dextromethorphan
NCT02775240 (33) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Dextrorphan
NCT02775240 (33) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Digoxin
NCT02775240 (33) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Maribavir
NCT02775240 (33) [back to overview]Parent/Metabolite Ratio of Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) for Dextromethorphan Over AUClast for Dextrorphan (AUClast Parent/Metabolite Ratio)
NCT02775240 (33) [back to overview]Pre-dose Concentration (C0) of Maribavir
NCT02775240 (33) [back to overview]Terminal Half-life (t1/2) of Dextrorphan
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Dextromethorphan
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Omeprazole and Its Metabolite 5-Hydroxyomeprazole
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Omeprazole and Its Metabolite 5-Hydroxyomeprazole
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of Cytochrome P450 (CYP450) Substrate-Midazolam
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Warfarin
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Dextromethorphan
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Caffeine
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Warfarin
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Midazolam
NCT02993471 (10) [back to overview]Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to 48 Hours (AUC[0-48h]) of CYP450 Substrate-Caffeine
NCT03415243 (29) [back to overview]Small Intestine Transit Time
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 90 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 75 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 60 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 120 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 45 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 30 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 240 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 180 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 15 Minutes of Administration
NCT03415243 (29) [back to overview]Percentage of Radiolabeled Drug Remaining in the Stomach After 105 Minutes of Administration
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 180 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 15 Minutes
NCT03415243 (29) [back to overview]Mean Time to Onset of Gastric Emptying
NCT03415243 (29) [back to overview]Mean Time to Complete Gastric Emptying
NCT03415243 (29) [back to overview]Mean Time for Gastric Emptying by Measuring 90 Percent Values
NCT03415243 (29) [back to overview]Mean Time for Gastric Emptying by Measuring 50 Percent Values
NCT03415243 (29) [back to overview]Mean Time for Gastric Emptying by Measuring 25 Percent Values
NCT03415243 (29) [back to overview]Gastric Emptying Half-Life
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 90 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 75 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 60 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 45 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 120 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 105 Minutes
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 240 Minutes
NCT03415243 (29) [back to overview]Total Area Under the Gastric Emptying Curve
NCT03415243 (29) [back to overview]Area Under the Gastric Emptying Curve From Time 0 to 30 Minutes
NCT03415243 (29) [back to overview]Number of Participants With Clinically Significant Change in Laboratory Test Values
NCT03471767 (7) [back to overview]Percentage of Participants Who Experienced a More Than 50% Reduction in Expired Carbon Monoxide (CO) Levels
NCT03471767 (7) [back to overview]Urinary Levels of Dextromethorphan
NCT03471767 (7) [back to overview]Change in Smoking Behavior
NCT03471767 (7) [back to overview]Change in Smoking Intensity
NCT03471767 (7) [back to overview]Medication Adherence
NCT03471767 (7) [back to overview]Medication Tolerance by Self-Reported Side Effects
NCT03471767 (7) [back to overview]Medication Tolerance by Serious Adverse Events
NCT03480009 (4) [back to overview]Mean Pain Scores Via Numeric Rating Scale (NRS-11)
NCT03480009 (4) [back to overview]Analgesic Usage During Medication Abortion
NCT03480009 (4) [back to overview]Analgesic Usage During Medication Abortion
NCT03480009 (4) [back to overview]Worst Pain Measurement Via Numeric Rating Scale (NRS-11)
NCT03538054 (2) [back to overview]Daily Self-reported Physical Activity
NCT03538054 (2) [back to overview]Daily Self-reported Pain Severity
NCT03714919 (5) [back to overview]Time in PACU
NCT03714919 (5) [back to overview]Number of Participants With Sedation, Nausea/Vomiting, or Hallucinations
NCT03714919 (5) [back to overview]Extubation Time
NCT03714919 (5) [back to overview]End of Surgery to Hospital Discharge
NCT03714919 (5) [back to overview]Average Pain Score
NCT03883581 (5) [back to overview]Change in Speech Intelligibility
NCT03883581 (5) [back to overview]Change in Patient-reported Outcome: Center for Neurologic Study-Bulbar Function Scale (CNS-BFS)
NCT03883581 (5) [back to overview]Change in Dynamic Imaging Grade of Swallowing Toxicity
NCT03883581 (5) [back to overview]Change in ALSFRS-R Bulbar Subscale Score
NCT03883581 (5) [back to overview]Bamboo Passage Reading Duration (in Seconds)
NCT04039022 (1) [back to overview]Incidence of Treatment-emergent AEs (TEAEs) Following Dosing With AXS-05
NCT04392011 (7) [back to overview]Midazolam and Dextromethorphan Half-life
NCT04392011 (7) [back to overview]Mitragynine Area Under the Concentration vs. Time Curve (AUC)
NCT04392011 (7) [back to overview]Mitragynine Cmax
NCT04392011 (7) [back to overview]Mitragynine Half Life
NCT04392011 (7) [back to overview]Midazolam and Dextromethorphan Cmax
NCT04392011 (7) [back to overview]Dextromethorphan Area Under the Concentration vs. Time Curve (AUC)
NCT04392011 (7) [back to overview]Midazolam Area Under the Concentration vs. Time Curve (AUC)

Change in CYP1A2 Drug Metabolism Phenotype With Age

Concentrations of caffeine metabolites 5-Acetylamino-6-amino-3-methyluracil (AAMU), 1-methylxanthine (1MX), 1-methyluric acid (1MU), and 1,7-dimethyluric acid (17MU) are quantified in urine and used to estimate the activity of cytochrome P450 1A2 using the well established (AAMU+1MX+1MU)/1,7U ratio. The longitudinal study design allows for changes in drug metabolism activity as a function of age which can be characterized via least squares regression where the slope of age vs. (AAMU+1MX+1MU)/1,7U ratio is examined for deviations from zero. (NCT00117715)
Timeframe: every 6 months for 5 years

Interventionunitless ratio (Mean)
Log ((AAMU+1MX+1MU)/1,7U) Milestone 1Log ((AAMU+1MX+1MU)/1,7U) Milestone 2Log ((AAMU+1MX+1MU)/1,7U) Milestone 3Log ((AAMU+1MX+1MU)/1,7U) Milestone 4Log ((AAMU+1MX+1MU)/1,7U) Milestone 5Log ((AAMU+1MX+1MU)/1,7U) Milestone 6Log ((AAMU+1MX+1MU)/1,7U) Milestone 7Log ((AAMU+1MX+1MU)/1,7U) Milestone 8Log ((AAMU+1MX+1MU)/1,7U) Milestone 9
Longitudinal Assessment Cohort-0.851-0.823-0.858-0.856-0.832-0.853-0.822-0.851-0.821

[back to top]

Change in CYP3A4 Drug Metabolism Phenotype With Age

Concentrations of dextromethorphan (DM) metabolites 3-hydroxymorphinan (3HM) and dextrorphan (DX) are quantified in urine and used to estimate the activity of cytochrome P450 3A4 using the well established 3HM/DX ratio. The longitudinal study design allows for changes in drug metabolism activity as a function of age which can be characterized via least squares regression where the slope of age vs. 3HM/DX ratio is examined for deviations from zero. (NCT00117715)
Timeframe: every 6 months for 5 years

Interventionunitless ratio (Mean)
Log (3HM/DX) Milestone 1Log (3HM/DX) Milestone 2Log (3HM/DX) Milestone 3Log (3HM/DX) Milestone 4Log (3HM/DX) Milestone 5Log (3HM/DX) Milestone 6Log (3HM/DX) Milestone 7Log (3HM/DX) Milestone 8Log (3HM/DX) Milestone 9
Longitudinal Assessment Cohort-0.069-0.117-0.142-0.158-0.109-0.136-0.133-0.122-0.169

[back to top]

Change in CYP2D6 Drug Metabolism Phenotype With Age

Concentrations of dextromethorphan(DM) and it's metabolite dextrorphan (DX) are quantified in urine and used to estimate the activity of cytochromes P450 2D6 using the well established DM/DX ratio. The longitudinal study design allows for changes in drug metabolism activity as a function of age which can be characterized via least squares regression where the slope of age vs. DM/DX ratio is examined for deviations from zero. (NCT00117715)
Timeframe: every 6 months for 5 years

Interventionunitless ratio (Mean)
Log (DM/DX) Milestone 1Log (DM/DX) Milestone 2Log (DM/DX) Milestone 3Log (DM/DX) Milestone 4Log (DM/DX) Milestone 5Log (DM/DX) Milestone 6Log (DM/DX) Milestone 7Log (DM/DX) Milestone 8Log (DM/DX) Milestone 9
Longitudinal Assessment Cohort-1.974-2.083-2.162-2.073-2.161-2.185-2.284-2.177-2.384

[back to top]

AUC0-t Simvastatin Acid

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for simvastatin acid (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Simvastatin 20 mg + Lomitapide 10 mg138.76

[back to top]

AUC0-t Nicotinic Acid

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for nicotinic acid (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
ER Niacin 1000 mg + Lomitapide 10 mg110.22

[back to top]

AUC0-t Nicotinuric Acid

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for nicotinuric acid (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
ER Niacin 1000 mg + Lomitapide 10 mg79.15

[back to top]

AUC0-t Rosuvastatin (Lomitapide 10 mg)

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for rosuvastatin (Lomitapide 10 mg) (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Rosuvastatin 20 mg + Lomitapide 10 mg102.05

[back to top]

AUC0-t Rosuvastatin (Lomitapide 60 mg)

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for rosuvastatin (Lomitapide 60 mg) (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Rosuvastatin 20 mg + Lomitapide 60 mg132.21

[back to top]

Percent Change From Baseline in Low-density Lipoprotein Cholesterol (LDL-C)

Percent change from Baseline in LDL-C (NCT00359281)
Timeframe: Baseline to Day 8

InterventionPercent Change (Mean)
Atorvastatin 20 mg + Lomitapide 10 mg-30.99
Simvastatin 20 mg + Lomitapide 10 mg-26.43
Ezetimibe 10 mg + Lomitapide 10 mg-28.36
Rosuvastatin 20 mg + Lomitapide 10 mg-41.74
Fenofibrate 145 mg + Lomitapide 10 mg-20.12
Atorvastatin 20 mg + Lomitapide 60 mg-66.02
Rosuvastatin 20 mg + Lomitapide 60 mg-63.20
Dextrometh-rophan 30 mg + Lomitapide 60 mg-46.07
ER Niacin 1000 mg + Lomitapide 10 mg-20.89

[back to top]

AUC0-t Total Ezetimibe

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for total ezetimibe (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Ezetimibe 10 mg + Lomitapide 10 mg105.71

[back to top]

AUC0-t Atorvastatin Acid (Lomitapide 60 mg)

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for atorvastatin acid (Lomitapide 60 mg) (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Atorvastatin 20 mg + Lomitapide 60 mg152.32

[back to top]

AUC0-t Fenofibric Acid

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for fenofibric acid (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Fenofibrate 145 mg + Lomitapide 10 mg89.62

[back to top]

AUC0-t Simvastatin

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for simvastatin (NCT00359281)
Timeframe: 0 to 24 hours

InterventionRatio (Geometric Mean)
Simvastatin 20 mg + Lomitapide 10 mg162.25

[back to top]

Area Under Concentration-time Curve From 0 to Last Measureable Concentration (AUC0-t) Atorvastatin Acid (Lomitapide 10 mg)

Geometric Mean Ratio ln(AUC0-t) Day 8/Day 1 for atorvastatin acid (Lomitapide 10 mg) (NCT00359281)
Timeframe: 0 to 24 hour

InterventionRatio (Geometric Mean)
Atorvastatin 20 mg + Lomitapide 10 mg110.97

[back to top]

SNOT-16 Score (Sino-Nasal Outcomes Test) at Day 3

The Sino-Nasal Outcomes Test (SNOT-16) assesses disease-specific quality of life for acute and chronic rhinosinusitis. This brief instrument assesses 16 sinus-related symptoms and was administered by phone. The respondent reported how much they were bothered by each item considering both its severity and frequency. Response options include no problem (0), mild or slight problem (1), moderate problem (2), severe problem (3). The SNOT-16 score is the mean score from all 16 items and ranges from 0 (minimal impact) to 3 (significant impact). (NCT00377403)
Timeframe: 4 days

InterventionUnits on a scale (Mean)
Intervention Arm1.12
Symptomatic Treatments Only1.14

[back to top]

Stage 1A: MTD of Cobimetinib in 14/14 Schedule

"AEs were graded according to NCI-CTCAE v3.0. A DLT was the basis for determining MTD in Stage 1A participants. The participants of Stage 1A are dose-escalation cohorts, starting at the MTD of the 21/7 schedule, were treated on a 14/14 schedule to determine the MTD. A DLT was defined as either of the following occurring during the Study Treatment Period:~Occurrence of a drug-related AE that, in the opinion of the CRC, was of potential clinical significance such that further dose escalation would expose participants to risk of irreversible medical harm; Nonhematologic toxicity: Grade 3 or 4 events, including Grade 3 nausea and/or vomiting and/or Grade 3 diarrhea, despite prophylaxis and/or treatment; Hematologic toxicity: Grade 4 thrombocytopenia. Grade 4 neutropenia of more than 4 days' duration; Grade 4 neutropenia of any duration with fever or documented infection. AEs (Grade 3 or higher) for which a clinical cause unrelated to cobimetinib was evident was not considered DLTs." (NCT00467779)
Timeframe: Stage 1A: Days 1 to 28 of Cycle 1

Interventionmg (Number)
Stage 1A - All Cohorts (14/14)100

[back to top]

Stage 1A: Cmax of Cobimetinib at Steady State

Cmax is defined as the maximum plasma concentration achieved after administration of cobimetinib in Stage 1A and was measured in steady state as ng/mL. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

Interventionng/mL (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)180.0
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)494.0
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)640.0
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)1160.0

[back to top]

Stage 1A: Cmax of Cobimetinib at Cycle 1 Day 1

Cmax is defined as the maximum plasma concentration achieved after administration of cobimetinib on on Day 1 in Stage 1A and was measured as ng/mL. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionng/mL (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)78.4
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)167.0
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)258.0
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)533.0

[back to top]

Stage 1A: AUC 0-24 of Cobimetinib at Cycle 1 Day 1

AUC0-24 for stage 1A was calculated on Day 1 with the measured data points from the time of administration of cobimetinib up to 24 h after administration by the trapezoidal formula. AUC 0-24 is the truncated AUC over a 24-hour sampling interval. The concentration-time curve is the result of time points of blood sampling and its measured concentration of free cobimetinib in the blood samplings. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionh*ng/mL (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)1140
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)2130
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)4020
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)7190

[back to top]

Stage 1A: Apparent Clearance of Cobimetinib at Steady State

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the blood. Apparent clearance was calculated only for participants who had a quantifiable AUC 0-24 in steady state. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

InterventionL/hr (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)25.1
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)14.9
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)21.9
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)6.9

[back to top]

Stage 1A: AUC 0-24/D of Cobimetinib at Steady State

AUC 0-24/D is the dose normalized truncated AUC over a 24-hour sampling interval. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

Interventionng*hr/mL/mg (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)49.8
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)84.2
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)115.0
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)172.0

[back to top]

Stage 1A: AUC 0-24 of Cobimetinib at Steady State

The area under the AUC0-24 for steady state in stage 1A was calculated with the measured data points from the time of administration of cobimetinib up to 24 h after administration by the trapezoidal formula. AUC 0-24 is the truncated AUC over a 24-hour sampling interval. The concentration-time curve is the result of time points of blood sampling and its measured concentration of free cobimetinib in the blood samplings. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

Interventionh*ng/mL (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)2990.0
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)6740.0
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)11500.0
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)21500.0

[back to top]

Stage 1A: Accumulation Ratio of Cobimetinib at Steady State

Accumulation Ratio: AUC0-24 at steady state divided by AUC0-24 on Cycle 1 Day 1. It was calculated only for participants who had a quantifiable AUC 0-24 at steady state. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

Interventionratio (Mean)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)2.32
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)3.14
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)2.6
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)5.15

[back to top]

Stage 1: Tmax of Cobimetinib at Steady State

Tmax is defined as the time to reach Cmax during stage 1 in steady state. Steady state was reached when overall intake of cobimetinib was in dynamic equilibrium with its elimination. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

Interventionhours (Median)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)4.0
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)1.0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)2.25
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)4.0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)3.0
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)2.0
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)3.0
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)2.5

[back to top]

Stage 1: Time to Maximum Concentration (Tmax) of Cobimetinib at Day 1, Cycle 1

Tmax is defined as the time to reach Cmax during stage 1 at Day 1 Cycle 1. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 1 and pre-dose on Cycle 1 Day 2

Interventionhours (Median)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)2.0
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)1.0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)1.5
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)3.0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)4.0
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)2.5
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)2.0
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)2.0

[back to top]

Stage 1: Maximum Tolerated Dose (MTD) of Cobimetinib in 21/7 Schedule

"AEs were graded according to the NCI-CTCAE v3.0. A DLT was determined from clinical findings during the Study Treatment Period (Cycle 1, Days 1). MTD was defined as the dose at which no DLTs were observed. DLT was defined as either of the following occurring during the Study Treatment Period. The occurrence of a drug-related AE that, in the opinion of the CRC, was of potential clinical significance such that further dose escalation would expose participants in higher dose cohorts to risk of irreversible medical harm or require medical treatment to avoid irreversible medical harm or non-hematologic toxicity~Grade 3 or 4 events, including Grade 3 nausea and/or vomiting and/or Grade 3 diarrhea, despite prophylaxis and/or treatment Hematologic toxicity~Grade 4 thrombocytopenia~Grade 4 neutropenia of greater than or equal to (≥) 4 days' duration~Grade 4 neutropenia of any duration with fever or documented infection" (NCT00467779)
Timeframe: Stage 1: Days 1 to 28 of Cycle 1

Interventionmilligrams (mg) (Number)
Stage 1 All Cohorts (21/7)60

[back to top]

Stage 1: Maximum Observed Concentration (Cmax) of Cobimetinib at Day 1, Cycle 1

Cmax is defined as the maximum plasma concentration achieved after administration of cobimetinib on Day 1, Cycle 1 in Stage 1 and was measured as nanograms per milliliter (ng/mL). (NCT00467779)
Timeframe: Stage 1: Pre-dose and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 1 and pre-dose on Cycle 1 Day 2

Interventionng/mL (Geometric Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)4.51
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)6.92
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)18.3
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)18.8
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)30.8
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)71.7
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)163.0
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)261.0

[back to top]

Stage 1: Half-Life (t1/2) of Cobimetinib at Steady State

t1/2 is the half-life of cobimetinib measured over the terminal phase by noncompartmental analysis in stage 1 in steady state. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

Interventionhours (Median)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)80.0
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)64.0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)47.8
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)66.0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)50.5
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)41.3
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)51.3
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)60.0

[back to top]

Stage 1: Cmax of Cobimetinib at Steady State

Cmax is defined as the maximum plasma concentration achieved after administration of cobimetinib in Stage 1 and was measured at steady state in ng/mL. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

Interventionng/mL (Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)13.5
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)19.8
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)28.7
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)53.0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)54.7
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)341.0
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)401.0
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)641.0

[back to top]

Stage 1: AUC 0-24/D of Cobimetinib at Steady State

AUC 0-24/D is the dose normalized truncated AUC over a 24-hour sampling interval. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

Interventionng*hr/mL/mg (Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)62.4
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)34
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)28.2
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)76.3
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)44.3
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)134
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)104
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)132

[back to top]

Stage 1: AUC 0-24 of Cobimetinib at Steady State

The area under the AUC0-24 for steady state in stage 1 was calculated with the measured data points from the time of administration of cobimetinib up to 24 h after administration by the trapezoidal formula. AUC 0-24 is the truncated AUC over a 24-hour sampling interval. The concentration-time curve is the result of time points of blood sampling and its measured concentration of free cobimetinib in the blood samplings. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

Interventionh*ng/mL (Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)202.0
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)288.0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)411.0
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)763.0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)886.0
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)5370.0
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)6250.0
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)10500.0

[back to top]

Stage 1: Area Under the Plasma Cobimetinib Concentration Curve From Time 0 to 24 Hours (AUC 0-24) Day 1, Cycle 1

The area under the concentrations-time curve (AUC0-24) was calculated with the measured data points from the time of administration of cobimetinib up to 24 h after administration by the trapezoidal formula. AUC 0-24 is the truncated AUC over a 24-hour sampling interval. The concentration-time curve is the result of time points of blood sampling and its measured concentration of free cobimetinib in the blood samples. AUC is measured as hours times nanograms per milliliter (h*ng/mL). (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionh*ng/mL (Geometric Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)56.2
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)68.0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)203.0
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)242.0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)440.0
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)785.0
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)1620.0
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)3060.0

[back to top]

Stage 1: Apparent Clearance of Cobimetinib at Steady State

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the blood. Apparent clearance was calculated only for participants who had a quantifiable AUC 0-24 in steady state. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

InterventionLiters per hour (L/hr) (Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)22.4
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)34.0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)37.1
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)13.5
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)22.6
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)14.0
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)11.8
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)11.6

[back to top]

Stage 1: Accumulation Ratio of Cobimetinib at Steady State

Accumulation Ratio: AUC0-24 at steady state divided by AUC0-24 on Cycle 1 Day 1. It was calculated only for participants who had a quantifiable AUC 0-24 at steady state. (NCT00467779)
Timeframe: Stage 1: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12-18 hours post-dose on Cycle 1 Day 1, Day 21, 24, 48, and 72 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22, 23, and 24, respectively)

Interventionratio (Mean)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)3.56
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)3.66
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)2.65
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)3.23
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)2.96
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)3.61
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)2.87
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)3.26

[back to top]

Stage 1 and 1A: Number of Participants With Dose Limiting Toxicities (DLTs)

"Adverse events (AE) were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) v3.0. DLT was defined as either of the following occurring during the Study Treatment Period. The occurrence of a drug-related AE that, in the opinion of the cohort review committee (CRC), was of potential clinical significance such that further dose escalation would expose participants in higher dose cohorts to risk of irreversible medical harm or require medical treatment to avoid irreversible medical harm or non-hematologic toxicity~Grade 3 or 4 events, including Grade 3 nausea and/or vomiting and/or Grade 3 diarrhea, despite prophylaxis and/or treatment Hematologic toxicity~Grade 4 thrombocytopenia~Grade 4 neutropenia of greater than or equal to (≥) 4 days' duration~Grade 4 neutropenia of any duration with fever or documented infection" (NCT00467779)
Timeframe: Stage 1 and 1A: Days 1 to 28 of Cycle 1

Interventionparticipants (Number)
Stage 1 Cohort 01 - Cobimetinib 0.05 mg/kg (21/7)0
Stage 1 Cohort 02 - Cobimetinib 0.10 mg/kg (21/7)0
Stage 1 Cohort 03 - Cobimetinib 0.20 mg/kg (21/7)0
Stage 1 Cohort 04 - Cobimetinib 10 mg (21/7)0
Stage 1 Cohort 05 - Cobimetinib 20 mg (21/7)0
Stage 1 Cohort 06 - Cobimetinib 40 mg (21/7)1
Stage 1 Cohort 07 - Cobimetinib 60 mg (21/7)1
Stage 1 Cohort 08 - Cobimetinib 80 mg (21/7)2
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)0
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)0
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)0
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)2

[back to top]

Stage 2A: Accumulation Ratio of Cobimetinib at Steady State

Accumulation Ratio AUC0-24 is ratio of AUC on Day 20: Day 1. (NCT00467779)
Timeframe: Stage 2A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15), and between Cycle 1 Days 26-28

Interventionratio (Mean)
Stage 2A Cohort 30 - Cobimetinib 100 mg (Expansion) (14/14)2.3

[back to top]

Stage 2A: Tmax of Cobimetinib at Steady State

Tmax is defined as the time to reach Cmax during stage 2A in steady state. (NCT00467779)
Timeframe: Stage 2A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15), and between Cycle 1 Days 26-28

Interventionhours (Median)
Stage 2A Cohort 30 - Cobimetinib 100 mg (Expansion) (14/14)3.0

[back to top]

Stage 2: Accumulation Ratio of Cobimetinib at Steady State

Accumulation ratio is AUC0-24 at steady state divided by AUC0-24 on Cycle 1 Day 1. It was calculated only for participants who had a quantifiable AUC 0-24 at steady state. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 21, 24 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22), and between Cycle 1 Days 26-28

Interventionratio (Geometric Mean)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)2.5

[back to top]

Stage 2: Tmax of Cobimetinib at Cycle 1 Day 1

Tmax is defined as the time to reach Cmax during stage 2 on Day 1. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionhours (Median)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)3.0

[back to top]

Stage 2: Tmax of Cobimetinib at Steady State

Tmax is defined as the time to reach Cmax during stage 2 in steady state. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 21, 24 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22), and between Cycle 1 Days 26-28

Interventionhours (Median)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)4.0

[back to top]

Stage 2:AUC 0-24 of Cobimetinib at Cycle 1 Day 1

The area under the AUC0-24 on Day 1 in stage 2 was calculated with the measured data points from the time of administration of cobimetinib up to 24 h after administration by the trapezoidal formula. AUC 0-24 is the truncated AUC over a 24-hour sampling interval. The concentration-time curve is the result of time points of blood sampling and its measured concentration of free cobimetinib in the blood samplings. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionh*ng/mL (Geometric Mean)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)3060.0

[back to top]

Stage 2:Half-Life of Cobimetinib at Steady State

T1/2 half-life of cobimetinib measured over the terminal phase by noncompartmental analysis. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 21, 24 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22), and between Cycle 1 Days 26-28

Interventionhours (Median)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)53.4

[back to top]

Stage III: AUC 0-24 of Dextromethorphan

AUC0-24 is the area under the plasma drug concentration curve over a 24-hour sampling interval and was determined both in presence (Cycle 1 Day 15) and absence (Cycle 1 Day 1) of cobimetinib. (NCT00467779)
Timeframe: Stage III: Predose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours after dextromethorphan administration on Days 1 and 15 of Cycle 1

Interventionh*ng/mL (Geometric Mean)
With cobimetinib - Cycle 1 Day 1 (n=17)Without cobimetinib - Cycle 1 Day1 (n=19)
Stage 3 Cohort 40 - Cobimetinib+Midazolam+Dextromethorphan24.825.8

[back to top]

Stage 2A: Apparent Clearance of Cobimetinib at Steady State

Apparent clearance is the plasma clearance of absorbed drug. (NCT00467779)
Timeframe: Stage 2A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15), and between Cycle 1 Days 26-28

InterventionL/hr (Mean)
Stage 2A Cohort 30 - Cobimetinib 100 mg (Expansion) (14/14)11.8

[back to top]

Stage 2A: AUC 0-24/D of Cobimetinib at Steady State

AUC 0-24/D is the dose normalized truncated AUC over a 24-hour sampling interval. (NCT00467779)
Timeframe: Stage 2A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15), and between Cycle 1 Days 26-28

Interventionh*ng/mL (Mean)
Stage 2A Cohort 30 - Cobimetinib 100 mg (Expansion) (14/14)84.8

[back to top]

Stage 2A: Cmax of Cobimetinib at Steady State

Cmax is the maximum plasma concentration achieved following the Day 20 dose in Stage 2A. (NCT00467779)
Timeframe: Stage 2A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15), and between Cycle 1 Days 26-28

Interventionng/mL (Geometric Mean)
Stage 2A Cohort 30 - Cobimetinib 100 mg (Expansion) (14/14)315.0

[back to top]

Stage 1A: Tmax of Cobimetinib at Steady State

Tmax is defined as the time to reach Cmax during stage 1A in steady state. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

Interventionhours (Median)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)2.0
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)2.0
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)3
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)6

[back to top]

Stage 2A: Half-Life of Cobimetinib at Steady State

(NCT00467779)
Timeframe: Stage 2A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15), and between Cycle 1 Days 26-28

Interventionhours (Median)
Stage 2A Cohort 30 - Cobimetinib 100 mg (Expansion) (14/14)42.7

[back to top]

Stage III: Cmax of Dextromethorphan

Cmax is defined as maximum observed plasma concentration and was determined both in the presence (Cycle 1 Day 15) and absence of cobimetinib (Cycle 1 Day 1). (NCT00467779)
Timeframe: Stage III: Predose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours after dextromethorphan administration on Days 1 and 15 of Cycle 1

Interventionng/mL (Geometric Mean)
With cobimetinib - Cycle 1 Day 15(n=17)Without cobimetinib - Cycle 1 Day 1 (n=20)
Stage 3 Cohort 40 - Cobimetinib+Midazolam+Dextromethorphan3.443.16

[back to top]

Stage III: AUC0-inf of Midazolam

AUC0-inf is the AUC from time 0 to infinity and was calculated both in the presence (Cycle 1 Day 15) and absence (Cycle 1 Day 1) of cobimetinib. (NCT00467779)
Timeframe: Stage III: Predose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours after dextromethorphan administration on Days 1 and 15 of Cycle 1

Interventionh*ng/mL (Geometric Mean)
With cobimetinib - Cycle 1 Day 15 (n=17)Without cobimetinib Cycle 1 Day 1 (n=19)
Stage 3 Cohort 40 - Cobimetinib+Midazolam+Dextromethorphan34.935.7

[back to top]

Stage III: AUC0-24 of Midazolam

AUC0-24 is the area under the plasma drug concentration curve over a 24-hour sampling interval and was calculated both in the presence (Cycle 1 Day 15) and absence (CXycle 1 Day 1) of cobimetinib. (NCT00467779)
Timeframe: Stage III: Predose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours after dextromethorphan administration on Days 1 and 15 of Cycle 1

Interventionh*ng/mL (Geometric Mean)
With cobimetinib - Cycle 1 Day 15 (n=14)Without cobimetinib - Cycle 1 Day 1 (n=11)
Stage 3 Cohort 40 - Cobimetinib+Midazolam+Dextromethorphan33.033.4

[back to top]

Stage III: AUC 0-inf of Dextromethorphan

AUC0-inf is AUC from time 0 to infinity and was calculated both in presence Cycle 1 Day 15) and absence (Cycle 1 Day 1) of cobimetinib. (NCT00467779)
Timeframe: Stage III: Predose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours after dextromethorphan administration on Days 1 and 15 of Cycle 1

Interventionh*ng/mL (Geometric Mean)
With cobimetinib - Cycle 1 Day 15 (n=13)Without cobimetinib - Cycle 1 Day1 (n=19)
Stage 3 Cohort 40 - Cobimetinib+Midazolam+Dextromethorphan29.118.9

[back to top]

Stage III: Cmax of Midazolam

Cmax is the maximum observed plasma concentration and was calculated both in the presence (Cycle 1 Day 15) and absence (Cycle 1 Day 1) of cobimetinib. (NCT00467779)
Timeframe: Stage III: Predose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours after dextromethorphan administration on Days 1 and 15 of Cycle 1

Interventionng/mL (Geometric Mean)
With cobimetinib - Cycle 1 Day15 (n=17)Without cobimetinib - Cycle 1 Day 1(n=20)
Stage 3 Cohort 40 - Cobimetinib+Midazolam+Dextromethorphan11.510.9

[back to top]

Stage 2: Apparent Clearance of Cobimetinib at Steady State

Clearance of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. Drug clearance is a quantitative measure of the rate at which a drug substance is removed from the blood. Apparent clearance was calculated only for participants who had a quantifiable AUC 0-24 in steady state. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 21, 24 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22), and between Cycle 1 Days 26-28

InterventionL/hr (Mean)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)9.8

[back to top]

Stage 2: AUC 0-24 of Cobimetinib at Steady State

The area under the AUC0-24 for steady state in stage 2 was calculated with the measured data points from the time of administration of cobimetinib up to 24 h after administration by the trapezoidal formula. AUC 0-24 is the truncated AUC over a 24-hour sampling interval. The concentration-time curve is the result of time points of blood sampling and its measured concentration of free cobimetinib in the blood samplings. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 21, 24 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22), and between Cycle 1 Days 26-28

Interventionh*ng/mL (Geometric Mean)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)10200.0

[back to top]

Stage 2: AUC 0-24/D of Cobimetinib at Steady State

AUC 0-24/D is the dose normalized truncated AUC over a 24-hour sampling interval. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 21, 24 hours post Cycle 1 Day 21 dose (Cycle 1 Day 22), and between Cycle 1 Days 26-28

Interventionh*ng/mL (Geometric Mean)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)102.0

[back to top]

Stage 2: Cmax of Cobimetinib at Cycle 1 Day 1

Cmax is defined as the maximum plasma concentration achieved after administration of cobimetinib in Stage 2 and was measured in ng/mL. (NCT00467779)
Timeframe: Stage 2: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionng/mL (Geometric Mean)
Stage 2 Cohort 20 - Cobimetinib 60 mg (Expansion) (21/7)184.0

[back to top]

Stage 1A: Tmax of Cobimetinib at Cycle 1 Day 1

Tmax is defined as the time to reach Cmax during stage 1A at Day 1. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 1, pre-dose on Cycle 1 Day 2

Interventionhours (Median)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)3.0
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)4.0
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)3.5
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)2.5

[back to top]

Stage 1A: t1/2 of Cobimetinib at Steady State

t1/2 is the half-life of cobimetinib measured over the terminal phase by noncompartmental analysis in stage 1A in steady state. (NCT00467779)
Timeframe: Stage 1A: Pre-dose & 0.5, 1, 1.5, 2, 3, 4, 6 hours post-dose on Cycle 1 Day 14, 24, 48, 72 hours post Cycle 1 Day 14 dose (Cycle 1 Day 15, 16, and 17, respectively)

Interventionhours (Median)
Stage 1A Cohort 01A - Cobimetinib 60 mg (14/14)59.4
Stage 1A Cohort 02A - Cobimetinib 80 mg (14/14)44.1
Stage 1A Cohort 03A - Cobimetinib 100 mg (14/14)51.6
Stage 1A Cohort 04A - Cobimetinib 125 mg (14/14)47.7

[back to top]

Mean Change From Baseline at Day 84 in Beck Depression Inventory (BDI-II) Total Score

The BDI-II is a 21-item self report instrument intended to assess the existence and severity of symptoms of depression, summed to give a single score. The BDI-II uses a 4-point for each item ranging from 0 to 3. A total score of 0-13 is considered minimal range, 14 to 19 is mild, 20 to 28 is moderate, and 29 to 63 is severe. (NCT00573443)
Timeframe: Baseline and Day 84

InterventionScores on a Scale (Mean)
AVP-923-30-1.59
AVP-923-20-1.03
Placebo-0.02

[back to top]

Mean Change From Baseline to Day 84 in Pain Rating Scale (PRS) of MS Subjects

Subjects with MS were instructed to also record daily the pain they experienced using the PRS. After evaluating the subject's ability to comply with these requirements, the investigator determined if a caregiver should complete the study diary and assessments. Subjects rated their pain over the past 12 hours on a scale of 0 to 10 (0=none, 10=worst pain ever experienced). (NCT00573443)
Timeframe: Baseline, Day 15, Day 29, Day 57, Day 84

InterventionScores on a Scale (Mean)
AVP-923-30-1.0
AVP-923-20-0.7
Placebo-0.4

[back to top]

Mean Change From Baseline to Day 84 in Neuropsychiatric Inventory (NPI-Q) Frequency and Severity Score (ITT Population)

The NPI is a retrospective (to 1 month) caregiver-informant interview assessing frequency and severity of 12 neuropsychiatric symptom domains. The NPI score is based on the sum of the severity ratings (0=absent, 1=mild, 3=severe). The 12 symptom domains include delusions, hallucinations, agitation/aggression, dysphoria/depression, anxiety, euphoria/elation, apathy/indifference, disinhibition, irritability/lability, aberrant motor behaviors, nighttime behavioral disturbances, and appetite/eating abnormalities. The NPI severity score is based on severity ratings (0=absent, 1=mild to 3=severe). (NCT00573443)
Timeframe: Baseline to Day 84

,,
InterventionScores on a Scale (Mean)
Frequency ScoreSeverity Score
AVP-923-20-2.56-1.59
AVP-923-30-1.62-0.74
Placebo-1.33-0.98

[back to top]

Mean Change From Baseline to Day 84 in Neuropsychiatric Inventory (NPI-Q) Frequency and Severity Score (EE Population)

The NPI is a retrospective (to 1 month) caregiver-informant interview assessing frequency and severity of 12 neuropsychiatric symptom domains. The NPI score is based on the sum of the severity ratings (0=absent, 1=mild, 3=severe). The 12 symptom domains include delusions, hallucinations, agitation/aggression, dysphoria/depression, anxiety, euphoria/elation, apathy/indifference, disinhibition, irritability/lability, aberrant motor behaviors, nighttime behavioral disturbances, and appetite/eating abnormalities. The NPI severity score is based on severity ratings (0=absent, 1=mild to 3=severe). (NCT00573443)
Timeframe: Baseline to Day 84

,,
InterventionScores on a Scale (Mean)
Frequency ScoreSeverity Score
AVP-923-20-3.09-1.81
AVP-923-30-1.68-0.80
Placebo-1.25-0.91

[back to top]

Mean Change From Baseline in CNS-LS Total Score by Visit

Center for Neurologic Studies-Lability Scale (CNS-LS) is an instrument for the measurement of PBA that has been validated for the use in patients with ALS and MS. It is a 7-item self-report questionnaire that measures the frequency and severity of PBA episodes, including assessments of labile laughter and labile tearfulness,and provides a score for total PBA (total score can range from 7-35). The following 5-point scoring was used: 1=Applies never, 2=Applies rarely, 3=Applies occasionally, 4=Applies frequently, 5=Applies most of the time. A score of 13 or higher may suggest PBA, and the higher the score the more severe the episodes. (NCT00573443)
Timeframe: Baseline, Day 15, Day 29, Day 57, Day 84

,,
InterventionScores on a Scale (Mean)
Day 15 (Visit2)Day 29 (Visit 3)Day 57 (Visit 4)Day 84 (Visit 5)
AVP-923-20-6.27-7.62-8.89-8.24
AVP-923-30-6.77-8.03-8.59-8.17
Placebo-4.58-5.70-5.66-5.72

[back to top]

Mean Change From Baseline at Day 84 in SF-36 (Short-Form) Health Survey Medical Outcome Score by Category

The SF-36 is designed to examine a person's perceived health status. The SF-36 includes one multi-item scale measuring eight health concepts: vitality, physical functioning, bodily pain, general health perceptions, physical role-, emotional role-, social role functioning, and mental health. Answers to each question are scored and summed to produce raw scale scores for each health concept which are then transformed to a 0 - 100 scale, a high score defining a more favorable health state. An aggregate summary measure is calculated by averaging the scores from the eight health concepts. (NCT00573443)
Timeframe: Baseline and Day 84

,,
InterventionScores on a Scale (Mean)
Physical Functioning ScaleRole Physical ScaleBodily Pain ScaleGeneral Health ScaleVitality ScaleSocial Functioning ScaleRole Emotional ScaleMental Health Scale
AVP-923-20-5.30-4.265.84-2.95-5.301.42-1.813.09
AVP-923-30-0.903.474.09-1.47-0.909.3411.555.53
Placebo-4.05-1.75-1.13-1.28-4.05-3.092.36-0.28

[back to top]

PBA Episode Rate Ratio (Post/Pre), Regression Adjusted

Episodes were counted each day and recorded in a daily diary. The outcome measure is the ratio of the episode rate over the 84-day treatment period to the rate during the baseline period, adjusted for study site, and underlying disease using longitudinal negative binomial regression. (NCT00573443)
Timeframe: Baseline to Day 84

InterventionUnit-free (ratio of episodes/week) (Least Squares Mean)
AVP-923-300.247
AVP-923-200.237
Placebo0.465

[back to top]

Difference in EEG Spike Counts at Six Months Compared to Baseline for Each Treatment Arm.

Difference in EEG spike count means pre and 6 months post-treatment in each of three treatment groups. (NCT00593957)
Timeframe: Initial and 6-month post-treatment

InterventionEEG spike counts per minute (Mean)
Dextromethorphan (DM)1 EEG Spike Counts at Baseline44.22
DM2 EEG Spike Counts at Baseline25.44
DM3 EEG Spike Counts at Baseline40.67
DM1 EEG Spike Count at 6 Months38.29
DM2 EEG Spike Counts at 6 Months27.58
DM3 EEG Spike Counts at 6 Months36.01

[back to top]

Mean SSI Score for Total Subjects at Baseline and 6 Months

Analysis of Difference in Mean Screen for Social Interaction (SSI) Score between 0-6 months for total sample (n=19). (NCT00593957)
Timeframe: 0-6 months

Interventionscores on a scale (Mean)
Total Sample SSI Mean Score at Baseline62.737
Total Sample SSI Mean Score at 6 Months68.684

[back to top]

Improvement in Receptive Language as Measured by the Mullen Scale.

The Mullen Receptive language scale pre and 6 months post DM, measured as a change in the mean score of language, by age in months. (NCT00593957)
Timeframe: Change in mean between Initial and 6-month follow-up

Interventionage in months (Mean)
DM1( 0.25 mg/kg /Day)0
DM2 (2.5 mg/kg/Day)-0.44
DM3 (5mg/kg/Day)03

[back to top]

Difference in SSI Mean Score at Six Months Compared to Baseline for Each Treatment Arm.

"The Screen for Social Interaction (SSI) is a 54-item parent/caregiver-report screening instrument that emphasizes reciprocal social interaction including joint attention skills. The items are positive (prosocial) and are scored on a four-point frequency scale (child displays the behavior almost never = 0 to almost all the time = 3). Thus lower scores reflect a slower or delayed development, and higher scores reflect more normative development. SSI total scores range from 0-162. There are no subscales. Difference in Screen for Social Interaction (SSI) mean scores between baseline and 6 months post-treatment for each treatment arm are reported." (NCT00593957)
Timeframe: Initial and 6 month followup

Interventionscores on a scale (Mean)
DM1( 0.25 mg/kg /Day) SSI Baseline40.25
DM2 (2.5 mg/kg/Day)SSI Baseline70.00
DM3 (5mg/kg/Day) SSI Baseline84.29
DM1( 0.25 mg/kg /Day) SSI 6 Months48.63
DM2 (2.5 mg/kg/Day) SSI 6 Months74.50
DM3 (5.0 mg/kg/Day) SSI 6 Months88.29

[back to top]

Plasma AUC Ratio of Day 1 and Day 8

"Assessment of the drug-drug interactions of Proellex® (CDB-4124) with cytochrome P450 isoenzymes CYP1A2, 2C9, 2C19, 2D6, and 3A4 in healthy female subjects administered 50 mg Proellex® once daily (QD). The Day 8 AUC was compared to the Day 1 AUC to determine inhibition.~For CYP1A2 the plasma paraxanthine/caffeine MR ratio (metabolic ratio) was used. For CYP2D6 the MR ratio of dextromethorphan/dextrorphan was used." (NCT00741468)
Timeframe: 8 days

InterventionRatio of geometric means Day 8 to Day 1 (Mean)
CYP1A21.093
CYP2C91.029
CYP2C191.104
CYP2D61.914
CYP3A42.245

[back to top]

Median of 3 Month Tamoxifen Steady State Plasma Concentrations According to CYP2D6 Phenotype Group

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 3 month Tamoxifen steady state plasma concentrations for each CYP2D6 phenotype group are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 3 Month

InterventionnM (Median)
PM/PMIM/PMIM/IMPM/EMEM/IMEM/EMEM/UM
¹³C-DM-BT219.6233.8272.3257.7279.1201.0313.3

[back to top]

Median of 3 Month N-desmethyl-tamoxifen (NDMT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 3 month NDMT steady state plasma concentrations for each CYP2D6 phenotype group are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 3 Month

InterventionnM (Median)
PM/PMIM/PMIM/IMPM/EMEM/IMEM/EMEM/UM
¹³C-DM-BT633.3578.0691.6643.0630.7457.8576.9

[back to top]

Median of 3 Month Endoxifen Steady State Concentrations (Endx Css) According to CYP2D6 Phenotype Group and Activity Score

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 3 month Endx Css for each CYP2D6 phenotype group and the corresponding activity score are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 3 Month

InterventionnM (Median)
EM/UM (3.0)EM/EM (2.0)EM/IM (1.5)IM/IM (1.0)EM/PM (1.0)IM/PM (0.5)PM/PM (0)
¹³C-DM-BT23.630.527.911.521.56.15.5

[back to top]

Median of 3 Month 4-hydroxy-tamoxifen (4HT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 3 month 4HT steady state plasma concentrations for each CYP2D6 phenotype group are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 3 Month

InterventionnM (Median)
PM/PMIM/PMIM/IMPM/EMEM/IMEM/EMEM/UM
¹³C-DM-BT3.131.53.523.625.413.644.21

[back to top]

Median of 6 Month 4-hydroxy-tamoxifen (4HT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 6 month 4HT steady state plasma concentrations for each CYP2D6 phenotype group are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 6 Month

InterventionnM (Median)
PM/PMIM/PMIM/IMPM/EMEM/IMEM/EMEM/UM
¹³C-DM-BT3.171.691.53.815.862.965.38

[back to top]

Median of 6 Month Endoxifen Steady State Concentrations (Endx Css) According to CYP2D6 Phenotype Group and Activity Score

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 6 month Endx Css for each CYP2D6 phenotype group and the corresponding activity score are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 6 Month

InterventionnM (Median)
EM/UM (3.0)EM/EM (2.0)EM/IM (1.5)IM/IM (1.0)EM/PM (1.0)IM/PM (0.5)PM/PM (0)
¹³C-DM-BT38.526.730.411.319.14.76.7

[back to top]

Median of 6 Month N-desmethyl-tamoxifen (NDMT) Steady State Plasma Concentrations According to CYP2D6 Phenotype Group

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 6 month NDMT steady state plasma concentrations for each CYP2D6 phenotype group are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 6 Month

InterventionnM (Median)
PM/PMIM/PMIM/IMPM/EMEM/IMEM/EMEM/UM
¹³C-DM-BT554.6542.4718.2546.5525.9401.8552.9

[back to top]

Median of 6 Month Tamoxifen Steady State Plasma Concentrations According to CYP2D6 Phenotype Group

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The median of 6 month Tamoxifen steady state plasma concentrations for each CYP2D6 phenotype group are summarized below. Refer to Outcome Measure 1 Data Table for details on the combination of genotype for each participant that are defined for each CYP2D6 phenotype group." (NCT00873366)
Timeframe: 6 Month

InterventionnM (Median)
PM/PMIM/PMIM/IMPM/EMEM/IMEM/EMEM/UM
¹³C-DM-BT194.0200.5279.2259.7234.7179.7301.4

[back to top]

Operating Characteristics of the ¹³C-dextromethorphan (13C-DM) Breath Test in Identifying Those Who Are CYP2D6 Genotypic Poor Metabolizers

"Participants were classified as having CYP2D6 decreased or non-decreased metabolism based on genotype and the co-prescription of inhibitors of the enzyme system.~The specific phenotype, alleles and their associated activity score (AS) assessed were as follows:~Ultrarapid metabolism (UM) or AS=2.0 (*1XN or *2XN), Extensive metabolism (EM) or AS=1.0 (*1, *2, and *2A), Intermediate metabolism (IM) or AS=0.5 (*9, *10, *17 and *41), and Poor metabolism (PM) or AS=0.0 (3, *4, *5, *7, *8, *11, and *12). The distribution of CYP2D6 genotypes grouped by CYP2D6 metabolism phenotype for each participants are summarized below." (NCT00873366)
Timeframe: Baseline

InterventionParticipants (Count of Participants)
EM/UM (*1/*2AXN)EM/UM (*1/*1XN)IM/UM (*41/*2AXN)EM/EM (*1/*1)EM/EM (*1/*2A)EM/EM (*1/*2)EM/EM (*2/*2)EM/EM (*2A/*2A)EM/IM (*1/*9)EM/IM (*1/*10)EM/IM (*1/*41)EM/IM (*2A/*9)EM/IM (*2A/*41)EM/PM (*1/*3)EM/PM (*1/*4)EM/PM (*2/*4)EM/PM (*2/*4XN)EM/PM (*2A/*4)EM/PM (*2A/*5)EM/PM (*2A/*6)IM/IM (*41/*41)IM/PM (*3/*41)IM/PM (*4/*9)IM/PM (*4/*10)IM/PM (*4/*41)PM/PM (*3/*4)PM/PM (*4/*4)
¹³C-DM-BT11112132122371218116111111411

[back to top]

Spearman's Rank Correlation Coefficient Between CYP2D6 Activity Score and Endoxifen Steady State Concentrations (Endx Css)

Spearman rank order correlation coefficients were used to assess the strength of the association between CYP2D6 genotype activity score and Endx Css (NCT00873366)
Timeframe: 3 Month and 6 Month

InterventionCorrelation coefficient (Number)
3 Month6 Month
¹³C-DM-BT0.470.56

[back to top]

Spearman's Rank Correlation Coefficient Between CYP2D6 Activity Score and Endoxifen/N-desmethyl-tamoxifen (Endx/NDMT) Ratio

Spearman rank order correlation coefficients were used to assess the strength of the association between CYP2D6 genotype activity score and Endx/NDMT ratio (NCT00873366)
Timeframe: 3 Month and 6 Month

InterventionCorrelation coefficient (Number)
3 Month6 Month
¹³C-DM-BT0.600.61

[back to top]

Spearman's Rank Correlation Coefficient Between CYP2D6 Genotype and ¹³Cdextromethorphan Breath Test (DM-BT)

Spearman rank order correlation coefficients were used to assess the strength of the association between CYP2D6 genotype activity score and DM-BT values. (NCT00873366)
Timeframe: Baseline, 3 month and 6 month

InterventionCorrelation coefficient (Number)
Baseline3 Month6 Month
¹³C-DM-BT0.550.580.55

[back to top]

Spearman's Rank Correlation Coefficient Between Baseline DM-BT and 3 Month Endoxifen/N-desmethyl-tamoxifen (Endx/NDMT) Ratio

Spearman rank order correlation coefficients were used to assess the strength of the association between baseline ¹³Cdextromethorphan breath test (DM-BT) and Endoxifen/N-desmethyl-tamoxifen (Endx/NDMT) Ratio (NCT00873366)
Timeframe: Baseline, 3 month

InterventionCorrelation coefficient (Number)
¹³C-DM-BT0.56

[back to top]

Spearman's Rank Correlation Coefficient Between 6 Month DM-BT and 6 Month Endoxifen Steady State Concentrations

Spearman rank order correlation coefficients were used to assess the strength of the association between baseline ¹³Cdextromethorphan breath test (DM-BT) and Endoxifen steady state concentrations (Endx Css) (NCT00873366)
Timeframe: 6 month

InterventionCorrelation coefficient (Number)
¹³C-DM-BT0.54

[back to top]

Spearman's Rank Correlation Coefficient Between 3 Month DM-BT and 3 Month Endoxifen Steady State Concentrations

Spearman rank order correlation coefficients were used to assess the strength of the association between baseline ¹³Cdextromethorphan breath test (DM-BT) and Endoxifen steady state concentrations (Endx Css) (NCT00873366)
Timeframe: 3 month

InterventionCorrelation coefficient (Number)
¹³C-DM-BT0.51

[back to top]

Spearman's Rank Correlation Coefficient Between Baseline DM-BT and 3 Month Endoxifen Steady State Concentrations

Spearman rank order correlation coefficients were used to assess the strength of the association between baseline ¹³Cdextromethorphan breath test (DM-BT) and Endoxifen steady state concentrations (Endx Css) (NCT00873366)
Timeframe: Baseline, 3 month

InterventionCorrelation coefficient (Number)
¹³C-DM-BT0.6

[back to top]

Change From Baseline in Perceived Numerical Cough Severity Scale for 6 Hour Post-Dose Period

"Perceived numerical cough severity score was assessed on an 11-point categorical rating scale in accordance to the question How much have you coughed in the last hour?, where 0 = did not cough at all and 10 = cough a lot. The change from baseline for the 6-hour post-dosing period was calculated as the average of change from baseline (i.e. baseline value minus the post baseline value) measurements of Hour 1 to Hour 6, thus the change from baseline values ranged from -10 to 10; higher score indicated a better improvement." (NCT01257542)
Timeframe: Baseline, 1, 2, 3, 4, 5, 6 hour post-dose

,
InterventionUnits on a scale (Mean)
BaselineChange for 6 hour post-dose period
Dextromethorphan Hydrobromide7.02.5
Placebo6.82.8

[back to top]

Change From Baseline in Perceived Verbal Cough Severity Scale for 6 Hour Post-Dose Period

"Perceived verbal cough severity score was assessed on a 5-point categorical rating scale in accordance to the question How much have you coughed in the last hour?, where 0 = not at all, 1 = a tiny bit, 2 = a little, 3 = some and 4 = a lot. The change from baseline over the 6-hour post-dosing period calculated as the average of change from baseline [that is (i.e.) baseline value minus the post baseline value] measurements of Hour 1 to Hour 6, thus the change from baseline ranged from -4 to 4; higher score indicated a better improvement." (NCT01257542)
Timeframe: Baseline, 1, 2, 3, 4, 5, 6 hours post-dose

,
InterventionUnits on a scale (Mean)
BaselineChange for 6 hour post-dose period
Dextromethorphan Hydrobromide2.91.1
Placebo3.01.3

[back to top]

Participants' Global Assessment of Cough: Cough Severity

"Participant global assessment of cough with the assistance of parent or legal guardian was scored on a 5-point categorical scale based on response to the question How much have you coughed in the past 6 hours? where 0 = not at all, 1 = a tiny bit, 2 = a little, 3 = some and 4 = a lot." (NCT01257542)
Timeframe: Within 5 minutes after Hour 6

InterventionUnits on a scale (Mean)
Placebo3.1
Dextromethorphan Hydrobromide3.0

[back to top]

Participants' Global Assessment of Cough: Relief From Cough

"Participant global assessment of cough with the assistance of parent or legal guardian was scored on a 5-point categorical scale based on response to the question From when you woke up this morning until now, how much better is your cough? where 0 = not at all better, 1 = a tiny bit better, 2 = a little better, 3 = better and 4 = a lot better." (NCT01257542)
Timeframe: Within 5 minutes after Hour 6

InterventionUnits on a scale (Mean)
Placebo2.9
Dextromethorphan Hydrobromide2.7

[back to top]

Total Cough Count

Total cough count was done by trained assessors using continuous digital video and audio recordings. (NCT01257542)
Timeframe: Up to 6 hours post-dose

InterventionCough counts (Mean)
Placebo238.7
Dextromethorphan Hydrobromide190.9

[back to top]

Change From Baseline in Verbal Cough Severity Scale at Hours 1, 2, 3, 4, 5 and 6

"Verbal cough severity score was assessed on a 5-point categorical rating scale in accordance to the question How much have you coughed in the last hour?, where 0 = not at all, 1 = a tiny bit, 2 = a little, 3 = some and 4 = a lot. The change from baseline values were derived by subtracting each post baseline value from the baseline value, and ranged from -4 to 4; higher score indicated a better improvement." (NCT01257542)
Timeframe: Baseline, 1, 2, 3, 4, 5, 6 hours

,
InterventionUnits on a scale (Mean)
Change at 1 hourChange at 2 hoursChange at 3 hoursChange at 4 hoursChange at 5 hoursChange at 6 hours
Dextromethorphan Hydrobromide0.90.90.91.11.21.2
Placebo1.01.21.31.41.71.5

[back to top]

Change From Baseline in Numerical Cough Severity Scale at Hours 1, 2, 3, 4, 5 and 6

"Numerical cough severity score was assessed on an 11-point categorical rating scale in accordance to the question How much have you coughed in the last hour?, wherein 0 = did not cough at all and 10 = cough a lot. The change from baseline was derived by subtracting the post baseline value from the baseline value and ranged from -10 to 10; higher score indicated a better improvement." (NCT01257542)
Timeframe: Baseline, 1, 2, 3, 4, 5, 6 hours

,
InterventionUnits on a scale (Mean)
Change at 1 hourChange at 2 hoursChange at 3 hoursChange at 4 hoursChange at 5 hoursChange at 6 hours
Dextromethorphan Hydrobromide2.02.12.32.53.02.8
Placebo2.02.52.72.93.63.4

[back to top]

Average Logarithms of Dextromethorphan (DM) Plasma Concentrations (Cmax) on Days 22 and 50

The average of the logarithms of the DM plasma concentrations on Days 22 and 50 were presented. (NCT01324232)
Timeframe: 0 to 3 hours post-dose on Day 22 and 50

InterventionLog Micrograms per Liter (Median)
AVP-923-204.019
AVP-923-304.493
AVP-923-454.758
Total4.394

[back to top]

Mean Change From Baseline in Pittsburgh Sleep Quality Index (PSQI) Scores at Day 85

PSQI, a self-rated questionnaire was used to assess sleep quality and disturbances over a 1-month time interval. A total of 19 individual items generated 7 component scores: subjective sleep quality, sleep latency, sleep duration, habitual sleep efficiency, sleep disturbances, use of sleeping medication, and daytime dysfunction. Each component was scored from 0 (no difficulty) to 3 (severe difficulty). The sum of the scores for the 7 components yielded 1 global score (ranging from 0 to 21). Higher PSQI score indicated worse quality of sleep. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. The analysis was carried out using an ANCOVA model, with the PSQI change from Baseline to Day 85 as the dependent variable, treatment group as a fixed effect, and the Baseline PQIS as a covariate. (NCT01324232)
Timeframe: Baseline; Day 85

Interventionunits on a scale (Least Squares Mean)
Placebo-0.51
AVP-923-20-1.02
AVP-923-30-1.36
AVP-923-45-1.71

[back to top]

Mean Overall Patient Global Impression of Change (PGIC) Scores at Day 85

The PGIC was a standard, validated 7-point categorical scale. The participant was asked to assess the overall change in his or her central neuropathic pain symptoms since entry into the study on a scale of 0 to 7 (0=much better; 7=much worse). Higher scores indicated worsening. (NCT01324232)
Timeframe: Day 85

Interventionunits on a scale (Mean)
Placebo3.58
AVP-923-204.42
AVP-923-303.70
AVP-923-453.64
Total3.84

[back to top]

Mean Change From Baseline in Expanded Disability Status Scale (EDSS) Scores at Days 22 and 85

"The EDSS, a method of quantifying disability in participants with MS was based on neurological examination of 8 functional systems (FS) (pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, cerebral, and other) that allowed neurologists to assign a FS score to each of these systems. Neurological findings in each FS were scored on a scale of 0 (low level of problems) to 5 (high level of problems). The other category was not rated numerically but measured disability related to a particular issue, like motor loss. A total averaged EDSS score was then calculated on a scale of 0 (normal) to 10 (death from MS). The total EDSS score was determined by 2 factors: gait and FS scores. A higher score indicated greater disability. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value." (NCT01324232)
Timeframe: Baseline; Days 22 and 85

,,,,
Interventionunits on a scale (Mean)
Day 22Day 85
AVP-923-20-0.1-0.1
AVP-923-30-0.2-0.1
AVP-923-45-0.00.0
Placebo-0.1-0.1
Total-0.1-0.1

[back to top]

Association Between the Dextromethorphan Plasma Concentration and the Change From Baseline Pain Rating Scale (PRS) Score to the Average Pain Rating Scale Score During Days 57 Through 84

PRS required participants to rate their pain over the past 12 hours (hrs) on a scale of 0 to 10 (0=no pain; 10=worst possible pain). Baseline (B) PRS was defined as the average of the PRS scores in the last 7 days collected prior to the B visit. If participants did not have at least 4 PRS scores during the last 7 days prior to the B visit, then the average of up to 7 of the most recent PRS scores available prior to the B visit was used. Post-B PRS was the average of Days 57 through 84 values. For participants who did not have any PRS scores during Days 57 through 84, the average of the last 7 available post-B PRS scores was used. Change from B was calculated as the post-B score minus B score. The average logarithms of dextromethorphan plasma concentrations (Cmax) on Days 22 and 50 are reported in primary outcome measure #2 below. Pearson correlation was calculated between Cmax as one group of data across the reporting groups and Change from Baseline in PRS score. (NCT01324232)
Timeframe: Baseline; Days 57 through 84 (PRS score); Days 22 and 50 (dextromethorphan plasma concentrations)

Interventionunits on a scale (Median)
Placebo-1.821
AVP-923-20-2.143
AVP-923-30-2.650
AVP-923-45-1.679
Total-2.000

[back to top]

Comparison of the Adjusted Mean Change From Baseline PRS Score to the Average PRS Score During Days 57 Through 84

The PRS required participants to rate their pain over the past 12 hours on a scale of 0 to 10 (0=no pain; 10=worst possible pain) by circling the number that best described their pain on average over the past 12 hours. Baseline PRS was defined as the average of the PRS scores in the last 7 days collected prior to the Baseline visit. If participants did not have at least 4 PRS scores during the last 7 days prior to the Baseline visit, then the average of up to 7 of the most recent PRS scores available prior to the Baseline visit was used. Post-Baseline PRS was the average of the Day 57 through 84 values. For participants who did not have any PRS scores during Days 57 through 84, the average of the last 7 available post-Baseline PRS scores was used. Change from Baseline was calculated as the post-Baseline score minus the Baseline score. (NCT01324232)
Timeframe: Baseline; Days 57 through 84

Interventionunits on a scale (Least Squares Mean)
Placebo-2.04
AVP-923-20-2.07
AVP-923-30-2.41
AVP-923-45-2.00
AVP-923-20 and AVP-923-30-2.24
AVP-923-30 and AVP-923-45-2.21
All AVP-923-2.16

[back to top]

Mean Change From Baseline in Beck Depression Inventory (BDI-II) Scores at Day 85

BDI-II, a 21-item, self-reported instrument was used to assess the existence and severity of symptoms of depression. Each item corresponded to a symptom of depression and as scored on a 4-point scale, ranging from 0 to 3. Participants were asked to consider each statement as it related to the way they have felt for the past 2 weeks. Each of the 21 items were summed to give a single score for the BDI-II (ranging from 0 to 63). A total score of 0 to 13 indicated minimal depression, a score of 14 to 19 indicated mild depression, a score of 20 to 28 indicated moderate depression, and a score of 29 to 63 indicated severe depression. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. The analysis was carried out using an ANCOVA model, with the BDI-II change from Baseline to Day 85 as the dependent variable, treatment group as a fixed effect, and the Baseline BDI-II as a covariate. (NCT01324232)
Timeframe: Baseline; Day 85

Interventionunits on a scale (Least Squares Mean)
Placebo0.83
AVP-923-20-0.26
AVP-923-300.21
AVP-923-451.15

[back to top]

Mean Change From Baseline in Fatigue Severity Scale (FSS) Scores at Days 57 Through 84

"The FSS questionnaire consisted of 9 statements that attempted to explore the severity of fatigue symptoms in participants with MS and other conditions, including chronic fatigue immune dysfunction syndrome and systemic lupus erythematosus, and was designed to differentiate fatigue from clinical depression because both share some of the same symptoms. Participants were asked to respond to each statement on a scale of 1 to 7, with 1 indicating Strongly Disagree and 7 indicating Strongly Agree. Total score ranging from 9 to 63, was computed as the sum of the sub-scores for all 9 statements; a higher score indicated increasing fatigue. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Post-Baseline FSS score was the average of Day 57 through 84 values. The analysis was carried out using an ANCOVA model, with the FSS change from Baseline to Days 85 as the dependent variable," (NCT01324232)
Timeframe: Baseline; Days 57 through 84

Interventionunits on a scale (Least Squares Mean)
Placebo-3.32
AVP-923-20-2.00
AVP-923-30-6.32
AVP-923-45-2.12

[back to top]

Mean Change From Baseline in MS Neuropsychological Screening Questionnaire (MSNQ) Scores at Day 85

MSNQ, a self-reporting, 15-item questionnaire was used to screen for cognitive impairment in participants with MS. Participants (or their informants) scored each item on a scale from 0 (not at all) to 4 (often and greatly interferes with life). The total MSNQ score was calculated as the sum of the sub-scores for all 15 questions and thus ranged from 0 to 60. A higher score indicated greater impairment. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. The analysis was carried out using an ANCOVA model, with the MSNQ score change from Baseline to Day 85 as the dependent variable, treatment group as a fixed effect, and the Baseline MSNQ as a covariate. (NCT01324232)
Timeframe: Baseline; Day 85

Interventionunits on a scale (Least Squares Mean)
Placebo-0.99
AVP-923-20-1.48
AVP-923-30-1.88
AVP-923-450.47

[back to top]

Mean Change From Baseline in Multiple Sclerosis Impact Scale-29 (MSIS-29) Scores at Day 85

MSIS-29, an instrument measuring the physical (20 items) and psychological (9 items) impact of MS from the participants' perspective was used to evaluate therapeutic effectiveness from the participants' perspective. Participants were asked to circle the response that best described the impact of MS on daily life on a scale of 0 (not at all) to 5 (extremely). The total MSIS-29 score ranged from 0 to 145, was calculated as the sum of the sub-scores for all 29 questions, with lower scores indicating better quality of life. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. The analysis was carried out using an ANCOVA model, with the MSIS-29 score change from Baseline to Day 85 as the dependent variable, treatment group as a fixed effect, and the Baseline MSIS-29 as a covariate. (NCT01324232)
Timeframe: Baseline; Day 85

Interventionunits on a scale (Least Squares Mean)
Placebo-4.84
AVP-923-20-4.34
AVP-923-30-6.50
AVP-923-45-1.41

[back to top]

Mean Change From Baseline in Symbol Digit Modalities Test (SDMT) Scores at Day 85

The SDMT assessed organic cerebral dysfunction in both children (8 years and older) and adults. The SDMT involved a simple substitution task that normal participants could easily perform. Using a reference key, the examinee had 90 seconds to pair specific numbers with given geometric figures. The SDMT score was the total correct response (not counting errors) in 90 seconds and ranged from 0 to 110. Lower scores indicated increased dysfunction. Baseline was defined as last non-missing measurement prior to dosing. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. The analysis was carried out using an ANCOVA model, with the SDMT change from Baseline to Day 85 as the dependent variable, treatment group as a fixed effect, and the Baseline SDMT as a covariate. (NCT01324232)
Timeframe: Baseline; Day 85

,,,
Interventionunits on a scale (Least Squares Mean)
Total correct responses, OralTotal correct responses, Written
AVP-923-202.963.62
AVP-923-301.910.16
AVP-923-452.060.20
Placebo0.591.85

[back to top]

Mean Numerical Rating Scale (NRS) Scores at Days 22, 50, and 85

The NRS was an 11-point scale for participant self-reporting of pain. The overall scores ranged from 0 (no spasticity) to 10 (worst possible spasticity). Higher scores indicated increased aggression. (NCT01324232)
Timeframe: Days 22, 50, and 85

,,,,
Interventionunits on a scale (Mean)
Day 22Day 50Day 85
AVP-923-203.562.893.13
AVP-923-303.414.063.87
AVP-923-453.903.543.36
Placebo3.293.243.66
Total3.543.423.50

[back to top]

AUC of Dextromethorphan, Midazolam and Omeprazole in the Presence of Fluoxetine

Our secondary outcome measure will be the interaction between fluoxetine and each CYP evaluated in the cocktail. A 50% increase in the AUC of caffeine (CYP1A2), dextromethorphan (CYP2D6), omeprazole (CYP2C19) or midazolam (CYP3A4) between treatment and control days is considered clinically significant. The interaction of fluoxetine with caffeine (CYP1A2) will be considered as a negative control for the study. These AUCs will be measured on study day 1 (control day) and study day 18 (NCT01361217)
Timeframe: The secondary outcome will be assessed within 2 months after the last subject is enrolled or at 2 years from the start of study enrollment, which ever is sooner.

Interventionnmol*hr/L (Mean)
caffeine control AUCCaffeine treatment AUCdextromethophan control AUCDextromethorphan treatment AUCOmeprazole control AUComeprazole treatment AUCmidazolam control AUCmidazolam treatment AUC
Midazolam, Caffeine, Omeprazole, Caffeine AUC After Fluoxetine4300043000681850120085003024

[back to top]

Lovastatin AUC in the Presence of Fluoxetine

Our primary outcome measure will be the interaction of fluoxetine with CYP3A4. A 50% increase in the AUC for lovastatin plus hydroxy-lovastatin acid (the active form of lovastatin) between treatment day 14 (study day 20) and control days (study day 2) is considered clinically significant. (NCT01361217)
Timeframe: The primary outcome will be assessed within 2 months after the last subject is enrolled or at 2 years from the start of study enrollment, which ever is sooner.

Interventionnmol*hr/L (Mean)
Lovastatin AUC After Fluoxetine Dosing170
Lovastatin Before Fluoxetine180

[back to top]

Mean Pain Intensity (Percent Change From Baseline)

Primary outcome was percent change from baseline in mean pain intensity (transformed Gracely Scale; 0-35). Baseline was defined as the week prior to randomization. The greater the percent change, the bigger the reduction in pain intensity. (NCT01435798)
Timeframe: 1st week of maintenance period (week prior to hospital admission for nested study; subjects traveled to Boston on days 6-7 of the maintenance period)

InterventionPercent change from baseline (Mean)
0% MTD Dex-0.006
25% MTD Dex-0.018
50% MTD Dex-0.073
100% MTD Dex-0.24

[back to top]

Changes in Mean Macular Sensitivity in the Study Eye at 24 Months Compared to Baseline

Microperimetry was used to assess macular sensitivity. (NCT01441102)
Timeframe: Baseline and 24 Months

InterventiondB (Mean)
Dextromethorphan Hydrobromide-3.9

[back to top]

Number of Participants Withdrawn From the Study Therapy Due to Vision Loss or Adverse Events

(NCT01441102)
Timeframe: Duration of the study, up to 24 months

Interventionparticipants (Number)
Dextromethorphan Hydrobromide1

[back to top]

Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 12 Months Compared to Baseline

Fluorescein angiography (FA) images were obtained via a standard digital imaging system (OIS, Sacramento, CA) at baseline and at Month 6, Month 12, Month 18, and Month 24. Three retinal specialists independently graded the area of late fluorescein leakage (at approximately 10 minutes) using a region-of-interest tool in an image analysis software package (NIH ImageJ, Bethesda, MD). (NCT01441102)
Timeframe: Baseline and 12 Months

Interventioneyes (Number)
Dextromethorphan Hydrobromide3

[back to top]

Changes in Mean Macular Sensitivity in the Study Eye at 6 Months Compared to Baseline

Microperimetry was used to assess macular sensitivity. (NCT01441102)
Timeframe: Baseline and 6 Months

InterventiondB (Mean)
Dextromethorphan Hydrobromide-2.1

[back to top]

Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 18 Months Compared to Baseline

Visual acuity was measured using the Early Treatment Diabetic Retinopathy Study (ETDRS) protocol. Acuity is measured as letters read on an ETDRS eye chart and the letters read equate to Snellen measurements. For example, if a participant reads between 84 and 88 letters, the equivalent Snellen measurement is 20/20. (NCT01441102)
Timeframe: Baseline and 18 Months

InterventionETDRS letters (Mean)
Dextromethorphan Hydrobromide4.00

[back to top]

Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 24 Months Compared to Baseline

Visual acuity was measured using the Early Treatment Diabetic Retinopathy Study (ETDRS) protocol. Acuity is measured as letters read on an ETDRS eye chart and the letters read equate to Snellen measurements. For example, if a participant reads between 84 and 88 letters, the equivalent Snellen measurement is 20/20. (NCT01441102)
Timeframe: Baseline and 24 Months

InterventionETDRS letters (Mean)
Dextromethorphan Hydrobromide5.50

[back to top]

Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 6 Months Compared to Baseline

Visual acuity was measured using the Early Treatment Diabetic Retinopathy Study (ETDRS) protocol. Acuity is measured as letters read on an ETDRS eye chart and the letters read equate to Snellen measurements. For example, if a participant reads between 84 and 88 letters, the equivalent Snellen measurement is 20/20. (NCT01441102)
Timeframe: Baseline and 6 Months

InterventionETDRS letters (Mean)
Dextromethorphan Hydrobromide0.60

[back to top]

Changes in Mean Macular Sensitivity in the Study Eye at 12 Months Compared to Baseline

Microperimetry was used to assess macular sensitivity. (NCT01441102)
Timeframe: Baseline and 12 Months

InterventiondB (Mean)
Dextromethorphan Hydrobromide-3.7

[back to top]

Change in Best-corrected Visual Acuity (BCVA) in the Study Eye at 12 Months Compared to Baseline

Visual acuity was measured using the Early Treatment Diabetic Retinopathy Study (ETDRS) protocol. Acuity is measured as letters read on an ETDRS eye chart and the letters read equate to Snellen measurements. For example, if a participant reads between 84 and 88 letters, the equivalent Snellen measurement is 20/20. (NCT01441102)
Timeframe: Baseline and 12 Months

InterventionETDRS letters (Mean)
Dextromethorphan Hydrobromide1.50

[back to top]

Changes in Mean Macular Sensitivity in the Study Eye at 18 Months Compared to Baseline

Microperimetry was used to assess macular sensitivity. (NCT01441102)
Timeframe: Baseline and 18 Months

InterventiondB (Mean)
Dextromethorphan Hydrobromide-4.9

[back to top]

Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 18 Months Compared to Baseline

Fluorescein angiography (FA) images were obtained via a standard digital imaging system (OIS, Sacramento, CA) at baseline and at Month 6, Month 12, Month 18, and Month 24. Three retinal specialists independently graded the area of late fluorescein leakage (at approximately 10 minutes) using a region-of-interest tool in an image analysis software package (NIH ImageJ, Bethesda, MD). (NCT01441102)
Timeframe: Baseline and 18 Months

Interventioneyes (Number)
Dextromethorphan Hydrobromide2

[back to top]

Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 24 Months Compared to Baseline

Fluorescein angiography (FA) images were obtained via a standard digital imaging system (OIS, Sacramento, CA) at baseline and at Month 6, Month 12, Month 18, and Month 24. Three retinal specialists independently graded the area of late fluorescein leakage (at approximately 10 minutes) using a region-of-interest tool in an image analysis software package (NIH ImageJ, Bethesda, MD). (NCT01441102)
Timeframe: Baseline and 24 Months

Interventioneyes (Number)
Dextromethorphan Hydrobromide4

[back to top]

Number of Study Eyes Demonstrating a Decrease in the Area of Late Leakage, as Measured by Fluorescein Angiography (FA), at 6 Months Compared to Baseline

Fluorescein angiography (FA) images were obtained via a standard digital imaging system (OIS, Sacramento, CA) at baseline and at Month 6, Month 12, Month 18, and Month 24. Three retinal specialists independently graded the area of late fluorescein leakage (at approximately 10 minutes) using a region-of-interest tool in an image analysis software package (NIH ImageJ, Bethesda, MD). (NCT01441102)
Timeframe: Baseline and 6 Months

Interventioneyes (Number)
Dextromethorphan Hydrobromide4

[back to top]

Percentage Change in Retinal Thickness in the Study Eye at 12 Months Compared to Baseline

Retinal thickness was assessed by spectral-domain optical coherence tomography (Cirrus HD-OCT; Carl Zeiss Meditec, Dublin, CA), a non-invasive imaging technique that uses long-wavelength light to capture micrometer-resolution cross-sectional images from biological tissue. Changes in OCT will be calculated using the ETDRS grid. Attention will be directed to changes in retinal thickness as measured by OCT in each of the 9 subfields of the grid. (NCT01441102)
Timeframe: Baseline and 12 Months

Interventionpercentage change in retinal thickness (Mean)
Dextromethorphan Hydrobromide6.54

[back to top]

Percentage Change in Retinal Thickness in the Study Eye at 18 Months Compared to Baseline

Retinal thickness was assessed by spectral-domain optical coherence tomography (Cirrus HD-OCT; Carl Zeiss Meditec, Dublin, CA), a non-invasive imaging technique that uses long-wavelength light to capture micrometer-resolution cross-sectional images from biological tissue. Changes in OCT will be calculated using the ETDRS grid. Attention will be directed to changes in retinal thickness as measured by OCT in each of the 9 subfields of the grid. (NCT01441102)
Timeframe: Baseline and 18 Months

Interventionpercentage change in retinal thickness (Mean)
Dextromethorphan Hydrobromide-0.99

[back to top]

Percentage Change in Retinal Thickness in the Study Eye at 24 Months Compared to Baseline

Retinal thickness was assessed by spectral-domain optical coherence tomography (Cirrus HD-OCT; Carl Zeiss Meditec, Dublin, CA), a non-invasive imaging technique that uses long-wavelength light to capture micrometer-resolution cross-sectional images from biological tissue. Changes in OCT will be calculated using the ETDRS grid. Attention will be directed to changes in retinal thickness as measured by OCT in each of the 9 subfields of the grid. (NCT01441102)
Timeframe: Baseline and 24 Months

Interventionpercentage change in retinal thickness (Mean)
Dextromethorphan Hydrobromide-7.44

[back to top]

Percentage Change in Retinal Thickness in the Study Eye at 6 Months Compared to Baseline

"Retinal thickness was assessed by spectral-domain optical coherence tomography (Cirrus HD-OCT; Carl Zeiss Meditec, Dublin, CA), a non-invasive imaging technique that uses long-wavelength light to capture micrometer-resolution cross-sectional images from biological tissue. The participant's eye that met the study eye eligibility criteria was selected as the study eye. For cases in which both eyes met the study eye eligibility criteria, the study eye was selected according to the choice of study eye in cases of bilateral disease selection criteria outlined in the eligibility criteria. The eye not chosen as the study eye is referred to as the fellow eye." (NCT01441102)
Timeframe: Baseline and 6 months

Interventionpercentage change in retinal thickness (Mean)
Dextromethorphan Hydrobromide-7.89

[back to top]

TP-DI Sub-study: Dextromethorphan to Dextrorphan Urine Concentration Ratio

(NCT01462318)
Timeframe: Week 43 (7 days prior to DAC HYP administration) and Week 53 (7 days after DAC HYP administration), pre-cocktail dose and for 12 hours after probe-drug cocktail administration

Interventionratio (Mean)
Dextromethorphan (Period 1)Dextromethorphan+DAC HYP (Period 2)
TP-DI Sub-study0.424680.48939

[back to top]

TP-DI Sub-study: Omeprazole/Hydroxyomeprazole Concentration Ratio at 2 Hours Post-omeprazole Dosing

(NCT01462318)
Timeframe: Week 43 (7 days prior to DAC HYP administration) and Week 53 (7 days after DAC HYP administration) at 2 hours after probe drug cocktail administration

Interventionratio (Mean)
Omeprazole (Period 1)Omeprazole+ DAC HYP (Period 2)
TP-DI Sub-study2.6731.028

[back to top]

Intensive PK Sub-study: Apparent Clearance (CL/F) of DAC HYP

(NCT01462318)
Timeframe: Day 141 (Week 20) at pre-dose and 8, 24, 72 and 120 hours post-dose and 7, 10, 14 and 21 days post-dose

InterventionL/day (Mean)
Main Study0.27

[back to top]

Intensive PK Sub-study: Apparent Volume of Distribution (V/F) of DAC HYP

(NCT01462318)
Timeframe: Day 141 (Week 20) at pre-dose and 8, 24, 72 and 120 hours post-dose and 7, 10, 14 and 21 days post-dose

InterventionLiters (Mean)
Main Study8.21

[back to top]

Intensive PK Sub-study: Elimination Half-life (t½) of DAC HYP

(NCT01462318)
Timeframe: Day 141 (Week 20) at pre-dose and 8, 24, 72 and 120 hours post-dose and 7, 10, 14 and 21 days post-dose

Interventionday (Mean)
Main Study21.92

[back to top]

Intensive PK Sub-study: Minimum Concentrations (Cmin) of DAC HYP

(NCT01462318)
Timeframe: Day 141 (Week 20) at pre-dose and 8, 24, 72 and 120 hours post-dose and 7, 10, 14 and 21 days post-dose

Interventionmcg/mL (Mean)
Main Study14.93

[back to top]

Intensive PK Sub-study: Area-Under-the-Curve From Start to End of the Dosing Interval (AUCtau) of DAC HYP

(NCT01462318)
Timeframe: Day 1 and Day 141 (Week 20) at pre-dose and 8, 24, 72, and 120 hours post-dose and 7, 10, 14, and 21 days post-dose

Interventionday*mcg/mL (Mean)
Week 0 (Day 1); n=25Week 20; n=24
Main Study255.25638.10

[back to top]

Intensive PK Sub-study: Cmax of DAC HYP

(NCT01462318)
Timeframe: Day 1 and Day 141 (Week 20) at pre-dose and 8, 24, 72, and 120 hours post-dose and 7, 10, 14 and 21 days post-dose

Interventionmcg/mL (Mean)
Day 1 (Week 0); n=25Day 141 (Week 20); n=24
Main Study12.6329.07

[back to top]

Intensive PK Sub-study: Time to Reach Maximum Concentration (Tmax) of DAC HYP

(NCT01462318)
Timeframe: Day 1 and Day 141 (Week 20) at pre-dose and 8, 24, 72, and 120 hours post-dose and 7, 10, 14 and 21 days post-dose

Interventionday (Mean)
Day 1 (Week 0); n=25Day 141 (Week 20); n=24
Main Study9.316.41

[back to top]

Number of Participants With Anti-DAC HYP Binding Antibodies (ADAbs): Electrochemiluminescent (ECL) Anti-Drug Antibody (ADA) Assay

Participants with post-baseline (PB) ADAbs through Week 44, in the treatment period (extends up to 42 days after the last dose during the main study), and in the post-treatment period (43 days after the last dose until the end of the post-treatment period dose). (NCT01462318)
Timeframe: Up to 44 weeks

Interventionparticipants (Number)
PB ADAbs through Week 44=negative; n=113PB ADAbs through Week 44=positive; n=113PB ADAbs in treatment period=negative; n=113PB ADAbs in treatment period=positive; n=113PB ADAbs in post-treatment period=negative; n=110PB ADAbs in post-treatment period=positive; n=110
Main Study783592218921

[back to top]

Number of Participants With Anti-DAC HYP Neutralizing Antibodies (NAbs): ECL ADA Assay

Participants with PB NAbs through Week 44, in the treatment period (extends up to 42 days after the last dose during the main study), and in the post-treatment period (43 days after the last dose until the end of the post-treatment period dose). (NCT01462318)
Timeframe: Up to 44 weeks

Interventionparticipants (Number)
PB NAbs through Week 44=negative; n=113PB NAbs through Week 44=positive; n=113PB NAbs in treatment period=negative; n=113PB NAbs in treatment period=positive; n=113PB NAbs in post-treatment period=negative; n=110PB NAbs in post-treatment period=positive; n=110
Main Study105810941046

[back to top]

TP-DI Sub-study: Area-Under-the-Curve From Zero to Infinity (AUCinf) of Each Probe Drug

AUCinf of each of the following cytochrome P450 (CYP) isoenzyme substrates: midazolam (CYP3A), S-warfarin + vitamin K (CYP2C9), and omeprazole (CYP2C19). The AUC from zero to 12 hours (AUC0-12) was calculated for caffeine (CYP1A2). (NCT01462318)
Timeframe: Week 43 (7 days prior to DAC HYP administration) and Week 53 (7 days after DAC HYP administration), pre-cocktail dose and at 0.5 and 1, 2, 3, 4, 6, 8, 10 , 24, 48, 72 and 96 hours post-probe drug cocktail administration

Interventionhr*ng/mL (Mean)
Midazolam (Period 1) AUCinf; n=20Midazolam+DAC HYP (Period 2) AUCinf; n=19S-warfarin (Period 1) AUCinf; n=17S-warfarin+DAC HYP (Period 2) AUCinf; n=18Omeprazole (Period 1) AUCinf; n=18Omeprazole+DAC HYP (Period 2) AUCinf; n=19Caffeine (Period 1) AUC0-12; n=20Caffeine+DAC HYP (Period 2) AUC0-12; n=20
TP-DI Sub-study786.75816.8719292.919609.32214.51770.035742.437449.2

[back to top]

TP-DI Sub-study: CL/F of Each Probe Drug

CL/F of each of the following CYP isoenzyme substrates: midazolam (CYP3A), warfarin + vitamin K (CYP2C9), and omeprazole (CYP2C19). (NCT01462318)
Timeframe: Week 43 (7 days prior to DAC HYP administration) and Week 53 (7 days after DAC HYP administration), pre-cocktail dose and at 0.5 and 1, 2, 3, 4, 6, 8, 10 , 24, 48, 72 and 96 hours post-probe drug cocktail administration

InterventionmL/hr (Mean)
Midazolam (Period 1); n=20Midazolam+DAC HYP (Period 2); n=19S-Warfarin (Period 1); n=17S-Warfarin+DAC HYP (Period 2); n=18Omeprazole (Period 1); n=18Omeprazole+DAC HYP (Period 2); n=19
TP-DI Sub-study7625.77298.6565.86541.4641612.441772.4

[back to top]

TP-DI Sub-study: Cmax of Each Probe Drug

Cmax of each of the following CYP isoenzyme substrates: midazolam (CYP3A), caffeine (CYP1A2), warfarin + vitamin K (CYP2C9), and omeprazole (CYP2C19). (NCT01462318)
Timeframe: Week 43 (7 days prior to DAC HYP administration) and Week 53 (7 days after DAC HYP administration), pre-cocktail dose and at 0.5 and 1, 2, 3, 4, 6, 8, 10 , 24, 48, 72 and 96 hours post-probe drug cocktail administration

Interventionng/mL (Mean)
Midazolam (Period 1); n=20Midazolam+DAC HYP (Period 2); n=19Caffeine (Period 1); n=20Caffeine+DAC HYP (Period 2); n=19S-Warfarin (Period 1); n=20S-Warfarin+DAC HYP (Period 2); n=19Omeprazole (Period 1); n=19Omeprazole+DAC HYP (Period 2); n=19
TP-DI Sub-study271.05311.214965.05399.5635.65649.74776.95771.16

[back to top]

Change in PedsQL Social Functioning Subscale Score, Pre- and Post-Intervention

Pediatric Quality of Life Inventory (PedsQL version 4). Social Functioning subscale. 5-point Likert scale from 0 (Never) to 4 (Almost always); Items are reversed scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Higher scores indicate better Health Related Quality of Life (QOL). (NCT01520363)
Timeframe: Baseline and at the end of the 3 month trial

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)58.53
Placebo Group54.64
Study Drug Group at 3 Months46.76
Placebo at 3 Months47.14

[back to top]

Change in PedsQL Total Score, Pre- and Post-Intervention

Pediatric Quality of Life Inventory (PedsQL version 4) total score. Each item is rated on a 5-point Likert scale from 0 (Never) to 4 (Almost always). Items are reversed scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. The Total Score is the sum of all the items over the number of items answered on all the Scales. Higher scores indicate better HRQOL. (NCT01520363)
Timeframe: Baseline evaluation and at the end of the 3 month study

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)54.018
Placebo Group54.157
Study Drug Group at 3 Months50.9235
Placebo at 3 Months51.7286

[back to top]

Change in Rett Syndrome Behavior Questionnaire Score, Pre- and Post-Intervention

The Rett Syndrome Behavior Questionnaire (RSBQ) total score was assessed. The total score ranges from 0 to 90, with 0 exhibiting no Rett syndrome related symptoms and 90 showing the greatest amount of symptoms (worse outcome). (NCT01520363)
Timeframe: Initial evaluation and at the end of the 3 month study

Interventionunits on a scale (Mean)
Study Drug-dextromethorphan (DM)33.8
Placebo Group33.3
Study Drug-dextromethorphan (DM) at 3 Months33.8
Placebo Group at 3 Months33.3

[back to top]

Change in Seizure Frequency, Pre- and Post-Intervention, 0-4 Year Age Group

Change in Frequency of seizure count baseline to follow-up for children aged 0-4 years (NCT01520363)
Timeframe: Baseline evaluation and at the end of the 3 month study

Interventionseizure count (Mean)
Study Drug-dextromethorphan (DM)3
Placebo Group3.2
Study Drug-dextromethorphan (DM) at 3 Months3.7
Placebo Group at 3 Months3.1

[back to top]

Change in Seizure Frequency, Pre-and Post-Intervention, 5-10 Year Age Group

Change in Frequency of seizures baseline to follow-up for children aged 5-10 years (NCT01520363)
Timeframe: Baseline evaluation and at the end of the 3 month study

Interventionseizure count (Mean)
Study Drug-dextromethorphan (DM)3.6
Placebo Group2.8
Study Drug Group at 3 Months3.36
Placebo at 3 Months3.37

[back to top]

Change in VABS: Socialization Domain Scores, Pre- and Post-Intervention

Vineland Adaptive Behavior Scales-II (VABS): Socialization Domain. Critical behaviors are scored on a Likert scale from 2=Usually, 1=Sometimes or Partially, 0= Seldom or Never. Socialization Domain raw scores range from: Minimum=0 to Maximum=152. A higher score is a better outcome. (NCT01520363)
Timeframe: Baseline and at the end of the 3 month trial

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)24.14
Placebo Group22.96
Study Drug-dextromethorphan (DM) at 3 Months24.05
Placebo Group at 3 Months22.48

[back to top]

Change in VABS: Motor Skills Domain Scores, Pre- and Post-Intervention

Vineland Adaptive Behavior Scales-II (VABS): Motor Skills Domain Scores individual items are scored on a Likert scale from 2=Usually, 1=Sometimes or Partially, 0= Seldom or Never. Motor Skills Domain raw scores range from: Minimum=0 to Maximum=100. A higher score is a better outcome. (NCT01520363)
Timeframe: Baseline evaluation and at the end of the 3 month study

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)9.86
Placebo Group11.36
Study Drug-dextromethorphan (DM) at 3 Months9.64
Placebo Group at 3 Months11.12

[back to top]

Change in VABS:Communication Domain Scores, Pre- and Post-Intervention

Vineland Adaptive Behavior Scales (VABS)-II Communication Domain Scores. The Communication Domain evaluates the receptive, expressive, and written communication skills of the child. Critical behaviors in each Subdomain item are rated as 2=Usually, 1=Sometimes or Partially, 0= Seldom or Never. Communication Domain raw scores range from: Minimum=0 to Maximum=198. A higher score is a better outcome. (NCT01520363)
Timeframe: Baseline and at the end of the 3 month trial

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)18.05
Placebo Group18.08
Study Drug-dextromethorphan (DM) at 3 Months17.91
Placebo Group at 3 Months17.96

[back to top]

Change in PedsQL Emotional Functioning Subscale Score, Pre- and Post-Intervention

Pediatric Quality of Life Inventory (PedsQL version 4). Emotional Functioning subscale. 5-point Likert scale from 0 (Never) to 4 (Almost always); Items are reversed scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Higher scores indicate better Health Related Quality of Life (QOL). (NCT01520363)
Timeframe: Baseline evaluation and at the end of the 3 month study

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)72.37
Placebo Group68.93
Study Drug-dextromethorphan (DM) at 3 Months68.621
Placebo Group at 3 Months68.214

[back to top]

Change in VABS:Daily Living Skills Domain Scores, Pre- and Post-Intervention

Vineland Adaptive Behavior Scales-II (VABS): Daily Living Skills Domain individual items are scored on a Likert scale from 2=Usually, 1=Sometimes or Partially, 0= Seldom or Never. The Daily Living Skills Domain measures personal behavior as well as domestic and community interaction skills. Daily Living Skills Domain raw scores range from Minimum=0 to Maximum=218. (NCT01520363)
Timeframe: Baseline and at the end of the 3 month trial

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)21.64
Placebo Group21.00
Study Drug-dextromethorphan (DM) at 3 Months20.95
Placebo Group at 3 Months19.84

[back to top]

Change in Ghuman-Folstein Screen for Social Interaction (SSI) Score, Pre- and Post-Intervention.

"The Ghuman-Folstein Screen for Social Interaction (SSI) assesses the change in behavior and temperament dysregulation as a total score.~The score ranges from 0-162, with 0 being most Impaired /has the strongest autism features and 162 having no impairment/no autism features." (NCT01520363)
Timeframe: Initial evaluation and at the end of the 3 month study. The test lasts 45 minutes

Interventionunits on a scale (Mean)
Study Drug-dextromethorphan (DM)73.1
Placebo Group71.3
Study Drug Group at 3 Months73.1
Placebo at 3 Months68.0

[back to top]

Change in Mullen, Expressive Language Sub-scale Scores, Pre- and Post-Intervention

The Mullen Scales of Early Learning (MULLEN) Expressive Language scale raw scores range from Minimum=0 to Maximum=50. A higher score is a better outcome. Age equivalents from 1 month to 70 months can be computed for each subscale separately. (NCT01520363)
Timeframe: Baseline and 3 months

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)5.33
Placebo Group6.73
Study Drug Group at 3 Months6.76
Placebo at 3 Months7.29

[back to top]

Change in Mullen; Fine Motor Sub-scale Scores, Pre- and Post-Intervention

The Mullen Scales of Early Learning (MULLEN) Fine motor scale raw scores range from Minimum=0 to Maximum=49. A higher score is a better outcome. Age equivalents from 1 month to 70 months can be computed for each subscale separately. (NCT01520363)
Timeframe: Baseline and 3 months

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)5.38
Placebo Group7.00
Study Drug Group at 3 Months5.05
Placebo at 3 Months7.04

[back to top]

Change in Mullen; Receptive Language Subscale Scores, Pre- and Post-Intervention

The Mullen Scales of Early Learning (MULLEN) Receptive Language scale raw scores range from Minimum=0 to Maximum=50. A higher score is a better outcome. Age equivalents from 1 month to 70 months can be computed for each subscale separately. (NCT01520363)
Timeframe: Baseline and 3 months

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)6.45
Placebo Group6.95
Study Drug Group at 3 Months6.68
Placebo at 3 Months6.55

[back to top]

Change in Mullen; Visual Reception Sub-scale Scores, Pre- and Post-Intervention

The Mullen Scales of Early Learning (MULLEN) Visual reception subscale raw scores range from Minimum=0 to Maximum=50. A higher score is a better outcome. Age equivalents from 1 month to 70 months can be computed for each subscale separately. (NCT01520363)
Timeframe: Initial evaluation and at the end of the 3 month trial

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)5.45
Placebo Group6.61
Study Drug Group at 3 Months6.64
Placebo at 3 Months6.57

[back to top]

Change in PedsQL Physical Functioning Subscale Score, Pre- and Post-Intervention

Pediatric Quality of Life Inventory (PedsQL version 4). Physical Functioning subscale. 5-point Likert scale from 0 (Never) to 4 (Almost always); Items are reversed scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Higher scores indicate better Health Related Quality of Life (QOL). (NCT01520363)
Timeframe: Initial evaluation and at the end of the 3 month study

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)42.717
Placebo Group48.973
Study Drug Group at 3 Months38.900
Placebo at 3 Months48.133

[back to top]

Change in PedsQL School Functioning Subscale Score, Pre- and Post-Intervention

Pediatric Quality of Life Inventory (PedsQL version 4). School Functioning subscale. 5-point Likert scale from 0 (Never) to 4 (Almost always); Items are reversed scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Higher scores indicate better Health Related Quality of Life (QOL). (NCT01520363)
Timeframe: Baseline evaluation and at the end of the 3 month study

Interventionscore on a scale (Mean)
Study Drug-dextromethorphan (DM)64.231
Placebo Group49.583
Study Drug Group at 3 Months55.769
Placebo at 3 Months44.592

[back to top]

Number of Participants With the Indicated Change in the Concomitant Use of Allowed Psychotropic Drugs Compared to Their Baseline Use

Concomitant medications (CMs) are defined as non-study medications with a start date on or before the final study visit, and that were either ongoing at the end of the study or had a stop date on or after the date of first dose of study drug. Drugs for the treatment of AD (e.g., donepezil, rivastigmine, galantamine, memantine) were allowed when administered at stable dose for at least 2 months prior to randomization. Concomitant use of the following medications was allowed, provided the participant had been on a stable dose for at least 1 month prior to randomization and remained stable throughout the study: medications for agitation secondary to AD; medications for nighttime management of insomnia or behavioral disturbances that included short-acting benzodiazepines, a low dose of alprazolam up to 0.5 milligrams (mg)/day, and a low dose of trazodone up to 50 mg/day; hypnotics for the treatment of insomnia; and certain classes of antidepressants. (NCT01584440)
Timeframe: up to Week 10

,,
Interventionparticipants (Number)
Any changeDose reducedDose increasedChange in frequencyUse discontinuedNew usage
AVP-923 in Stage 1/AVP-923 in Stage 2910127
Placebo in Stage 1/AVP-923 in Stage 2801126
Placebo in Stage 1/Placebo in Stage 213111211

[back to top]

Change in the Caregiver Strain Index (CSI) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The CSI is a 13-question tool that is used to measure strain related to care provision and to identify families with potential caregiving concerns. There is at least one item for each of the following major domains: Employment, Financial, Physical, Social, and Time. A 0 (No) to 1 (Yes) scale is used for each of the 13 questions; thus the total score ranges from 0 to 13. Higher scores signify higher stress levels. Positive responses to 7 or more items on the index indicate a greater level of strain. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-1.2-0.2
Placebo-0.60.1

[back to top]

Change in the NPI Agitation/Aggression Domain Score From Day 1 (Stage 1 Baseline) to Day 8 and Day 22 and From Day 36 (Stage 2 Baseline) to Day 43 and Day 57

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. The Agitation/Aggression domain was designed to collect information on the behavioral aspects of agitation/aggression in participants with probable AD and clinically meaningful agitation secondary to AD. Each NPI domain is rated by the caregiver for symptom frequency and severity. Symptom frequency is rated as: 1, occasionally; 2, often; 3, frequently; 4, very frequently. Symptom severity is rated as: 1, mild; 2, moderate; 3, marked. The total domain score is calculated as the frequency score multiplied by the severity score and thus ranges from 1 to 12. A higher score represents worsening symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Days 8 and 22; Day 36 (Stage 2 Baseline); Days 43 and 57

,
Interventionscore on a scale (Mean)
Day 8Day 22Day 43Day 57
AVP-923-2.2-2.7-0.5-1.2
Placebo-1.4-1.6-1.3-1.3

[back to top]

Change in the Alzheimer's Disease Cooperative Study-Activities of Daily Living Inventory (ADCS-ADL) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The ADCS-ADL inventory measures basic activities of daily living such as dressing, conversation, eating, bathing, and grooming. The 19-item version, covering mainly basic ADL, is used to assess participants with more severe disabilities. The ADCS-ADL uses a scale from 0 to 54. Lower scores indicate declining ability. Change from Baseline is calculated as the post-Baseline value minus the Baseline value. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-0.9-2.0
Placebo-0.8-0.6

[back to top]

Change in the Cornell Scale for Depression in Dementia (CSDD) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The CSDD is a 19-item interview specifically developed to assess the signs and symptoms of major depression in participants with dementia. Each of the 19 items is rated for severity on a scale of 0 to 2 (0, absent; 1, mild or intermittent; 2, severe). The total score is calculated as the sum of the item scores and thus ranges from 0 to 38. Higher scores signify more severe depression. Scores above 10 indicate probable major depression. Scores above 18 indicate definite major depression. Scores below 6, as a rule, are associated with the absence of significant depressive symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-1.0-0.9
Placebo0.6-0.7

[back to top]

Change in the Individual NPI Domain Scores From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. Each NPI domain is rated by the caregiver for symptom frequency and severity. Symptom frequency is rated as: 1, occasionally; 2, often; 3, frequently; 4, very frequently. Symptom severity is rated as: 1, mild; 2, moderate; 3, marked. The total domain score is calculated as the frequency score multiplied by the severity score and thus ranges from 1 to 12. A higher score represents worsening symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. Sleep/nighttime behavior disorders = S/NB disorders. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36, DelusionsDay 36, HallucinationsDay 36, Depression/dysphoriaDay 36, AnxietyDay 36, Euphoria/elationDay 36, Apathy/indifferenceDay 36, DisinhibitionDay 36, Irritability/labilityDay 36, Aberrant motor behaviorDay 36, S/NB disordersDay 36, Appetite/eating changesDay 70, DelusionsDay 70, HallucinationsDay 70, Depression/dysphoriaDay 70, AnxietyDay 70, Euphoria/elationDay 70, Apathy/indifferenceDay 70, DisinhibitionDay 70, Irritability/labilityDay 70, Aberrant motor behaviorDay 70, S/NB disordersDay 70, Appetite/eating changes
AVP-923-0.80.2-0.9-0.6-0.3-0.9-0.9-2.2-1.2-1.0-1.20.1-0.10.2-1.0-0.2-0.5-0.8-1.0-0.80.00.1
Placebo-0.6-0.1-1.0-1.2-0.1-0.5-0.7-1.2-0.4-0.1-1.2-0.7-0.40.2-0.3-0.00.4-0.8-0.70.4-0.10.4

[back to top]

Change in the Mini-Mental State Examination (MMSE) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline), as Analyzed by the Specified SPCD Methodology

The MMSE is a brief test that is used to screen for cognitive impairment. The MMSE scale comprises 11 questions or simple tasks concerning orientation, memory, attention, and language to evaluate the participant's cognitive state. The anchor values are not consistent for each task. The MMSE total score is calculated by summing the item scores across all 11 tasks. A participant's total possible MMSE score ranges from 0 to 30 points. Higher scores indicate milder cognitive impairment. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-9230.20.3
Placebo-0.3-0.5

[back to top]

Change in the Neuropsychiatric Inventory (NPI) Agitation/Aggression Domain Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. The Agitation/Aggression domain was designed to collect information on the behavioral aspects of agitation/aggression in participants with probable Alzheimer's Disease (AD) and clinically meaningful agitation secondary to AD. Each NPI domain is rated by the caregiver for symptom frequency and severity. Symptom frequency is rated as: 1, occasionally; 2, often; 3, frequently; 4, very frequently. Symptom severity is rated as: 1, mild; 2, moderate; 3, marked. The total domain score is calculated as the frequency score multiplied by the severity score and thus ranges from 1 to 12. A higher score represents worsening symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. Data are reported for only those participants contributing data to the analysis. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-3.3-2.0
Placebo-1.7-0.8

[back to top]

Change in the NPI-CDS for the Agitation/Aggression Domain From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. For the Agitation/Aggression domain, the caregiver is asked to rate how emotionally distressing they find the symptom behavior on the following scale: 0, not at all; 1, minimally; 2, mildly; 3, moderately; 4, severely; 5, very severely or extremely. A higher score represents increased distress. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-1.4-0.5
Placebo-0.6-0.7

[back to top]

Change in the NPI-CDS NPI4A Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. For the NPI4A NPI-CDS score, the caregiver is asked to rate how emotionally distressing they find the symptom behavior (for the Agitation/Aggression, Irritability/Lability, Anxiety, and Aberrant Motor Behavior domains) on the following scale: 0, not at all; 1, minimally; 2, mildly; 3, moderately; 4, severely; 5, very severely or extremely. The NPI4A NPI-CDS score ranges from 0 to 20. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-3.3-1.5
Placebo-1.7-1.0

[back to top]

Change in the NPI-CDS NPI4D Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. For the NPI4D NPI-CDS score, the caregiver is asked to rate how emotionally distressing they find the symptom behavior (for the Agitation/Aggression, Irritability/Lability, Disinhibition, and Aberrant Motor Behavior domains) on the following scale: 0, not at all; 1, minimally; 2, mildly; 3, moderately; 4, severely; 5, very severely or extremely. The NPI4D NPI-CDS score ranges from 0 to 20. A higher score represents increased distress. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-3.3-1.8
Placebo-1.5-1.2

[back to top]

Change in the Quality of Life-Alzheimer's Disease (QoL-AD) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The QoL-AD is a brief, 13-item measure designed specifically to obtain a rating of the participant's quality of life (QoL) from both the participant and the caregiver. It uses simple and straightforward language and responses and includes assessments of the individual's relationships with friends and family, concerns about finances, physical condition, mood, and an overall assessment of life quality. Caregivers complete the measure as a questionnaire about the participants' QoL, whereas participants complete it in interview format about their own QoL. Each of the 13 items is rated on a 4-point scale, with 1 indicating a poor QoL and 4 indicating an excellent QoL. Total scores range from 13 to 52, with a higher score indicating a better QoL. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36, CargiverDay 70, CaregiverDay 36, ParticipantDay 70, Participant
AVP-9230.4-0.31.31.5
Placebo0.30.9-0.00.7

[back to top]

Change in the Sum of the Agitation/Aggression, Irritability/Lability, Anxiety, and Aberrant Motor Behavior NPI Domain (NPI4A) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. Each NPI domain is rated by the caregiver for symptom frequency and severity. Symptom frequency is rated as: 1, occasionally; 2, often; 3, frequently; 4, very frequently. Symptom severity is rated as: 1, mild; 2, moderate; 3, marked. The total domain score is calculated as the frequency score multiplied by the severity score and thus ranges from 1 to 12. The NPI4A score is the sum of the Agitation/Aggression, Irritability/Lability, Anxiety, and Aberrant Motor Behavior domain scores, and thus ranges from 4 to 48. A higher score represents worsening symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-7.3-4.8
Placebo-4.5-1.4

[back to top]

Change in the Sum of the Agitation/Aggression, Irritability/Lability, Disinhibition, and Aberrant Motor Behavior NPI Domain (NPI4D) Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. Each NPI domain is rated by the caregiver for symptom frequency and severity. Symptom frequency is rated as: 1, occasionally; 2, often; 3, frequently; 4, very frequently. Symptom severity is rated as: 1, mild; 2, moderate; 3, marked. The total domain score is calculated as the frequency score multiplied by the severity score and thus ranges from 1 to 12. The NPI4D score is the sum of the Agitation/Aggression, Irritability/Lability, Disinhibition, and Aberrant Motor Behavior domain scores, and thus ranges from 4 to 48. A higher score represents worsening symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-7.6-4.6
Placebo-4.0-1.9

[back to top]

Change in the Total Neuropsychiatric Inventory-Caregiver Distress Score (NPI-CDS) From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. For each domain, the caregiver is asked to rate how emotionally distressing they find the symptom behavior on the following scale: 0, not at all; 1, minimally; 2, mildly; 3, moderately; 4, severely; 5, very severely or extremely. The total NPI-CDS score is calculated as the sum of all 12 domain scores and thus ranges from 0 to 60. A higher score represents increased distress. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-6.6-2.6
Placebo-3.6-2.0

[back to top]

Change in the Total NPI Score From Day 1 (Stage 1 Baseline) to Day 36 (Stage 2 Baseline) and From Day 36 to Day 70

The NPI is a retrospective interview covering 12 neuropsychiatric symptom domains and is used to evaluate psychopathology, neuropsychiatric manifestations, and caregiver distress. Each NPI domain is rated by the caregiver for symptom frequency and severity. Symptom frequency is rated as: 1, occasionally; 2, often; 3, frequently; 4, very frequently. Symptom severity is rated as: 1, mild; 2, moderate; 3, marked. The total domain score is calculated as the frequency score multiplied by the severity score and thus ranges from 1 to 12. The total score is calculated as a sum of all 12 domain scores and thus ranges from 12 to 144. A higher score represents worsening symptoms. Change from Baseline is calculated as the post-Baseline score minus the Baseline score. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionscore on a scale (Mean)
Day 36Day 70
AVP-923-13.5-6.0
Placebo-8.5-2.5

[back to top]

Number of Participants Using Rescue Medications

Participants were allowed to receive oral lorazepam as rescue medication for the short-term treatment of symptoms of agitation, if deemed necessary by the investigator. Lorazepam was to be administered at doses up to 1.5 mg/day, and dosing was not to exceed 3 days in any 7-day period. (NCT01584440)
Timeframe: up to Week 10

,,
Interventionparticipants (Number)
OverallDay 8 (Visit 2)Day 22 (Visit 3)Day 36 (Visit 4)Day 43 (Visit 5)Day 57 (Visit 6)Day 70 (Visit 70)
AVP-923 in Stage 1/AVP-923 in Stage 25035212
Placebo in Stage 1/AVP-923 in Stage 211333333
Placebo in Stage 1/Placebo in Stage 27133543

[back to top]

Number of Participants With the Indicated Categorical Response on the Patient Global Impression of Change (PGI-C) for the Caregiver Domain at Day 36 (Stage 2 Baseline) and Day 70 Compared to Their Response at Day 1 (Stage 1 Baseline)

The PGI-C uses a 7-point scale to assess treatment response and to rate the global impression of clinical change in a participant's agitation. The participant is asked to rate their impression of change from Baseline as: 1, very much improved; 2, much improved; 3, minimally improved; 4, no change; 5, minimally worse; 6, much worse; 7, very much worse. Baseline is defined as the last non-missing assessment prior to Stage 1 randomization. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionparticipants (Number)
Day 36, Very much improvedDay 36, Much improvedDay 36, Minimally improvedDay 36, No changeDay 36, Minimally worseDay 36, Much worseDay 36, Very much worseDay 70, Very much improvedDay 70, Much improvedDay 70, Minimally improvedDay 70, No changeDay 70, Minimally worseDay 70, Much worseDay 70, Very much worse
AVP-9231024202013103101410420
Placebo22330402341341113940

[back to top]

Number of Participants With the Indicated Categorical Response on the PGI-C for the Caregiver Domain at Day 70 Compared to Their Response at Day 1 (Stage 1 Baseline)

The PGI-C uses a 7-point scale to assess treatment response and to rate the global impression of clinical change in a participant's agitation. The participant is asked to rate their impression of change from Baseline as: 1, very much improved; 2, much improved; 3, minimally improved; 4, no change; 5, minimally worse; 6, much worse; 7, very much worse. For placebo non-responders re-randomized to AVP-923 or placebo at Stage 2, Baseline is defined as the last non-missing assessment prior to Stage 2 re-randomization. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 70

,
Interventionparticipants (Number)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVery much worse
AVP-9239262513530
Placebo5917141040

[back to top]

Number of Participants With the Indicated Response on the Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change Rating (mADCS-CGIC) Scale Agitation Domain at Day 36 and Day 70 Compared to Their Response at Day 1 (Stage 1 Baseline)

The intent of the ADCS version of the CGIC is to provide a means to reliably assess the global impression of change from Baseline in a clinical trial. The mADCS-CGIC is a modification of the ADCS-CGIC instrument that focuses specifically on agitation. The participant is asked to rate their impression of change from Baseline as: 1, marked improvement; 2, moderate improvement; 3, minimal improvement; 4, no change; 5, minimal worsening; 6, moderate worsening; 7, marked worsening. Baseline is defined as the last non-missing assessment prior to Stage 1 randomization. (NCT01584440)
Timeframe: Day 1 (Stage 1 Baseline); Day 36 (Stage 2 Baseline); Day 70

,
Interventionparticipants (Number)
Day 36, Marked improvementDay 36, Moderate improvementDay 36, Minimal improvementDay 36, No changeDay 36, Minimal worseningDay 36, Moderate worseningDay 36, Marked worseningDay 70, Marked improvementDay 70, Moderate improvementDay 70, Minimal improvementDay 70, No changeDay 70, Marked worseningDay 70, Moderate worseningDay 70, Minimal worsening
AVP-92382228227104101012330
Placebo11343481260451114901

[back to top]

Number of Participants With the Indicated Response on the mADCS-CGIC Scale Agitation Domain at Day 70 Compared to Their Response at Day 36 (Stage 2 Baseline)

The intent of the ADCS version of the CGIC is to provide a means to reliably assess the global impression of change from Baseline in a clinical trial. The mADCS-CGIC is a modification of the ADCS-CGIC instrument that focuses specifically on agitation. The participant is asked to rate their impression of change from Baseline as: 1, marked improvement; 2, moderate improvement; 3, minimal improvement; 4, no change; 5, minimal worsening; 6, moderate worsening; 7, marked worsening. For placebo non-responders re-randomized to AVP-923 or placebo at Stage 2, Baseline is defined as the last non-missing assessment prior to Stage 2 re-randomization. (NCT01584440)
Timeframe: Day 36 (Stage 2 Baseline); Day 70

,
Interventionparticipants (Number)
Marked improvementModerate improvementMinimal improvementNo changeMarked worseningModerate worseningMinimal worsening
AVP-9230111511410
Placebo42819621

[back to top]

Number of Participants With the Indicated Type of Adverse Event

Treatment-emergent adverse events (TEAEs) are defined as AEs that first occurred, or worsened, after the first dose of study medication and within 30 days after the permanent discontinuation of the study medication (first dose date ≤ AE start date ≤ date of last dose + 30 days) (NCT01584440)
Timeframe: up to Week 10

,
InterventionParticipants (Count of Participants)
Participants with ≥ 1 TEAEParticipants with discontinuations due to a TEAEParticipants who died
All AVP-9239380
Placebo5540

[back to top]

Primary Safety Endpoints

Number of serious adverse events (NCT01630811)
Timeframe: Week 0 through week 25

InterventionParticipants (Count of Participants)
Nuedexta0
Placebo0

[back to top]

Change in Maladaptive Behaviors

Demonstrate a change in frequency and intensity of maladaptive behaviors as measured by the Aberrant Behavior Checklist (ABC) Irritability subscale in subjects given Nuedexta 8 weeks over subjects given placebo. This checklist consists of 20 questions relating to behavior and the reported total score is on a scale from 0 to 60. A lower score can be interpreted as less frequent and/or less intense presentation of the undesirable behavior. The below values are the difference in ABC scores from baseline to 8 weeks. A negative difference indicates improved behavior. (NCT01630811)
Timeframe: Baseline and 8 weeks

Interventionunits on a scale (Mean)
Nuedexta-6.62
Placebo-1.08

[back to top]

Change in Aggressive Behavior

Demonstrate a trend towards reduced aggressive behavior as measured by Overt Aggression Scale (OAS). It consists of a scale from 0 - 40, and a lower score can be interpreted as less frequent and/or less intense presentation of the undesirable behavior. Reported is the mean difference in scores from baseline to 8 weeks. A positive score indicates more aggressive behavior and a negative score indicates less aggressive behavior. (NCT01630811)
Timeframe: Baseline and 8 weeks

Interventionunits on a scale (Mean)
Nuedexta-1.92
Placebo0.08

[back to top]

Adjusted Geometric Mean Cmax of Dextromethorphan With and Without the Coadministration of Belatacept - PK Evaluable Population

Cmax was measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for dextromethorphan with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. The poor metabolizer of CYP2D6 was excluded from the statistical analysis. Inje cocktail components (dextromethorphan) measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionng/mL (Geometric Mean)
Day 1 Inje Cocktail0.927
Day 4 Inje Cocktail Plus Belatacept0.800
Day 7 Inje Cocktail0.855
Day 11 Inje Cocktail0.794

[back to top]

Adjusted Geometric Mean Cmax of Caffeine With and Without the Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. Cmax was measured in ng/mL. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionng/mL (Geometric Mean)
Day 1 Inje Cocktail4696.505
Day 4 Inje Cocktail Plus Belatacept4450.019
Day 7 Inje Cocktail4332.326
Day 11 Inje Cocktail4479.687

[back to top]

Mean Change From Baseline in Sitting Heart Rate - All Treated Participants

Heart Rate was taken after the participant had been sitting quietly for at least 5 minutes and the heart rate was measured in beats per minute (bpm). Heart Rates were obtained at screening visit, Day -1, and at 0 hour (pre-dose), 0.5 hour (post dose), and 2 hours (post dose) on Days 1, 4, 7, 11. Baseline was defined as last non-missing result with a collection date-time less than the date-time of the first active dose of study drug. (NCT01766050)
Timeframe: Baseline and 0.5 and 2.0 hours Post Dose on Days 1, 4, 7, and 11

,,,
Interventionbpm (Mean)
Heart Rate 0.5 hour post dose (N=22, 21, 21, 20)Heart Rate 2.0 hour post dose (N=22, 21, 21, 20)
Day 1 Inje Cocktail-3.3-3.5
Day 11 Inje Cocktail-1.63.1
Day 4 Inje Cocktail Plus Belatacept-4.20.2
Day 7 Inje Cocktail-1.80.5

[back to top]

Cmax of Inje Cocktail Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail component metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail component metabolites were each measured using HPLC with MS/MS detection. Cmax was measured in ng/mL. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng/mL (Geometric Mean)
1'-Hydroxy-Midazolam (N=22, 21, 21, 20)E-3174 (N=22, 21, 21, 20)5-Hydroxyomeprazole (N=22, 21, 21, 20)Dextrorphan (N=21,20,20,19)Paraxanthine (N=17,19,18,18)
Day 1 Inje Cocktail11.12623353421291
Day 11 Inje Cocktail12.32913373311358
Day 4 Inje Cocktail Plus Belatacept11.92763603241367
Day 7 Inje Cocktail11.43003323121320

[back to top]

AUC(INF) of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without Coadministration of Belatacept - PK Evaluable Population

Area under the plasma concentration-time curve from time zero extrapolated to infinite time [AUC(INF)] was measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng*h/mL (Geometric Mean)
1'-hydroxy-midazolam (N=22,21,20,20)E-3174 (N=22,20,21,20)5-hydroxyomeprazole (N=22,20,21,17dextrorphan (N=21,20,20,19)paraxanthine (N=7,8,6,6)
Day 1 Inje Cocktail30.521031010167922401
Day 11 Inje Cocktail35.121821063168221874
Day 4 Inje Cocktail Plus Belatacept33.82060990161022905
Day 7 Inje Cocktail32.92187938156222063

[back to top]

Adjusted Geometric Mean Area Under the Concentration Time Curve (AUC) From Zero to Last Concentration (0-T) and AUC Extrapolated to Infinity (INF) of Midazolam With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population

AUC(0-T): area under the plasma concentration-time curve from zero to the last time of the last quantifiable concentration and AUC (INF): AUC from time zero extrapolated to infinite time were measured in ng*h/mL. Samples for the assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for midazolam with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. Midazolam measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7and 11

,,,
Interventionng*h/mL (Geometric Mean)
AUC (0-T);N=22, 21, 21, 20)AUC (INF);N=22, 21, 21, 20)
Day 1 Inje Cocktail65.54867.743
Day 11 Inje Cocktail67.71769.841
Day 4 Inje Cocktail Plus Belatacept68.83371.039
Day 7 Inje Cocktail63.30365.555

[back to top]

Apparent Total Body Clearance (CLT/F) of the Inje Cocktail Components (Midazolam, Losartan, Omeprazole, Dextromethorphan and Caffeine) With and Without Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of dextromethorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. CLT/F was measured as liters/hour (L/h) (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
InterventionL/h (Geometric Mean)
Midazolam (N=22, 21, 21, 20)Losartan (N=22, 21, 21, 20)Omeprazole (N=22, 19, 21, 18)Dextromethorphan (N=18,17,18,18)Caffeine (N=22, 21, 21, 18)
Day 1 Inje Cocktail73.814829.239164.99
Day 11 Inje Cocktail71.415125.339684.83
Day 4 Inje Cocktail Plus Belatacept71.314623.742475.30
Day 7 Inje Cocktail77.314624.742644.97

[back to top]

Number of Participants With Marked Hematology and Urinalysis Laboratory Abnormalities - All Treated Participants

Samples for laboratory tests were obtained at Screening visit, Day -1 or prior to dosing on Day 1, Days 3, 6, 10, 46 ±2, and at early termination, after 10 hours fasting. Leukocytes: *10^9 cells per liter (c/L) < 0.85*Pre-Rx if Pre-Rx < LLN or <0.9*LLN if LLN <= Pre-Rx or Pre-Rx is missing. Neutrophils (absolute): *10^12 c/L < 0.85* Pre-Rx if Pre-Rx < 1.5, <1.5 if Pre-Rx >= 1.5, < 1.5 if Pre-Rx missing. Urine blood from dipstick: >=2 if Pre-Rx <1 or was missing or if Pre-Rx >=1. Urinary microscopic white blood cells (WBC) and red blood cells (RBC) >= 2 if Pre-Rx <2 or if Pre-Rx was missing or >=4 if Pre-Rx >=2. (NCT01766050)
Timeframe: Day -1 to Day 46 ±2 days or at early termination

Interventionparticipants (Number)
Leukocytes <0.85*Pre-Rx *10^9 c/L (N=22)Neutrophils <0.85*Pre-Rx *10^12 c/L (N=22)Urine Blood >= 2 (N=22)Urinary RBC microscopic >= 2 (N=8)Urinary WBC microscopic >= 2 (N=8)
Inje Cocktail Alone and Inje Cocktail Plus Belatacept14211

[back to top]

Number of Participants With Marked Serum Chemistry Laboratory Abnormalities - All Treated Participants

Samples for laboratory tests were obtained at Screening visit, Day -1 or prior to dosing on Day 1, Days 3, 6, 10, 46, and at early termination, after 10 hours fasting. Upper limits of normal (ULN); Lower limits of normal (LLN); Pre-therapy (Rx); micromoles per liter (µmol/L); millimoles per liter (mmol/L); grams per liter (g/L); Units per liter (U/L); Aspartate Aminotransferase (AST); Blood Urea Nitrogen (BUN) Total Bilirubin: >1.1*ULN if Pre-Rx<= ULN or Pre-Rx is missing, or >1.2*Pre-Rx if Pre-Rx >ULN. AST: >1.25*Pre-Rx if Pre-Rx >ULN or 1.25*ULN if Pre-Rx <= ULN or Pre-Rx is missing. BUN: >1.1*ULN if Pre-Rx<= ULN or Pre-Rx is missing, or >1.2*Pre-Rx if Pre-Rx >ULN. Phosphorus: <0.85*LLN if Pre-RX >= LLN or is missing or if Pre-Rx < LLN. total Protein: <0.9*LLN if Pre-Rx>= LLN or is missing or Pre-Rx > LLN. Creatine Kinase: >1.5*Pre-Rx if Pre-Rx > ULN or is missing or Pre-Rx is <= ULN. Lactate Dehydrogenase: >1.25*ULN if Pre-Rx <= ULN or missing, >1.5*Pre-Rx if Pre-Rx > ULN. (NCT01766050)
Timeframe: Day -1 to Day 46 ±2 days or at early termination

Interventionparticipants (Number)
Total bilirubin >1.1*ULN µmol/L (N=22)AST >1.25*Pre-Rx U/L (N=22)BUN >1.1*ULN mmol/L (N=22)Inorganic Phosphorus <0.85*LLN mmol/L (N=22)Total Protein <0.9*LLN g/L (N=22)Creatine Kinase > 1.5* Pre-Rx U/L (N=22)Lactate Dehydrogenase >1.25*ULN U/L (N=22)
Inje Cocktail Alone and Inje Cocktail Plus Belatacept1111121

[back to top]

Number of Participants With Out-of-Range Electrocardiogram Intervals - All Treated Participants

Participants had 12-Lead electrocardiograms (ECGs) performed at Screening Visit, Day 1 prior to dosing, Day 46 ±2, and at early termination. Definition of out-of-range: PR Interval >210 milliseconds (msec); QRS > 120 msec, QT > 500 msec or > 30 msec change from baseline (Day 1); QT with Fridericia correction (QTcF) > 450 msec or change from baseline of > 30 msec to <= 60 msec or change from baseline > 60 msec. (NCT01766050)
Timeframe: Day 1 to Day 46 ±2 days or at early termination

Interventionparticipants (Number)
Day 46 (N=18)Early Termination (N=2)
Inje Cocktail Alone and Inje Cocktail Plus Belatacept00

[back to top]

Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Omeprazole With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population

AUC(0-T): Area under the plasma concentration-time curve from time zero zero to the time of the last quantifiable concentration and AUC (INF): AUC extrapolated to infinity were measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for omeprazole with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. Inje cocktail components (omeprazole) measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng*h/mL (Geometric Mean)
AUC (0-T); (N= 22, 21, 21, 20)AUC (INF); (N= 22, 19, 21, 18)
Day 1 Inje Cocktail1361.0241368.593
Day 11 Inje Cocktail1653.4911779.717
Day 4 Inje Cocktail Plus Belatacept1577.0171632.455
Day 7 Inje Cocktail1671.1731679.693

[back to top]

Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Losartan With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population

AUC (0-T): area under the concentration curve from time 0 to the time of the last quantifiable concentration and AUC (INF) extrapolated to infinity were measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for losartan with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. Inje cocktail components (losartan) measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng*h/mL (Geometric Mean)
AUC (0-T) N=22, 21, 21, 20)AUC (INF) N=22, 21, 21, 20)
Day 1 Inje Cocktail332.191338.033
Day 11 Inje Cocktail332.400338.646
Day 4 Inje Cocktail Plus Belatacept336.531341.644
Day 7 Inje Cocktail337.487343.508

[back to top]

AUC(0-T) of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without Coadministration of Belatacept - PK Evaluable Population

Area under the plasma concentration-time curve from zero to the last time of the last quantifiable concentration [AUC(0-T)] was measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng*h/mL (Geometric Mean)
1'-hydroxy-midazolam (N=22, 21, 21, 20)E-3174 (N= 22,21,21,20)5-hydroxyomeprazole (N=22,21,21,20)dextrorphan (N=21,20,20,19)paraxanthine (N=17,19,18,18)
Day 1 Inje Cocktail28.91995997163620592
Day 11 Inje Cocktail33.62072995164421353
Day 4 Inje Cocktail Plus Belatacept31.81999967157021077
Day 7 Inje Cocktail32.52089929151920680

[back to top]

Plasma Half-Life (T-HALF) of the Inje Cocktail Components (Midazolam, Losartan, Omeprazole, Dextromethorphan, and Caffeine) With and Without Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of dextromethorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. T-HALF was measured in hours (h). (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionh (Mean)
Midazolam (N=22, 21, 21, 20)Losartan (N=22, 21, 21, 20)Omeprazole (N= 22, 19, 21, 18)Dextromethorphan (N=18, 17, 18, 15)Caffeine (N=22, 21, 21, 18)
Day 1 Inje Cocktail4.012.961.196.765.61
Day 11 Inje Cocktail4.052.871.366.706.03
Day 4 Inje Cocktail Plus Belatacept4.432.561.396.985.71
Day 7 Inje Cocktail4.273.301.286.336.17

[back to top]

Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Dextromethorphan With and Without the Coadministration of Belatacept - PK Evaluable Population

AUC(0-T): area under the plasma concentration-time curve from time zero to the time of the last quantifiable concentration and AUC (INF): AUC extrapolated to infinity, were measured as ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for dextromethorphan with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. The poor metabolizer of CYP2D6 was excluded from the statistical analysis. Inje cocktail components (dextromethorphan) measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng*h/mL (Geometric Mean)
AUC (0-T); N= 21, 19, 19, 19AUC (INF); N= 18, 17, 18, 15
Day 1 Inje Cocktail6.1766.220
Day 11 Inje Cocktail5.9496.359
Day 4 Inje Cocktail Plus Belatacept5.6515.455
Day 7 Inje Cocktail6.1956.412

[back to top]

Ratio of 1'-Hydroxy-Midazolam AUC(0-T) to Midazolam AUC(0-T) and 1'-Hydroxy-Midazolam AUC(INF) to Midazolam AUC(INF), Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC (INF)] With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (1'-hydroxy-midazolam) to parent (midazolam) ratio was corrected for molecular weight. AUC (0-T) and AUC (INF) measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionratio (Geometric Mean)
MR_AUC (0-T) ratio; N=22, 21, 21, 20MR_(INF) ratio; N=22, 21, 20, 20
Day 1 Inje Cocktail0.4210.428
Day 11 Inje Cocktail0.4720.478
Day 4 Inje Cocktail Plus Belatacept0.4470.460
Day 7 Inje Cocktail0.4950.479

[back to top]

Ratio of Paraxanthine (Cmax) to Caffeine (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (paraxanthine) to parent (caffeine) ratio was corrected for molecular weight. Cmax measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components and their metabolites were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionratio (Geometric Mean)
Day 1 Inje Cocktail0.303
Day 4 Inje Cocktail Plus Belatacept0.337
Day 7 Inje Cocktail0.333
Day 11 Inje Cocktail0.323

[back to top]

Ratio of E-3174 (Cmax) to Losartan (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (E-3174) to parent (losartan) ratio was corrected for molecular weight. Cmax measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionratio (Geometric Mean)
Day 1 Inje Cocktail2.31
Day 4 Inje Cocktail Plus Belatacept2.24
Day 7 Inje Cocktail2.26
Day 11 Inje Cocktail2.26

[back to top]

Ratio of 5-Hydroxyomeprazole (Cmax) to Omeprazole (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (5-hydroxyomeprazole) to parent (omeprazole) ratio was corrected for molecular weight. Cmax measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionratio (Geometric Mean)
Day 1 Inje Cocktail0.548
Day 4 Inje Cocktail Plus Belatacept0.471
Day 7 Inje Cocktail0.424
Day 11 Inje Cocktail0.478

[back to top]

Ratio of 5-Dextrorphan (Cmax) to Dextromethorphan (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (5-dextrorphan) to parent (dextromethorphan) ratio was corrected for molecular weight. Cmax measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionratio (Geometric Mean)
Day 1 Inje Cocktail280
Day 4 Inje Cocktail Plus Belatacept303
Day 7 Inje Cocktail275
Day 11 Inje Cocktail303

[back to top]

Ratio of 5-Dextrorphan AUC(0-T) to Dextromethorphan AUC(0-T) and 5-Dextrorphan AUC(INF) to Dextromethorphan AUC(INF), Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC (INF)] With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (5-dextrorphan ) to parent (dextromethorphan) ratio was corrected for molecular weight. AUC (0-T) and AUC (INF) measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionratio (Geometric Mean)
MR_AUC (0-T) ratio; N=21,20,20,19MR_AUC (INF) ratio; N=18,17,18,15
Day 1 Inje Cocktail201173
Day 11 Inje Cocktail193173
Day 4 Inje Cocktail Plus Belatacept200177
Day 7 Inje Cocktail177170

[back to top]

Ratio of 5-Hydroxyomeprazole AUC(0-T) to Omeprazole AUC(0-T) and 5-Hydroxyomeprazole AUC(INF) to Omeprazole AUC(INF) , Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC(INF)] With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (5-Hydroxyomeprazole) to parent (omeprazole) ratio was corrected for molecular weight. AUC (0-T) and AUC (INF) measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionratio (Geometric Mean)
MR_AUC (0-T) ratio; N=22,21,21,20MR_AUC (INF) ratio; N=22,19,21,17
Day 1 Inje Cocktail0.7000.705
Day 11 Inje Cocktail0.6100.679
Day 4 Inje Cocktail Plus Belatacept0.6090.558
Day 7 Inje Cocktail0.5510.553

[back to top]

Ratio of E-3174 AUC(0-T) to Losartan AUC(0-T) and E3174 AUC (INF) to Losartan AUC (INF) Corrected for Molecular Weight [MR_AUC(0-T), MR_AUC(INF)] With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (E-3174) to parent (losartan) ratio was corrected for molecular weight. AUC (0-T) and AUC (INF) measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionratio (Geometric Mean)
MR_AUC (0-T) ratio; N=22,21,21,20MR_AUC (INF) ratio; N=22,20,21,20
Day 1 Inje Cocktail5.816.02
Day 11 Inje Cocktail6.176.37
Day 4 Inje Cocktail Plus Belatacept5.755.99
Day 7 Inje Cocktail5.996.16

[back to top]

Ratio of Paraxanthine AUC(0-T) to Caffeine AUC(0-T) and Paraxanthine AUC (INF) to Caffeine AUC (INF), Corrected for Molecular Weight [MR_AUC(0-T) and MR_AUC (INF)] With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (paraxanthine) to parent (caffeine) ratio was corrected for molecular weight. AUC (0-T) and AUC (INF) measured in ng*h/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionratio (Geometric Mean)
MR_AUC(0-T) ratio; N=17,19,18,18MR_AUC(INF) ratio; N=7, 8, 6, 6
Day 1 Inje Cocktail0.6270.827
Day 11 Inje Cocktail0.6030.781
Day 4 Inje Cocktail Plus Belatacept0.6770.827
Day 7 Inje Cocktail0.6350.799

[back to top]

T-HALF of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without the Coadministration of Belatacept - PK Evaluable Population

Plasma half-life (T-HALF) was measured in hours (h). Samples for assessment of plasma concentrations of Inje cocktail components metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionh (Mean)
1'-Hydroxy-Midazolam (N=22,21,20,20)E-3174 (N=22, 20, 21, 20)5-Hydroxyomeprazole (N=22,20,21,17)Dextrorphan (N=21,20,20,19)Paraxanthine (N=7,8,6,6)
Day 1 Inje Cocktail5.294.861.434.267.33
Day 11 Inje Cocktail5.314.881.544.087.44
Day 4 Inje Cocktail Plus Belatacept6.354.571.554.366.86
Day 7 Inje Cocktail5.614.751.524.237.33

[back to top]

Time of Maximum Observed Plasma Concentration (Tmax) of the Inje Cocktail Components (Midazolam, Losartan, Omeprazole, Dextromethorphan, and Caffeine) With and Without Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of dextromethorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. Tmax was measured in hours (h). (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionh (Median)
Midazolam (N=22,21,21,20)Losartan (N= 22, 21, 21, 20)Omeprazole (N=22,21,21,20)Dextromethorphan (N=21, 19, 19, 19)Caffeine (N= 22,21,21,18)
Day 1 Inje Cocktail0.501.522.003.001.00
Day 11 Inje Cocktail1.01.503.003.001.50
Day 4 Inje Cocktail Plus Belatacept0.502.003.003.001.00
Day 7 Inje Cocktail0.501.503.003.001.50

[back to top]

Tmax of Inje Cocktail Component Metabolites (1'-Hydroxy-Midazolam, E-3174, 5-Hydroxyomeprazole, Dextrorphan, and Paraxanthine) With and Without the Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail components metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. The poor metabolizer of CYP2D6 was excluded from summary of Dextrorphan parameters. Inje cocktail components were each measured using HPLC with MS/MS detection. Time of maximum observed plasma concentration (Tmax) was measured in hours (h). (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionh (Median)
1'-Hydroxy-Midazolam (N=22,21,21,20)E-3174 (N=22, 21, 21, 20)5-Hydroxyomeprazole (N=22,21,21,20)Dextrorphan (N=21,20,20,19)Paraxanthine (N=17, 19, 18, 18)
Day 1 Inje Cocktail1.004.002.002.008.00
Day 11 Inje Cocktail1.004.002.532.008.00
Day 4 Inje Cocktail Plus Belatacept1.004.003.002.008.00
Day 7 Inje Cocktail1.004.003.002.008.00

[back to top]

Ratio of 1'-Hydroxy-Midazolam (Cmax) to Midazolam (Cmax), Corrected for Molecular Weight (MR_Cmax) With and Without Coadministration of Belatacept - PK Evaluable Population

Metabolite (1'-hydroxy-midazolam) to parent (midazolam) ratio was corrected for molecular weight. Cmax measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components and the metabolites of those components were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionratio (Geometric Mean)
Day 1 Inje Cocktail0.432
Day 4 Inje Cocktail Plus Belatacept0.476
Day 7 Inje Cocktail0.497
Day 11 Inje Cocktail0.490

[back to top]

Adjusted Geometric Mean Cmax of Omeprazole With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population

Cmax: Maximum observed plasma concentration was measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for omeprazole with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. Inje cocktail components (omeprazole) measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionng/mL (Geometric Mean)
Day 1 Inje Cocktail583.047
Day 4 Inje Cocktail Plus Belatacept734.776
Day 7 Inje Cocktail753.126
Day 11 Inje Cocktail686.929

[back to top]

Mean Change From Baseline in Heart Rate at Study Discharge (Day 46±2 Days)

Heart Rate was taken after the participant had been sitting quietly for at least 5 minutes and was measured in beats per minute (bpm). Hear rate was taken on Day 46 (day of discharge) during the follow up period. Baseline was defined as last non-missing result with a collection date-time less than the date-time of the first active dose of study drug. (NCT01766050)
Timeframe: Baseline and Day 46 ±2 days

Interventionbpm (Mean)
Post Treatment1.0

[back to top]

Adjusted Geometric Mean Maximum Drug Concentration (Cmax) of Midazolam With and Without the Coadministration of Belatacept - Pharmacokinetic (PK) Evaluable Population

Samples for the assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for midazolam with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. Cmax measured in nanograms per milliliter (ng/mL). Inje cocktail components (Midazolam) measured using High Performance Liquid Chromatography (HPLC) with Tandem Mass Spectrometry (MS/MS) Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionng/mL (Geometric Mean)
Day 1 Inje Cocktail24.376
Day 4 Inje Cocktail Plus Belatacept24.152
Day 7 Inje Cocktail22.211
Day 11 Inje Cocktail24.220

[back to top]

Adjusted Geometric Mean Cmax of Losartan With and Without the Coadministration of Belatacept - PK Evaluable Population

Cmax measured in ng/mL. Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) and their metabolites (1'-hydroxy-midazolam, E-3174, 5-hydroxyomeprazole, dextrorphan, and paraxanthine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Adjusted geometric mean for losartan with and without belatacept, along with adjusted geometric mean ratios for Days 4, 7, and 11 versus Day 1, respectively, are presented. Inje cocktail components (losartan) measured using HPLC with MS/MS Detection. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

Interventionng/mL (Geometric Mean)
Day 1 Inje Cocktail119.789
Day 4 Inje Cocktail Plus Belatacept118.663
Day 7 Inje Cocktail127.184
Day 11 Inje Cocktail126.461

[back to top]

Mean Change From Baseline in Sitting Systolic and Diastolic Blood Pressure - All Treated Participants

Systolic and Diastolic blood pressures were taken after the participant had been sitting quietly for at least 5 minutes and the pressures were measured in millimeters of mercury (mm Hg). Pressures were obtained at screening visit, Day -1, and at 0 hour (pre-dose), 0.5 hour (post dose), and 2 hours (post dose) on Days 1, 4, 7, 11. Baseline was defined as last non-missing result with a collection date-time less than the date-time of the first active dose of study drug. (NCT01766050)
Timeframe: Baseline and 0.5 and 2.0 hours Post Dose on Days 1, 4, 7, and 11

,,,
Interventionmm Hg (Mean)
Systolic 0.5 hour post dose (N=22,21,21,20)Systolic 2.0 hour post dose (N=22,21,21,20)Diastolic 0.5 hour post dose (N=22,21,21,20)Diastolic 2.0 hour post dose (N=22,21,21,20)
Day 1 Inje Cocktail-0.60.2-1.70.9
Day 11 Inje Cocktail-4.7-2.9-2.2-1.0
Day 4 Inje Cocktail Plus Belatacept0.12.0-0.2-2.0
Day 7 Inje Cocktail-4.9-2.7-2.2-3.4

[back to top]

Mean Change From Baseline in Systolic and Diastolic Blood Pressure at Study Discharge (Day 46±2 Days)

Systolic and Diastolic blood pressures were taken after the participant had been sitting quietly for at least 5 minutes and the pressures were measured in millimeters of mercury (mm Hg). Systolic and Diastolic blood pressures were taken on Day 46 (day of discharge from the study). Baseline was defined as last non-missing result with a collection date-time less than the date-time of the first active dose of study drug. (NCT01766050)
Timeframe: Baseline and Day 46 ±2 days

Interventionmm Hg (Mean)
Systolic (N=18)Diastolic (N=18)
Post Treatment-5.9-5.7

[back to top]

Number of Participants With Adverse Events (AEs), Serious AEs (SAEs), Deaths, and AEs Leading to Discontinuation - All Treated Participants

Adverse events were coded according to the Medical Dictionary for Regulatory Activities (MedDRA), version 15.1. AE=any new unfavorable symptom, sign, or disease or worsening of a preexisting condition that may not have a causal relationship with treatment. SAE=a medical event that at any dose results in death, persistent or significant disability/incapacity, or drug dependency/abuse; is life-threatening, an important medical event, or a congenital anomaly/birth defect; or requires or prolongs hospitalization. Events captured from Day 1 (pre-dose) to last day prior to discharge (Day 46 ±2). In the total group, a participant with an AE is only counted once (ie, data reflected in Days 1, 4, 7, and 11 below could be the same participant with an AE on multiple days of the study). (NCT01766050)
Timeframe: Day 1 to Day of discharge (Day 46±2)

,,,,
Interventionparticipants (Number)
Adverse EventsSAEsDeathsAEs leading to discontinuation
All Participants5001
Day 1 Inje Cocktail4000
Day 11 Inje Cocktail1000
Day 4 Inje Cocktail Plus Belatacept2000
Day 7 Inje Cocktail1001

[back to top]

Adjusted Geometric Mean AUC (0-T) and AUC (INF) of Caffeine With and Without the Coadministration of Belatacept - PK Evaluable Population

Samples for assessment of plasma concentrations of Inje cocktail components (midazolam, losartan, omeprazole, dextromethorphan and caffeine) were collected at time = 0 (predose), 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 12, and 24 hours relative to the dosing of belatacept on Day 4 and Inje cocktail dosing on Days 1, 4, 7, and 11, respectively. Inje cocktail components were each measured using HPLC with MS/MS detection. AUC (0-T) and AUC (INF) were measured as ng*h/mL. (NCT01766050)
Timeframe: Pre-dose to 24 hours after dose of the Inje Cocktail on Days 1, 4, 7 and 11

,,,
Interventionng*h/mL (Geometric Mean)
AUC (0-T); N= 22, 21, 21, 18AUC (INF); N= 22, 21, 21, 18
Day 1 Inje Cocktail37394.540084.1
Day 11 Inje Cocktail38407.041537.6
Day 4 Inje Cocktail Plus Belatacept35200.337647.8
Day 7 Inje Cocktail36853.440149.9

[back to top]

Change From Baseline in the Dyskinesia and Other PD Symptoms Score As Assessed by Patient Global Impression of Change (PGIC)

The PGIC was a 7-point scale used to assess treatment response as judged by the participant. The participant was asked to assess change in dyskinesia symptoms and change in overall PD symptoms (e.g., slowness, stiffness, balance) on a score range of 1, much improved; 2, improved; 3, minimally improved; 4, no change; 5, minimally worse; 6, worse; 7, much worse. The average PGIC score was calculated at each visit by treatment group. Baseline is Visit 1 (Day 1) or Visit 3 (Day 29); Post-Baseline is Visit 2 (Day 14) or Visit 4 (Day 42). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT01767129)
Timeframe: Baseline; Post-Baseline (Days 14 and 42)

,
Interventionscore on a scale (Least Squares Mean)
DyskinesiaOther PD symptoms
AVP-923-452.74.2
Placebo3.74.1

[back to top]

Change From Baseline in MDS-UDysRS Scores for Part 1 and 2

"The MDS-UDysRS was developed to evaluate involuntary movements often associated with treated PD. Part 1 (On-Dyskinesia ['jerking or twisting movements that occur when your medicine is working']) and Part 2 (Off-Dyskinesia [spasms or cramps that can be painful and occur when your PD medications are not taken or are not working]) of the MDS-UDysRS were assessed by a blinded rater. Parts 1B and 2B (participant questionnaires) were completed at home by the participant. Part 1 comprised of 11 items, and Part 2 comprised of 4 items. All items were assigned a score of: 0, normal; 1, slight; 2, mild; 3, moderate; 4, severe. The total score for Parts 1 and 2 ranged from 0 to 44 and from 0 to 16, respectively. Higher scores indicate increased symptom severity. Post-Baseline was Visit 2 (Day 14) or Visit 4 (Day 42). Change from Baseline was calculated as the post-Baseline value minus the Baseline value." (NCT01767129)
Timeframe: Baseline (Day 1); Post-Baseline (Day 14 or 42)

,
Interventionscore on a scale (Least Squares Mean)
Part 1Part 2
AVP-923-45-3.9-0.7
Placebo0.000.6

[back to top]

"Change From Baseline in PD Motor Diary Ratings Of Duration Of On-time Without Bothersome Dyskinesia"

The PD motor diary is a home diary used to assess functional status in participants with PD with motor fluctuations and dyskinesia. Participants were instructed to complete the PD motor diary at home for a minimum of 3 consecutive days within the 7 days prior to Visits 2 and 4 (Days 14 and 42). Baseline is Visit 1 (Day 1) or Visit 3 (Day 29); Post-Baseline is Visit 2 (Day 14) or Visit 4 (Day 42). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT01767129)
Timeframe: Baseline; Post-Baseline (Days 14 and 42)

Interventionhours (Least Squares Mean)
AVP-923-450.5
Placebo0.5

[back to top]

Change From Baseline in PDQ-39 Single Index (PDQ-39-SI) Scores at the End of Each Treatment Period

The PDQ-39 was the most widely used PD-specific measure of health status. It consists of 39 questions, covering 8 aspects of quality of life: mobility; activities of daily living; emotional wellbeing; stigma; social support; cognitive impairment (cognitions); communication; bodily discomfort. The instrument was developed on the basis of interviews with people diagnosed with PD. The PDQ-39-SI) score was calculated as the weighted addition of scores on all 8 dimensions of the PDQ-39. The total score ranged from 0 (no disease impact) to 100 (severe disease impact). Baseline was Visit 1 (Day 1) or Visit 3 (Day 29); Post-Baseline was Visit 2 (Day 14) or Visit 4 (Day 42). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT01767129)
Timeframe: Baseline; Post-Baseline (Day 14 or 42)

Interventionscore on a scale (Least Squares Mean)
AVP-923-45-4.2
Placebo2.2

[back to top]

Change From Screening in the Montreal Cognitive Assessment (MoCA) Calculated Score at the End of Each Treatment Period

The MoCA was designed as a rapid screening instrument for mild cognitive dysfunction. It assesses different cognitive domains: attention and concentration, executive functions, memory, language, visuoconstructional skills, conceptual thinking, calculations, and orientation. The total score is calculated by summing the items from each domain. Total scores can range from 0 to 30; lower scores indicate cognitive dysfunction. A final total score of 26 and above is considered normal. Change from Screening was calculated as the post-Screening value minus the Screening value. (NCT01767129)
Timeframe: Screening (Day -28); End of each treatment period (Days 14 or 42)

Interventionscore on a scale (Least Squares Mean)
AVP-923-450.1
Placebo0.6

[back to top]

Least Squares Mean Disability Area Under the Curve (AUC) Score As Assessed By Modified Movement Disorder Society-Unified Dyskinesia Rating Scale (MDS-UDysRS) Part 4

The MDS-UDysRS was developed to evaluate involuntary movements often associated with treated PD. Part 4 of the MDS-UDysRS objectively measures the disability associated with levodopa-induced dyskinesia. Disability was assessed via video analysis by unbiased blinded central raters. Disability was evaluated for communication, drinking from a cup, and ambulation items. Each item was rated from 0 (no dyskinesia) to 4 (most severe disability), with a sum range of 0 to 12. A score of '0' was assigned to questions associated with the dressing task because it was not performed due to placement of the IV line. A higher score indicated more severe symptoms. (NCT01767129)
Timeframe: Over the 2-hour levodopa infusion period on the last day of each treatment period (Day 14 and Day 42)

Intervention(Score on a scale)*hour (Least Squares Mean)
AVP-923-45409.4
Placebo428.3

[back to top]

Least Squares Mean Dyskinesia Severity Area Under the Curve (AUC) Score As Assessed By Modified Movement Disorder Society-Unified Dyskinesia Rating Scale (MDS-UDysRS) Part 3

The MDS-UDysRS was developed to evaluate involuntary movements often associated with treated Parkinson's disease (PD). Levodopa-Induced Dyskinesia severity was assessed via video analysis by unbiased blinded central raters, and was calculated using the Intensity Scale from Part 3 of the MDS-UDysRS. The Intensity Scale was made up of seven body parts: face, neck, right arm/shoulder, left arm/shoulder, trunk, right leg/hip, and left leg/hip. Each body part was scored on a variety of disability items (communication, drinking, and ambulation [walking]) on a scale of 0 (normal) to 4 (incapacitating dyskinesia) with a maximum total score of 28. For each body part, the highest disability score was summed to calculate the intensity score. A score of '0' was assigned to questions associated with the dressing task which were not performed due to the placement of the treatment infusion (IV) line. A higher score indicated more severe symptoms. (NCT01767129)
Timeframe: Over the 2-hour levodopa infusion period on the last day of each treatment period (Day 14 and Day 42)

Intervention(Score on a scale)*hour (Least Squares Mean)
AVP-923-45966.0
Placebo1049.4

[back to top]

Least Squares Mean Motor Movement Area Under the Curve Score As Assessed by Modified Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS), Part III

The Modified MDS-UPDRS (4 parts) was used to assess the severity of parkinsonian disability. Part III of the MDS-UPDRS was used as an objective measure of the severity of parkinsonian disability per motor examination with focus on the treated side tremor and motor fluctuations. The motor examination were videotaped using a standardized protocol for review by an expert central rater blinded to the treatment schedule. A total of 11 items were scored as follows: 0 (normal), 1 (slight), 2 (mild), 3 (moderate), 4 (severe), with a maximum total score of 44. For each part, a higher score indicated more severe symptoms. Due to placement of the IV line, a score of '0' was assigned to rigidity questions. (NCT01767129)
Timeframe: Over the 2-hour levodopa infusion period on the last day of each treatment period (Day 14 and Day 42)

Intervention(Score on a scale)*hour (Least Squares Mean)
AVP-923-456568.9
Placebo6397.0

[back to top]

Least Squares Mean Timed Finger Tapping Area Under the Curve (AUC) Score

Timed finger tapping test was used to quantify the upper extremity impairment in participants with idiopathic PD. Timed finger tapping was assessed using the Objective Parkinson's Disease Measurement (OPDM) System. Participants were instructed to tap for 60 seconds while speed and accuracy were assessed across 4 tests; right and left two finger test (m,n keystrokes) and one finger test (p,q keystrokes). The mean peak finger tapping score was calculated using the individual peak values. A higher score signifies improvement (faster typing, more accuracy), while a lower score signifies increased symptom severity. (NCT01767129)
Timeframe: Over the 2-hour levodopa infusion period on the last day of each treatment period (Day 14 and Day 42)

Intervention(Score on a scale)*hour (Least Squares Mean)
AVP-923-45820.9
Placebo772.1

[back to top]

Change From Baseline in MDS-UPDRS Scores for Part I, II, and IV

The MDS-UPDRS was used to assess the status of PD. Parts I and II (completed by the participant) and Part IV (completed by a rater) of the MDS-UPDRS provided a subjective measure of parkinsonian disability. Part I measured non-motor experiences of daily living, Part II measured motor experiences of daily living, and Part IV measures motor complications associated with PD. Each part was comprised of a series of questions, and each question was scored from 0 (normal) to 4 (severe). Part I and Part II were each comprised of 13 items; the total score ranges from 0 (normal) to 52 (severe). Part III was comprised of 33 items; the total score ranges from 0 (normal) to 132 (severe). Part IV was comprised of 6 items; the total score ranged from 0 (normal) to 24 (severe). For each part, a higher score indicated more severe symptoms. Post-Baseline was Visit 2 (Day 14) or Visit 4 (Day 42). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT01767129)
Timeframe: Baseline (Day 1); Post-Baseline (Day 14 or 42)

,
Interventionscore on a scale (Least Squares Mean)
Part IPart IIPart IV
AVP-923-45-1.2-0.0-2.9
Placebo-0.1-0.2-0.5

[back to top]

Change From Baseline in Parkinson's Disease Questionnaire-39 (PDQ-39) Domain Scores at the End of Each Treatment Period

The PDQ-39 was a self-reported questionnaire consisting of 39 questions assessing Parkinson's disease-specific health quality of life covering 8 aspects of quality of life: mobility; activities of daily living; emotional wellbeing; stigma; social support; cognitive impairment (cognitions); communication; bodily discomfort. Each item was scored on 5-point scale: 0 = Never (better in outcome), 1 = Occasionally, 2 = Sometimes, 3 = Often, 4 = Always (worse in outcome). Total scores ranged between 0 to 156. Higher scores indicated poor quality of life. Baseline was Visit 1 (Day 1) or Visit 3 (Day 29); Post-Baseline was Visit 2 (Day 14) or Visit 4 (Day 42). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. (NCT01767129)
Timeframe: Baseline; Post-Baseline (Day 14 or 42)

,
Interventionscore on a scale (Least Squares Mean)
MobilityActivities of Daily LivingEmotional Well BeingStigmaSocial supportCognitive impairment (Cognitions)CommunicationBodily discomfort
AVP-923-450.2-4.6-4.5-3.7-6.0-4.1-3.7-7.4
Placebo2.03.92.90.33.80.22.4-2.2

[back to top]

Percentage of Participants With Clinical Global Impression-Change (CGI-C) Score at Day 90

CGI-C, an investigator-assessed scale was used to measure the overall treatment response. CGI-C, a 7-point (1-7) scale was rated as: very much improved, much improved, minimally improved, no change, minimally worse, much worse, or very much worse. Data is presented as percentage of participants with CGI-C score at Day 90. Percentages within a measure may not sum to 100.0 due to rounding. Percentages use the count of participants with non-missing data as the denominator. (NCT01799941)
Timeframe: Day 90 (Final visit)

,,,
InterventionPercentage of participants (Number)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVeru much worse
Dementia30.447.113.78.80.00.00.0
Overall33.742.913.48.80.80.40.0
Stroke33.041.814.37.72.21.10.0
Traumatic Brain Injury39.738.211.810.30.00.00.0

[back to top]

Percentage of Participants With Patient Global Impression-Change (PGI-C) Score at Day 90

PGI-C, a participant/participant's caregiver-assessed scale was used to measure participant overall treatment response. PGI-C, a 7-point (1-7) scale was rated as: very much improved, much improved, minimally improved, no change, minimally worse, much worse, or very much worse. Data is presented as percentage of participants with PGI-C score at Day 90. Percentages within a measure may not sum to 100.0 due to rounding. Percentages use the count of participants with non-missing data as the denominator. (NCT01799941)
Timeframe: Day 90 (Final visit)

,,,
InterventionPercentage of participants (Number)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVeru much worse
Dementia28.448.014.77.81.00.00.0
Overall32.639.818.88.00.40.40.0
Stroke33.733.721.710.90.00.00.0
Traumatic Brain Injury37.335.820.94.50.01.50.0

[back to top]

Percentage of Participants With PBA Remission

PBA remission was defined as participants with one or more episodes reported at the baseline (Day 1) visit and zero episodes reported at the Day 30 (Visit 1) or Day 90 (Final visit). Data is reported as percentage of participants with no reported episodes over the previous 7 days (prior to visit) at Day 30 (Visit 1) and Day 90 (Final visit). (NCT01799941)
Timeframe: Day 30 (Visit 1) and Day 90 (Final visit)

,,,
InterventionPercentage of participants (Number)
Day 30 (n= 296, 108, 102, 86)Day 90 (n= 260, 102, 92, 66)
Dementia13.031.4
Overall20.335.4
Stroke22.534.8
Traumatic Brain Injury26.742.4

[back to top]

Percentage of Participants With Treatment Satisfaction Survey

The treatment satisfaction survey was a 5 point single question survey that was administered by the site staff to the participant/participant's caregiver. Participants were asked to rate their response to treatment satisfaction as: very dissatisfied, somewhat dissatisfied, neither satisfied nor dissatisfied, somewhat satisfied, and very satisfied. Data is presented as percentage of participants with treatment satisfaction at Day 90. Percentages within a measure may not sum to 100.0 due to rounding. Percentages use the count of participants with non-missing data as the denominator. (NCT01799941)
Timeframe: Day 90 (Final visit)

,,,
InterventionPercentage of participants (Number)
Very dissatisfiedSomewhat dissatisfiedNeither satisfied nor dissatisfiedSomewhat satisfiedVery satisfied
Dementia4.96.913.721.652.9
Overall7.75.411.528.047.5
Stroke12.05.48.731.542.4
Traumatic Brain Injury6.03.011.932.846.3

[back to top]

Mean Change From Baseline in Center for Neurologic Study-Lability Scale (CNS-LS) Score at Day 30

The CNS-LS was a seven-item, self-administered questionnaire, completed by the participant or participant's caregiver that provided a quantitative measure of the perceived frequency and severity of Pseudobulbar Affect (PBA) episodes. It consisted of two subscales measuring labile laughter (four items) and labile crying (three items). Each item was rated on a scale from 1 (applies never) to 5 (applies most of the time). The total score was calculated as the sum of the item values that resulted in a score ranging from 7 (no symptoms) to 35 (maximum symptom severity and frequency). A single continuous variable was created for the reported time point. The change in CNS-LS was calculated as the score from the Day 30 assessment minus the Baseline CNS-LS measure. A negative change represented a decrease in CNS-LS score over time following the baseline assessment indicating a perceived decrease in frequency and severity of PBA episodes. (NCT01799941)
Timeframe: Day 30

InterventionUnits on a scale (Mean)
Overall-5.43
Dementia-4.60
Stroke-6.17
Traumatic Brain Injury-5.58

[back to top]

Mean Change From Baseline in Center for Neurologic Study-Lability Scale (CNS-LS) Score at Day 90

The CNS-LS was a seven-item, self-administered questionnaire, completed by the participant or participant's caregiver that provided a quantitative measure of the perceived frequency and severity of Pseudobulbar Affect (PBA) episodes. It consisted of two subscales measuring labile laughter (four items) and labile crying (three items). Each item was rated on a scale from 1 (applies never) to 5 (applies most of the time). The total score was calculated as the sum of the item values that resulted in a score ranging from 7 (no symptoms) to 35 (maximum symptom severity and frequency). A single continuous variable was created for the reported time point. The change in CNS-LS was calculated as the score from the Day 90 assessment minus the Baseline CNS-LS measure. A negative change represented a decrease in CNS-LS score over time following the baseline assessment indicating a perceived decrease in frequency and severity of PBA episodes. (NCT01799941)
Timeframe: Day 90 (Final visit)

InterventionUnits on a scale (Mean)
Overall-7.69
Dementia-7.22
Stroke-7.59
Traumatic Brain Injury-8.54

[back to top]

Mean Change From Baseline in Quality of Life Visual Analog Scale (QOL-VAS) Score at Day 90

"The QOL-VAS, a participant reported scale of quality of life (QOL) was used to measure the impact of PBA episodes on the participant's QOL over the previous 7 days (prior to visit) at Baseline (Day 1) and Day 90 (Final visit). The assessment was completed by a participant placing a mark on a horizontal line that extends from 0 not (affected) at all to 10 significantly (affected). The participant's mark was measured and recorded at each time point. The change in QOL-VAS score from baseline to day 90 visit, defined as the day 90 score minus the baseline score, was analyzed. Data is reported as mean QOL-VAS score; a positive change in score represented an increase in participant's quality of life." (NCT01799941)
Timeframe: Day 90 (Final visit)

InterventionUnits on a scale (Mean)
Overall-3.13
Dementia-3.20
Stroke-2.66
Traumatic Brain Injury-3.67

[back to top]

Mean Pseudobulbar Affect (PBA) Episode Count Per Week by Visit

PBA episode count was an investigator assessed measure in which the participant/participant's daytime caregiver was asked to identify, count and recall the total episodes of exaggerated/uncontrollable laughing or crying over the previous 7 days (prior to visit) at Baseline (Day 1), Day 30 (Visit 1), and Day 90 (Final visit). The response categories for this question were: 0, 1- 2, 3-5, 6-10, >10. The original responses from participants were converted to estimate the continuous number of PBA episodes by taking the mid-point of the original response ranges and multiplying that value by 7. Data is presented as mean PBA count per week. (NCT01799941)
Timeframe: Baseline (Day 1), Day 30 (Visit 1), and Day 90 (Final visit)

,,,
InterventionCount/week (Mean)
BaselineDay 30 (n= 296, 108, 102, 86)Day 90 (n= 260, 102, 92, 66)
Dementia25.6812.858.41
Overall21.339.106.23
Stroke19.626.955.26
Traumatic Brain Injury17.946.944.20

[back to top]

Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

AEs (defined as any untoward medical occurrence in a clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product) and SAEs (defined as any untoward medical occurrence that at any dose resulted in death, was life-threatening [ie, the participant was at immediate risk of death from the AE as it occurred; this did not include an event that, had it occurred in a more severe form or was allowed to continue, might have caused death], required inpatient hospitalization or prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity, or was as a congenital anomaly/birth defect (in the child of a participant who was exposed to the study drug) were assessed during the study. (NCT01799941)
Timeframe: From signing of informed consent up to 30 days after receiving the last dose of study drug or up to approximately 120 days

,,,
InterventionParticipants (Number)
AEsSAEs
Dementia4914
Overall13223
Stroke405
Traumatic Brain Injury434

[back to top]

Percentage Change From Baseline in PBA Episode Count Per Week

The change from the baseline PBA rate was measured using Mixed Effects Poisson Regression Model (adjusted for gender and age [≤ 65 years]). (NCT01799941)
Timeframe: Day 30 (Visit 1) and Day 90 (Final visit)

,,,
Interventionpercent change (Mean)
Day 30 (n= 296, 108, 102, 86)Day 90 (n=260, 102, 92, 66)
Dementia-50.0-67.7
Overall-57.5-72.3
Stroke-64.9-74.5
Traumatic Brain Injury-61.3-78.5

[back to top]

Percentage of Participants With ≥ 50% Reduction in PBA Episode Count Per Week

Data is reported as the percentage of participants with ≥ 50% reduction in PBA episode count/week. (NCT01799941)
Timeframe: Day 30 (Visit 1) and Day 90 (Final visit)

,,,
InterventionPercentage of participants (Number)
Day 30 (n= 289, 107, 100, 82)Day 90 (n= 254, 101, 90, 63)
Dementia58.976.2
Overall63.778.0
Stroke65.076.7
Traumatic Brain Injury68.382.5

[back to top]

Percentage of Participants With ≥ 75% Reduction in PBA Episode Count Per Week

Data is reported as the percentage of participants with ≥ 75% reduction in PBA episode count/week. (NCT01799941)
Timeframe: Day 30 (Visit 1) and Day 90 (Final visit)

,,,
InterventionPercentage of participants (Number)
Day 30 (n= 289, 107, 100, 82)Day 90 (n= 254, 101, 90, 63)
Dementia33.657.4
Overall42.257.1
Stroke42.047.8
Traumatic Brain Injury53.769.8

[back to top]

Visual Analog Scale - Swallowing Score

Visual analog scales are useful for measuring complex clinical events and offer the advantage of self-administration and responsiveness to change over time. The scales designed for this study inventory three domains of bulbar function: speech, swallowing and salivation (sialorrhea). For each of these, subjects score themselves by indicating their level of function on a scale of 1 (severe impairment) to 10 (normal). Scores range from 1 to 10; the higher the score, the more normal the function. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)7.23
Matching Placebo6.93

[back to top]

Average Water Swallowing Test (WST)

The Water Swallowing Test (WST) estimates swallowing speed, a useful and reproducible measure. While sitting, subjects are asked to drink 30 milliliters (mL) of liquid. The time for subjects to complete this task is a sensitive measure for the detection of swallowing dysfunction and is a simple measure for serial assessment of subjects. The test will be completed three times, with the best two scores recorded to obtain an average score. Following completion of the WST, the subject's swallowing abilities (choking, spillage, and effort) will be observed. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionseconds (Least Squares Mean)
Active Drug (Nuedexta)12.16
Matching Placebo13.11

[back to top]

Average Solids Swallowing Test

The Time Swallowing Test assesses the subject's ability to swallow solids. For this test, the subject will be asked to consume a tablespoon of cereal containing 5 cheerios. The subject will be instructed to close their mouth, chew and subsequently swallow the bolus. The time to complete this task will be recorded. The test will be completed three times to obtain an average score. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionseconds (Least Squares Mean)
Active Drug (Nuedexta)18.53
Matching Placebo19.45

[back to top]

Ashworth Spasticity Scale Score - Right Leg

This is a standard measure for spasticity that has been used in numerous ALS clinical trials to assess spasticity due to upper motor neuron dysfunction in ALS. Data is generated from the clinical exam and scored from 1-5, the lowest score indicating normal tone and the highest muscle rigidity. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)1.94
Matching Placebo1.82

[back to top]

Ashworth Spasticity Scale Score - Right Arm

This is a standard measure for spasticity that has been used in numerous ALS clinical trials to assess spasticity due to upper motor neuron dysfunction in ALS. Data is generated from the clinical exam and scored from 1-5, the lowest score indicating normal tone and the highest muscle rigidity. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)1.65
Matching Placebo1.53

[back to top]

Ashworth Spasticity Scale Score - Left Leg

This is a standard measure for spasticity that has been used in numerous ALS clinical trials to assess spasticity due to upper motor neuron dysfunction in ALS. Data is generated from the clinical exam and scored from 1-5, the lowest score indicating normal tone and the highest muscle rigidity. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)1.91
Matching Placebo1.91

[back to top]

ALS Functional Rating Scale- Revised (ALSFRS-R) Total Score

The ALSFRS-R is a quickly administered (5 min) ordinal rating scale used to determine a subject's assessment of their capability and independence in 12 functional activities. There are 12 questions, graded by the subject 0-4 (4 is normal). Score of 0 (worst) to 48 (best). Reflects speech and swallowing, fine motor skills, large motor skills, and breathing. (NCT01806857)
Timeframe: Average between Screening Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)34.15
Matching Placebo33.7

[back to top]

Ashworth Spasticity Scale Score - Left Arm

This is a standard measure for spasticity that has been used in numerous ALS clinical trials to assess spasticity due to upper motor neuron dysfunction in ALS. Data is generated from the clinical exam and scored from 1-5, the lowest score indicating normal tone and the highest muscle rigidity. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)1.62
Matching Placebo1.67

[back to top]

Visual Analog Scale - Speech Scores

Visual analog scales are useful for measuring complex clinical events and offer the advantage of self-administration and responsiveness to change over time. The scales designed for this study inventory three domains of bulbar function: speech, swallowing and salivation (sialorrhea). For each of these, subjects score themselves by indicating their level of function on a scale of 1 (severe impairment) to 10 (normal). Scores range from 1 to 10; the higher the score, the more normal the function. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)4.97
Matching Placebo4.11

[back to top]

Visual Analog Scale - Salivation (Sialorrhea) Score

Visual analog scales are useful for measuring complex clinical events and offer the advantage of self-administration and responsiveness to change over time. The scales designed for this study inventory three domains of bulbar function: speech, swallowing and salivation (sialorrhea). For each of these, subjects score themselves by indicating their level of function on a scale of 1 (severe impairment) to 10 (normal). Scores range from 1 to 10; the higher the score, the more normal the function. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)6.78
Matching Placebo6.78

[back to top]

Timed Reading of Test Paragraph Result

Subjects will be asked to read 'The Rainbow Passage' a commonly used test paragraph utilized by speech pathologists to assess speech rate (words/minute). Study staff will time the subject to determine how many words the subject reads per minute. It is used primarily because it contains every sound in the English language. Subjects will also be observed for loudness, nasality, and intelligibility. (NCT01806857)
Timeframe: Average between Baseline Visit to Visit 3

Interventionwords per minute (Least Squares Mean)
Active Drug (Nuedexta)107.12
Matching Placebo103.37

[back to top]

Center for Neurologic Study - Lability Scale (CNS-LS) Total Score

The Center for Neurologic Study-Lability Scale (CNS-LS) is a 7-item self report scale that assesses pseudobulbar affect (PBA) by measuring the perceived frequency of PBA episodes (laughing or crying). Each item is scored using a 5-point Likert scale, from 1 (applies never) to 5 (applies most of the time). Scores range from 5-35. The higher the score, the worse the PBA. (NCT01806857)
Timeframe: Average between Screening Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Nuedexta)10.79
Matching Placebo13.72

[back to top]

Bulbar Function Scale (CNS-BFS) Total Score

"The Center for Neurologic Study-Bulbar Function Scale (CNS-BFS) is a 21-item self report scale that assesses three domains of bulbar function: speech, swallowing and salivation. Scores for each question range from 1 (does not apply) to 5 (applies most of the time). The higher the score, the worse the speech, swallowing and salivation (sialorrhea). [Range of score: 21-105]~The scale was modeled on the Center for Neurologic Study Emotional Lability Scale (CNS-LS) that has been a robust endpoint in four clinical trials. The scale was validated in a large population of ALS patients (n=122) and detects impaired bulbar function at a sensitivity of 90% and a specificity of 0.97%. Test re-test correlation was 0.92% at six-months (n=53)." (NCT01806857)
Timeframe: Average between Screening Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Neudexta)53.45
Matching Placebo59.31

[back to top]

Bulbar Function Scale (CNS-BFS) Swallowing Score

The Center for Neurologic Study-Bulbar Function Scale (CNS-BFS) is a 21-item self report scale that assesses three domains of bulbar function: speech, swallowing and salivation. Scores for each question range from 1 (does not apply) to 5 (applies most of the time). The higher the score, the worse the swallowing. There are 7 swallowing questions, with a score range of 7 to 35. (NCT01806857)
Timeframe: Average between Screening Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Neudexta)17.14
Matching Placebo18.92

[back to top]

Bulbar Function Scale (CNS-BFS) Speech Score

The Center for Neurologic Study-Bulbar Function Scale (CNS-BFS) is a 21-item self report scale that assesses three domains of bulbar function: speech, swallowing and salivation. Scores for each question range from 1 (does not apply) to 5 (applies most of the time). The higher the score, the worse the speech. There are 7 speech questions, with a score range of 7 to 35. (NCT01806857)
Timeframe: Average between Screening Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Neudexta)22.22
Matching Placebo24.57

[back to top]

Bulbar Function Scale (CNS-BFS) Sialorrhea Score

The Center for Neurologic Study-Bulbar Function Scale (CNS-BFS) is a 21-item self report scale that assesses three domains of bulbar function: speech, swallowing and salivation. Scores for each question range from 1 (does not apply) to 5 (applies most of the time). The higher the score, the worse the salivation (sialorrhea). There are 7 salivation (sialorrhea) questions, with a score range of 7 to 35. (NCT01806857)
Timeframe: Average between Screening Visit to Visit 3

Interventionunits on a scale (Least Squares Mean)
Active Drug (Neudexta)14.28
Matching Placebo15.81

[back to top]

Beck Scale for Suicidal Ideation (BSI)

Mean change in Brief Inventory Symptom from baseline to week 10. The BSS is a self-report 19-item scale preceded by five screening items. The BSS and its screening items are intended to assess a patient's thoughts, plans and intent to commit suicide. All 24 items are rated on a three-point scale (0 to 2). In this study, scores from the five screening items were included in the overall score. Therefore, total scores could range from 0 to 48, with higher scores reflecting more severe symptoms. (NCT01882829)
Timeframe: Baseline and Week 10

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-2.5

[back to top]

Sheehan Disability Scale

The Sheehan Disability Scale (SDS) is a self-rated scale which assesses illness-related disability in three areas of functioning: work, social and family. The SDS assess disability or functional impairment across three domains: work/school, social life/leisure activities and family life/home responsibilities. Each domain is scored from 0 (not at all) to 10 (very severely). The three domains can be summarized to evaluate global functional impairment by adding the scores of each of the three domains, resulting in global SDS score ranges from 0 (unimpaired) to 30 (highly impaired). (NCT01882829)
Timeframe: At baseline and Visit 6 (week 10)

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-4.3

[back to top]

Range of Impaired Functioning Tool

"The Range of Impaired Functioning Tool a brief scale for assessing functional impairment related to medical or psychiatric illness and has been demonstrated to possess good psychometric properties. The LIFE-RIFT has a total score and individual domain scores for the following areas of functioning: household duties, work, recreation, relationships with family, relationships with friends, schoolwork, and global life satisfaction (the satisfaction item is patient rated).~Higher scores indicate poorer functioning; scores ≥2 reflect impaired functioning in that domain. Results are reported for the total sum with full range from 3 (no impairment) to 60 (severe impairment), which is based on all individual domain scores." (NCT01882829)
Timeframe: At baseline and Visit 6 (week 10)

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-3.7

[back to top]

Quick Inventory of Depressive Symptomatology, Self Report (QIDS-SR)

The Quick Inventory of Depressive Symptomatology, Self Report (QIDS-SR) is a 16-item self rated instrument designed to assess the severity of depressive symptoms (30). The 16 items cover the nine symptom domains of major depression, and are rated on a scale of 0-3. Total score ranges from 0 to 27, with ranges of 0-5 (normal), 6-10 (mild), 11-15 (moderate), 16-20 (moderate to severe), and 21+ (severe). (NCT01882829)
Timeframe: up to 12 weeks

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-5.9

[back to top]

Quality of Life Enjoyment and Satisfaction Questionnaire Short Form

The Quality of Life Enjoyment and Satisfaction Questionnaire Short Form is a reliable and valid self-report measure designed to obtain sensitive measures of the degree of enjoyment and satisfaction experienced by individuals. The raw total score ranges from 14 to 70. Higher scores reflect better oucomes. (NCT01882829)
Timeframe: At baseline and Visit 6 (week 10)

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)0.1

[back to top]

Patient Rated Inventory of Side Effects (PRISE)

Frequency of observed adverse events over the study treatment period as captured by the PRISE. The Patient Rated Inventory of Side Effects (PRISE) assesses the presence of treatment side effects in nine organ/function systems (gastrointestinal, nervous system, heart, eyes/ears, skin, genital/urinary, sleep, sexual functioning, and other). (NCT01882829)
Timeframe: up to 12 weeks

Interventionevents (Number)
Nuedexta (Dextromethorphan/Quinidine)25

[back to top]

Montgomery-Asberg Depression Rating Scale

The Montgomery-Asberg Depression Rating Scale is a 10-item instrument used for the evaluation of depressive symptoms in adults and for the assessment of any changes to those symptoms. Each of the 10 items is s scored 0 (normal) to 6 (severe depression) with overall score ranges from 0 (normal) to 60 (severe depression). Primary outcome is change in MADRS at Visit 6 (Week 10). Higher values represent a worse outcome. (NCT01882829)
Timeframe: At baseline and visit 6 (week 10)

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-13.0

[back to top]

Massachusetts General Hospital Cognitive and Physical Functioning Questionnaire (CPFQ)

The Massachusetts General Hospital Cognitive and Physical Functioning Questionnaire (CPFQ) is a brief scale to measure cognitive and executive dysfunction in mood and anxiety disorders, and possesses good reliability and validity. The Massachusetts General Hospital CPFQ was developed to assess each of the 7 most common complaints of depressed patients reporting fatigue or cognitive/executive problems. The CPFQ consists of 7 questions, each rated on a scale from 1 to 6, with 1 indicating greater than normal functioning, 2 indicating normal functioning and with higher numbers indicating poorer functioning. Total score range from 7 (greater than normal function) to 42 (poor function). (NCT01882829)
Timeframe: At baseline and Visit 6 (week 10)

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-4.1

[back to top]

HAM-A

Change in Hamilton Anxiety Rating Scale (HAM-A) score from baseline to Week 10. It consists of 14 items, each defined by a series of symptoms. Each item is rated on a 5-point scale, ranging from 0 (not present) to 4 (severe), with a total score range of 0-56, where <17 indicates mild severity, 18-24 mild to moderate severity and >25-30 moderate to severe. (NCT01882829)
Timeframe: Baseline and Week 10

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-5.7

[back to top]

Columbia-Suicide Severity Rating Scale (C-SSRS)

The Columbia-Suicide Severity Rating Scale (C-SSRS) is a comprehensive, semi-structured interview measure that uniquely measures the full spectrum of suicidality including passive and active suicidal ideation, suicidal intent as well as suicidal behaviors. Full range from 0 (low intensity suicidal ideation to 9 (high intensity suicidal ideation). (NCT01882829)
Timeframe: up to 12 weeks

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)0.1

[back to top]

Clinical Global Impression (CGI) Scale

The Clinical Global Impression (CGI) scale assesses overall treatment response in psychiatric patients and has good reliability and validity metrics. The administration time is 2 minutes. This scale consists of three items: Severity of Illness (item 1); Global Improvement (item 2); and Efficacy Index (item 3). Item 1 is rated on a seven-point scale (1 = normal, 7 = among the most extremely ill patients) as is item 2 (1 = very much improved, 7 = very much worse). Full scale is 1 to 14. (NCT01882829)
Timeframe: up to 12 weeks

Interventionchange in units on a scale (Mean)
Nuedexta (Dextromethorphan/Quinidine)-1.5

[back to top]

Change in Cough Reflex Sensitivity to Capsaicin

increase in C5 (decrease in cough reflex sensitivity). Capsaicin cough challenge involves subjects breathing in incremental doubling concentrations of aerosolized capsaicin, 1 minute apart, until the concentration of capsaicin (micromolar) inducing 5 or more coughs (C5) is reached. (NCT02062710)
Timeframe: 2 hours after study drug administration

Interventionlog C5 (uM) (Mean)
Diphenhydramine/Phenylephrine/Cocoa0.97
Dextromethorphan0.80
Placebo0.57

[back to top]

Glasgow Coma Scale (GCS)

Glasgow Coma Scale (GCS) is assessed by physical neurological examination of the subject by a qualified neurologist. GSC is a common scoring system used to describe the level of consciousness in a person following a traumatic brain injury. The initial score correlates with the severity of brain injury and prognosis. It estimates Coma severity based on Eye (4), Verbal (5), and Motor (6) criteria with the following total score of between 3 (indicating deep unconsciousness) and 15 (indicating no issues). (NCT02142712)
Timeframe: Baseline

InterventionGlasgow Coma Scale (Number)
Famotidine15
Pantoprazole15
Diphenhydramine15

[back to top]

National Institutes of Health Stroke Severity (NIHSS) Scale

NIHSS is a tool used by healthcare providers to objectively quantify the degree of impairment caused by a stroke. It is composed of 11 items. Each item scores a specific ability between a score of 0-4. Usually, for each item, a score of 0 indicates normal function in that specific ability, while a higher score indicates some level of impairment. The individual scores from each item are added together to calculate a patient's total NIHSS score. The maximum possible score is 42, with the minimum score being a 0. (NCT02142712)
Timeframe: Baseline

InterventionNIHSS scale (Number)
Famotidine3
Pantoprazole3
Diphenhydramine0

[back to top]

Adverse Events

Compare the number of adverse events in the Nuedexta arm vs. the placebo arm. (NCT02176018)
Timeframe: Baseline (Day 0) to Treatment Period Month 3 (Day 116)

InterventionNumber of Adverse Events (Number)
Nuedexta9
Placebo35

[back to top]

Headache Days in Treatment Period Month 3

The average number of headache days at treatment period month 3 (28 day period) for the Nuedexta arm and the Placebo arm. Headache days were defined as any patient reported head pain during the prior 24 hour period. (NCT02176018)
Timeframe: End of Treatment Period Month 3 (Day 116)

InterventionNumber of headache days (Mean)
Nuedexta7.1
Placebo9.77

[back to top]

50% Headache Reduction

The number of subjects with at least a 50% reduction in number of headache days comparing baseline to each visit (treatment period months 1, 2, and 3: 28 day for each month) in the Nuedexta arm vs. the placebo arm. (NCT02176018)
Timeframe: Baseline (Day 0) to Treatment Period Month 3 (Day 116)

,
InterventionParticipants (Count of Participants)
Treatment Month 1Treatment Month 2Treatment Month 3
Nuedexta678
Placebo028

[back to top]

Headache Days in Each Treatment Period Month

The average number of headache days at treatment period months 1, 2, and 3 (28 day periods for each month) for the Nuedexta arm and the Placebo arm. Headache days were defined as any patient reported head pain during the prior 24 hour period. (NCT02176018)
Timeframe: Treatment Month 1, Treatment Month 2, and End of Treatment Period Month 3 (Day 116)

,
InterventionNumber of Headache days (Mean)
Treatment Month 1Treatment Month 2Treatment Month 3
Nuedexta7.17.057.10
Placebo10.889.479.77

[back to top]

Headache Duration in Each Treatment Period Month

The average headache duration (time of onset to pain free) at treatment period months 1, 2, and 3 (28 day periods for each month) for the Nuedexta arm and the Placebo arm. Headache duration was measured in hours. (NCT02176018)
Timeframe: Treatment Month 1, Month 2, and End of Treatment Period Month 3 (Day 116)

,
InterventionHours (Mean)
Treatment Month 1Treatment Month 2Treatment Month 3
Nuedexta5.396.336.60
Placebo5.305.755.46

[back to top]

Headache Health Score

Headache Health Score at Visit 2 (Day 28), Visit 3 (Day 57), Visit 4 (Day 85), and Visit 5 (Day 116) for the Nuedexta arm and the placebo arm. The Headache Health Score is measured using a scale from 0 to 100, with higher scores indicating less headache impact on the subject's life. (NCT02176018)
Timeframe: Visit 2 (Day 28), Visit 3 (Day 57), Visit 4 (Day 85), and Visit 5 (Day 116)

,
Interventionunits on a scale (Mean)
Visit 2 (Day 28)Visit 3 (Day 57)Visit 4 (Day 85)Visit 5 (Day 116)
Nuedexta78.7187.6287.1487.57
Placebo81.0780.3082.2884.10

[back to top]

Headache Severity in Each Treatment Period Month

The average headache severity at treatment period months 1, 2, and 3 (28 day periods for each month) for the Nuedexta arm and the Placebo arm. Headache pain severity was measured on a scale from 1 = Mild, to 3 = Severe. Higher numbers indicating more severe headache pain. (NCT02176018)
Timeframe: Treatment Month 1, Month 2, and End of Treatment Period Month 3 (Day 116)

,
Interventionunits on a scale (Mean)
Treatment Month 1Treatment Month 2Treatment Month 3
Nuedexta1.691.811.74
Placebo2.041.961.86

[back to top]

Migraine Days in Each Treatment Period Month

The average number of migraine days at treatment period months 1, 2, and 3 (28 day periods for each month) for the Nuedexta arm and the Placebo arm. (NCT02176018)
Timeframe: Treatment Month 1, Month 2, and End of Treatment Period Month 3 (Day 116)

,
InterventionNumber of Migraine days (Mean)
Treatment Month 1Treatment Month 2Treatment Month 3
Nuedexta5.655.856.05
Placebo7.656.947.29

[back to top]

Acute Medication Use in Each Treatment Period Month

The average number of doses of acute medication taken at treatment period months 1, 2, and 3 (28 day periods for each month) for the Nuedexta arm and the Placebo arm (NCT02176018)
Timeframe: Treatment Month 1, Month 2, and End of Treatment Period Month 3 (Day 116)

,
InterventionNumber of medication doses (Mean)
Treatment Month 1Treatment Month 2Treatment Month 3
Nuedexta9.459.5011.55
Placebo13.0611.4110.94

[back to top]

Migraine Disability Assessment Scale (MIDAS)

"MIDAS scores at Visit 2 and Visit 5 (end of treatment period month 3). The MIDAS is a questionnaire consisting of five (5) how many days in the last 3 months... questions. Thus the range for the MIDAS is from, 0 to a maximum possible score 93 (31 days X 3 months). The MIDAS is scored according to the following:~0-5, MIDAS Grade I, Little or No Disability 6-10, MIDAS Grade II, Mild Disability 11-20, MIDAS Grade III, Moderate Disability 21+, MIDAS Grade IV, Severe Disability" (NCT02176018)
Timeframe: Visit 2 (Day 28) to Visit 5 (Day 116)

,
Interventionunits on a scale (Mean)
Visit 2 (Day28)Visit 5 (Day 116)
Nuedexta34.9522.50
Placebo32.8830.35

[back to top]

Percent Change in Peak Pain Intensity

Primary outcome was percent change from baseline in mean pain intensity at Cmax (transformed Gracely Scale; 0-35). Higher values on the Gracely scale represent greater pain intensity; the greater the percent change from baseline in mean pain intensity, the bigger the reduction in pain intensity. (NCT02218203)
Timeframe: 30 minutes post-infusion (Cmax)

Interventionpercentage change from baseline (Mean)
Dextromethorphan/ 0mg/kg Lidocaine Combination-2.5
Dextromethorphan/1mg/kg Lidocaine Combination-7.9
Dextromethorphan/2mg/kg Lidocaine Combination-11.2
Dextromethorphan/4mg/kg Lidocaine Combination-19.2

[back to top]

Adverse Drug Events: CYP2D6 Metabolizer Status

Occurrence of adverse drug events will be captured and stratified by CYP2D6 metabolizer status (Poor Metabolizer (PM), Extensive Metabolizer (EM), Intermediate Metabolizer (IM), and Ultra rapid Metabolizer). (NCT02293096)
Timeframe: 6 weeks

Interventionparticipants with events (Number)
CYP2D6 metabolizer status: EMOther Metabolizer Statuses
Metoprolol Succinate, Genotyping, Clinical Factors, and Phenotyping30

[back to top]

Adverse Drug Events: ADRB1 Genotype

Occurrence of adverse drug events will be captured and stratified by ADRB1 genotype (strong responder, good responder, non-responder). (NCT02293096)
Timeframe: 6 weeks

Interventionparticipants with events (Number)
ADRB1 genotype: Strong ResponderADRB1 genotype: Good ResponderADRB1 genotype: Non-responder
Metoprolol Succinate, Genotyping, Clinical Factors, and Phenotyping120

[back to top]

Heart Rate Decline

10 % decline from pre-initiation heart rate will considered a HR decline success. Number of of participants with at least 10% decline is reported. (NCT02293096)
Timeframe: 4-6 weeks

InterventionParticipants (Count of Participants)
Metoprolol Succinate, Genotyping, Clinical Factors, and Phenotyping67

[back to top]

Blood Pressure Decline

Participants with at least a 10% decrease in SBP (NCT02293096)
Timeframe: 4-6 weeks status post initiation

InterventionParticipants (Count of Participants)
Metoprolol Succinate, Genotyping, Clinical Factors, and Phenotyping85

[back to top]

Good European League Against Rheumatism (EULAR) Therapeutic Response Rate

(NCT02368093)
Timeframe: 6 months

Interventionparticipants (Number)
Dextromethorphan Hydrobromide6
Placebo4

[back to top]

Mean of Total Cough Counts: Between Dose 1 to Dose 2 on Day 1

Total cough count was collected by the cough recording device VitaloJAKTM in an ambulatory setting. The VitaloJAKTM device recorded continuous digital audio obtained through both a lapel microphone clipped to the participant's clothing at the neck or upper chest level, and a chest wall sensor attached to the participant's chest at the top of the sternum. Data was captured on a data card and the vitalograph analyst evaluated cough counts. (NCT02651116)
Timeframe: Between Dose 1 to Dose 2 on Day 1

Interventioncough counts (Mean)
Dextromethorphan Hydrobromide32.73
Placebo47.03

[back to top]

Mean of Total Cough Counts: Between Dose 2 on Day 1 to Dose 3 on Day 2

Total cough count was collected by the cough recording device VitaloJAKTM in an ambulatory setting. The VitaloJAKTM device recorded continuous digital audio obtained through both a lapel microphone clipped to the participant's clothing at the neck or upper chest level, and a chest wall sensor attached to the participant's chest at the top of the sternum. Data was captured on a data card and the vitalograph analyst evaluated cough counts. (NCT02651116)
Timeframe: Between Dose 2 on Day 1 to Dose 3 on Day 2 (second dose of Day 1 to first dose of Day 2)

Interventioncough counts (Mean)
Dextromethorphan Hydrobromide9.70
Placebo11.44

[back to top]

Mean of Total Cough Counts: Between Dose 3 to Dose 4 on Day 2

Total cough count was collected by the cough recording device VitaloJAKTM in an ambulatory setting. The VitaloJAKTM device recorded continuous digital audio obtained through both a lapel microphone clipped to the participant's clothing at the neck or upper chest level, and a chest wall sensor attached to the participant's chest at the top of the sternum. Data was captured on a data card and the vitalograph analyst evaluated cough counts. (NCT02651116)
Timeframe: Between Dose 3 to Dose 4 on Day 2 (between first and second dose of Day 2)

Interventioncough counts (Mean)
Dextromethorphan Hydrobromide19.32
Placebo33.62

[back to top]

Mean of Total Cough Time Accumulated Over a 24-Hour Period Post-First Dose on Day 1

Time (in seconds) accumulated over a 24-hour period when cough events occurred was collected by the cough recording device VitaloJAKTM in an ambulatory setting. The VitaloJAKTM device recorded continuous digital audio obtained through both a lapel microphone clipped to the participant's clothing at the neck or upper chest level, and a chest wall sensor attached to the participant's chest at the top of the sternum. Data was captured on a data card and the vitalograph analyst evaluated total cough time accumulated. (NCT02651116)
Timeframe: Over for 24 hours post-first dose on Day 1

Interventionseconds (Mean)
Dextromethorphan Hydrobromide350.5
Placebo502.7

[back to top]

Change From Baseline in Afternoon Cough Frequency Assessed at Afternoon on Day 2, 3, and 4

"Participants on specified time points were asked to respond to the following question: how much have you been coughing this afternoon on a 5-point scale: 0= not at all, 1= a tiny bit, 2= a little, 3= some and 4= a lot. Higher scores indicated higher frequency of cough in afternoon time." (NCT02651116)
Timeframe: Baseline (afternoon visit on Day 1 before first dose); Before the afternoon dose on Day 2, and 3; Anytime in afternoon of Day 4

,
Interventionunits on a scale (Mean)
BaselineChange at Day 2Change at Day 3Change at Day 4
Dextromethorphan Hydrobromide3.2-0.7-1.5-1.9
Placebo3.4-0.6-1.4-1.8

[back to top]

Change From Baseline in Afternoon Cough Severity Assessed at Afternoon on Day 2, 3, and 4

"Participants on specified time points were asked to respond to the following question: how bad is your cough this afternoon on a 5-point scale: 0= no cough, 1= a tiny bit bad, 2= a little bad, 3= bad and 4= very bad. Higher scores indicated more severe cough in afternoon time." (NCT02651116)
Timeframe: Baseline (afternoon visit on Day 1 before first dose); Before the afternoon dose on Day 2, and 3; Anytime in afternoon of Day 4

,
Interventionunits on a scale (Mean)
BaselineChange at Day 2Change at Day 3Change at Day 4
Dextromethorphan Hydrobromide2.8-0.7-1.4-1.7
Placebo3.1-0.6-1.4-1.6

[back to top]

Change From Baseline in Child Global Question Assessed at Afternoon on Day 2, 3, and 4

"Participants on specified time points were asked to respond to the following question: how bad is your cold today, on a 5-point scale; 0= no cold, 1= a tiny bit bad, 2= a little bad, 3= bad, and 4= very bad. Higher scores indicated worse cold." (NCT02651116)
Timeframe: Baseline (afternoon visit on Day 1 before first dose); Before the afternoon dose on Day 2, and 3; Anytime in afternoon of Day 4

,
Interventionunits on a scale (Mean)
BaselineChange at Day 2Change at Day 3Change at Day 4
Dextromethorphan Hydrobromide3.2-1.1-1.6-2.1
Placebo3.3-0.9-1.6-1.7

[back to top]

Change From Baseline in Impact of Cough on Sleep Assessed in Morning at Day 2, 3, and 4

"Participants on specified time points were asked to respond to the following question: last night in bed, how much did your cough keep you awake, on a 5-point scale: 0= not at all, 1= a tiny bit, 2= a little, 3= some and 4= a lot. Higher scores indicated worse impact of cough on sleep." (NCT02651116)
Timeframe: Baseline (morning screening visit on Day 1); Within 30 minutes of waking, before morning dose on Days 2, 3, and 4

,
Interventionunits on a scale (Mean)
BaselineChange at Day 2Change at Day 3Change at Day 4
Dextromethorphan Hydrobromide2.8-0.8-1.3-1.8
Placebo3.0-0.7-1.4-1.9

[back to top]

Change From Baseline in Morning Cough Frequency Assessed in Morning at Day 2, 3, and 4

"Participants on specified time points were asked to respond to the following question: from when you woke up this morning until now, how much have you been coughing, on a 5-point scale: 0= not at all, 1= a tiny bit, 2= a little, 3= some and 4= a lot. Higher scores indicated higher frequency of cough in morning time." (NCT02651116)
Timeframe: Baseline (morning screening visit on Day 1); Within 30 minutes of waking, before morning dose on Days 2, 3, and 4

,
Interventionunits on a scale (Mean)
BaselineChange at Day 2Change at Day 3Change at Day 4
Dextromethorphan Hydrobromide3.4-1.2-1.5-2.0
Placebo3.3-0.7-1.1-1.8

[back to top]

Change From Baseline in Morning Cough Severity Assessed in Morning at Day 2, 3, and 4

"Participants on specified time points were asked to respond to the following question: how bad is your cough this morning, on a 5-point scale: 0= no cough, 1= a tiny bit bad, 2= a little bad, 3= bad and 4= very bad. Higher scores indicated more severe cough in morning time." (NCT02651116)
Timeframe: Baseline (morning screening visit on Day 1); Within 30 minutes of waking, before morning dose on Days 2, 3, and 4

,
Interventionunits on a scale (Mean)
BaselineChange at Day 2Change at Day 3Change at Day 4
Dextromethorphan Hydrobromide3.1-1.1-1.4-1.9
Placebo3.1-0.6-1.3-1.8

[back to top]

Pediatric Global Assessment of Satisfaction With Study Medication: By Participant, and Caregiver

"Participants at the end of the study were asked to respond to the following question: How would you rate the study medication for taking away your cough? on a 7-point scale: 0= excellent, 1= very good, 2= good, 3= fair, 4= poor, 5= very poor, and 6= terrible. Higher scores indicated poorer satisfaction with study medication. Within 20 minutes after participants completed the assessment parents/legally acceptable representative were asked to respond to the question: How would you rate the study medication for taking away your child's cough? on a 7-point scale: 0= excellent, 1= very good, 2= good, 3= fair, 4= poor, 5= very poor, and 6= terrible. Higher scores indicated poorer satisfaction with study medication." (NCT02651116)
Timeframe: For participants: at the end of the study on Day 4; For parents/legally acceptable representatives: within 20 minutes after participant completed assessment at the end of the study on Day 4

,
Interventionunits on a scale (Mean)
By Participant:By Caregiver:
Dextromethorphan Hydrobromide1.71.8
Placebo1.61.9

[back to top]

Mean of Total Cough Counts: Over 24 Hours Post-First Dose on Day 1

Total cough count was collected by the cough recording device VitaloJAKTM in an ambulatory setting. The VitaloJAKTM device recorded continuous digital audio obtained through both a lapel microphone clipped to the participant's clothing at the neck or upper chest level, and a chest wall sensor attached to the participant's chest at the top of the sternum. Data was captured on a data card and the vitalograph analyst evaluated cough counts. (NCT02651116)
Timeframe: Over for 24 hours post-first dose on Day 1

Interventioncough counts (Mean)
Dextromethorphan Hydrobromide457.1
Placebo676.8

[back to top]

Mean of Total Cough Counts: Between Dose 1 to Dose 2 on Day 1, and Between Dose 3 to Dose 4 on Day 2

Total cough count was collected by the cough recording device VitaloJAKTM in an ambulatory setting. The VitaloJAKTM device recorded continuous digital audio obtained through both a lapel microphone clipped to the participant's clothing at the neck or upper chest level, and a chest wall sensor attached to the participant's chest at the top of the sternum. Data was captured on a data card and the vitalograph analyst evaluated cough counts. In this outcome measure, as planned combined data is reported for first dosing interval (Dose 1 to Dose 2) on Day 1 and first dosing interval (Dose 3 to Dose 4) on Day 2. (NCT02651116)
Timeframe: Duration between Dose 1 to Dose 2 on Day 1 (between first and second dose of Day 1) plus duration between Dose 3 to Dose 4 on Day 2 (between first and second dose of Day 2)

Interventioncough counts (Mean)
Dextromethorphan Hydrobromide26.13
Placebo40.39

[back to top]

Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of Caffeine

PK: AUC zero to infinity of caffeine after single dose of drug cocktail on Day 1 in Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48 hr Postdose

Interventionnanograms*hour per milliliter (ng*h/mL) (Geometric Least Squares Mean)
100 mg Caffeine32500
200 mg Abemaciclib + 100 mg Caffeine47100

[back to top]

Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of Dextromethorphan

PK: AUC (zero to infinity) of dextromethorphan after single dose of drug cocktail on Day 1 in Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: 1, 2, 4, 6, 8, 10, 24, 48, 72 hr Postdose

Interventionng*h/mL (Geometric Mean)
30 mg Dextromethorphan32.6
200 mg Abemaciclib + 30 mg Dextromethorphan32.1

[back to top]

Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of Midazolam

PK: AUC (zero to infinity) of midazolam after single dose of drug cocktail on Day 1 in Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24 hr Postdose

Interventionng*h/mL (Geometric Mean)
0.2 mg Midazolam7.34
200 mg Abemaciclib + 0.2 mg Midazolam6.03

[back to top]

Pharmacokinetics: Area Under the Concentration Versus Time Curve [AUC(0-infinity)] of S-Warfarin

AUC (zero to infinity) of S-warfarin after single dose of drug cocktail on Day 1 in Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48, 72, 96 hr Postdose

Interventionng*h/mL (Geometric Mean)
10 mg Warfarin21400
200 mg Abemaciclib + 10 mg Warfarin20600

[back to top]

Mean Change From Baseline at 24 Hours in Pulse Rate in Period 1

Mean change from baseline in pulse rate over 24 hours (h) postdose following single dose drug cocktail in Period 1. (NCT02688088)
Timeframe: Day 8: Baseline, 24 h postdose

InterventionBeats per minute (bpm) (Mean)
Drug Cocktail Period 1-1.3

[back to top]

Pharmacokinetics: Maximum Concentration (Cmax) of Caffeine

Maximum concentration of caffeine after single dose of drug cocktail on Day 1 in Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, and 48 hours (hr) Postdose

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
100 mg Caffeine2890
Abemaciclib + 100 mg Caffeine2950

[back to top]

Pharmacokinetics: Maximum Concentration (Cmax) of Dextromethorphan

Maximum concentration of dextromethorphan after single dose of drug cocktail on Day 1 of Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 1, 2, 4, 6, 8, 10, 24, 48, 72 hr postdose

Interventionng/mL (Geometric Mean)
30 mg Dextromethorphan3.18
200 Abemaciclib + 30 mg Dextromethorphan3.30

[back to top]

Pharmacokinetics: Maximum Concentration (Cmax) of Midazolam

Maximum concentration of midazolam after single dose of drug cocktail on Day 1 of Period 1 and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24 hr Postdose

Interventionng/mL (Geometric Mean)
0.2 mg Midazolam (Drug Cocktail)2.12
200 mg Abemaciclib + 0.2 mg Midazolam1.75

[back to top]

Mean Change From Baseline at 24 Hours in Pulse Rate in Period 2

Mean change from baseline in pulse rate over 24 hours (h) postdose following single dose drug cocktail in Period 2, Day 1. (NCT02688088)
Timeframe: Day 1: Baseline, 24 h postdose

Interventionbpm (Mean)
200 mg Abemaciclib-0.2

[back to top]

Pharmacokinetics: Maximum Concentration (Cmax) S-Warfarin

Maximum concentration of S-warfarin after single dose of drug cocktail on Day 1 in Period 1and in combination with Abemaciclib on Day 8 in Period 2. (NCT02688088)
Timeframe: Days 1 and 8: Predose, 0.5 1, 2, 3, 4, 6, 8, 12, 48, 72, 96 hr Postdose

Interventionng/mL (Geometric Mean)
10 mg Warfarin561
200 mg Abemaciclib + 10 mg Warfarin526

[back to top]

Mean Change From Baseline at 24 Hours in Systolic and Diastolic Blood Pressure in Period 1

Mean change from predose in systolic and diastolic blood pressure (BP) over 24 hours (h) postdose following single dose drug cocktail in Period 1. (NCT02688088)
Timeframe: Day 8: Baseline, 24 h postdose

Interventionmillimeter of mercury (mmHg) (Mean)
Systolic BPDiastolic BP
Drug Cocktail Period 1-2.5-0.7

[back to top]

Mean Change From Baseline at 24 Hours in Systolic and Diastolic Blood Pressure in Period 2

Mean change from baseline in systolic and diastolic blood pressure (BP) at 24 h postdose following 200 mg abemaciclib and drug cocktail. (NCT02688088)
Timeframe: Day 8: Baseline, 24 h postdose

InterventionmmHg (Mean)
Systolic BPDiastolic BP
200 mg Abemaciclib + Drug Cocktail-11.6-6.1

[back to top]

Mean Change From Baseline at 24 Hours in Systolic and Diastolic Blood Pressure in Period 2

Mean change from baseline in systolic and diastolic blood pressure (BP) over 24 hours (h) postdose following single dose of abemaciclib in Period 2, Day 1. (NCT02688088)
Timeframe: Day 1: Baseline, 24 h postdose

InterventionmmHg (Mean)
Systolic BPDiastolic BP
200 mg Abemaciclib-7.8-1.8

[back to top]

Mean Change From Baseline at 24 Hours in Pulse Rate in Period 2

Mean change from baseline in pulse rate at 24 h postdose following 200 mg abemaciclib and drug cocktail. (NCT02688088)
Timeframe: Day 8: Baseline, 24 h postdose

Interventionbpm (Mean)
200 mg Abemaciclib + Drug Cocktail4.1

[back to top]

Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) of Dextrorphan

AUClast is the area under the plasma concentration versus time curve from the time of dosing to the last measurable concentration. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
Treatment A2200
Treatment B2110

[back to top]

Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) of Dextromethorphan

AUClast is the area under the plasma concentration versus time curve from the time of dosing to the last measurable concentration. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
Treatment A7.06
Treatment B6.77

[back to top]

Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) of Digoxin

AUClast is the area under the plasma concentration versus time curve from the time of dosing to the last measurable concentration. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
Treatment A23.0
Treatment B26.7

[back to top]

Apparent Oral Clearance (CL/F) of Digoxin

CL/F is equal to dose/AUC0-infinity (dose divided by area under the plasma concentration versus time curve extrapolated to infinity [AUC0-infinity]). (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionLiter per hour (L/h) (Geometric Mean)
Treatment A15.8
Treatment B13.4

[back to top]

Area Under the Plasma Concentration Versus Time Curve From Time Zero to the End of the Dosing Interval at Steady-State (AUCtau) of Maribavir

AUCtau is the area under the plasma concentration versus time curve from the time zero to the end of the dosing interval at steady-state. (NCT02775240)
Timeframe: Pre-dose, 0.25,0.5,1,1.5,2,3,4,5,6,8,12 hours post-dose on Day 13

InterventionMicrogram*hour per milliliter (mcg*h/mL) (Geometric Mean)
Treatment B91.5

[back to top]

Concentration at the End of Dosing Interval (Ctau) of Maribavir

Ctau is the concentration of maribavir at the end of the dosing interval. (NCT02775240)
Timeframe: Pre-dose, 0.25,0.5,1,1.5,2,3,4,5,6,8,12 hours post-dose on Day 13

InterventionMicrogram per milliliter (mcg/mL) (Geometric Mean)
Treatment B2.13

[back to top]

Apparent Oral Clearance (CL/F) of Maribavir

CL/F is equal to dose/AUCtau (dose divided by area under the curve from time 0 to the end of the dosing interval at steady state [AUCtau]) (NCT02775240)
Timeframe: Pre-dose, 0.25,0.5,1,1.5,2,3,4,5,6,8,12 hours post-dose on Day 13

InterventionLiter per hour (L/h) (Geometric Mean)
Treatment B2.19

[back to top]

Volume of Distribution Divided by the Fraction of Dose Absorbed (Vz/F) of Dextromethorphan

Vz/F is the volume of distribution associated with the terminal slope following extravascular administration divided by the fraction of dose absorbed. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

,
InterventionLiter (L) (Number)
Participant 1Participant 2Participant 3Participant 4Participant 5Participant 6
Treatment A2360.5414008.68NA2519.116142.8510836.77
Treatment B3904.44NA11078.423931.635796.8716835.78

[back to top]

Terminal Half-life (t1/2) of Dextromethorphan

Terminal half-life (t1/2) is the time in hours required for the concentration of the drug to reach half of its original value. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

,
InterventionHour (h) (Number)
Participant 1Participant 2Participant 3Participant 4Participant 5Participant 6
Treatment A15.869.09NA9.826.866.71
Treatment B16.17NA6.438.725.587.03

[back to top]

Parent/Metabolite Ratio of Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) for Dextromethorphan Over AUC0-infinity for Dextrorphan (AUC0-infinity Parent/Metabolite Ratio)

AUC0-infinity parent/metabolite ratio is the ratio of AUC0-infinity for dextromethorphan over AUC0-infinity for dextrorphan. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

,
InterventionRatio of AUC0-infinity (Number)
Participant 1Participant 2Participant 3Participant 4Participant 5Participant 6
Treatment A0.1770.009NA0.0960.0280.013
Treatment B0.127NA0.0140.0560.0250.008

[back to top] [back to top]

First-order Rate Constant (Lambda z) Associated With the Terminal (Log-linear) Portion of the Curve of Dextromethorphan

Lambda z is the first-order rate constant associated with the terminal (log-linear) portion of the plasma concentration versus time curve, determined as the negative slope of the terminal log-linear phase of the curve. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

,
InterventionPer hour (/h) (Number)
Participant 1Participant 2Participant 3Participant 4Participant 5Participant 6
Treatment A0.040.08NA0.070.100.10
Treatment B0.04NA0.110.080.120.10

[back to top]

Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) of Dextromethorphan

AUC0-infinity is the area under the plasma concentration versus time curve extrapolated to infinity, calculated using the observed value of the last non-zero concentration. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

,
InterventionNanogram*hour per milliliter (ng*h/mL) (Number)
Participant 1Participant 2Participant 3Participant 4Participant 5Participant 6
Treatment A290.7428.10NA168.6948.3326.79
Treatment B179.19NA25.1495.9541.6518.08

[back to top]

Apparent Oral Clearance (CL/F) of Dextromethorphan

CL/F is equal to dose/AUC0-infinity (dose divided by area under the plasma concentration versus time curve extrapolated to infinity [AUC0-infinity]) (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

,
InterventionLiter per hour (L/h) (Number)
Participant 1Participant 2Participant 3Participant 4Participant 5Participant 6
Treatment A103.191067.64NA177.84620.711119.90
Treatment B167.42NA1193.51312.65720.231659.07

[back to top]

Volume of Distribution Divided by the Fraction of Dose Absorbed (Vz/F) of Digoxin

Vz/F is the volume of distribution associated with the terminal slope following extravascular administration divided by the fraction of dose absorbed. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionLiter (L) (Geometric Mean)
Treatment A946
Treatment B809

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax) of Maribavir

Tmax is the time to reach the maximum observed drug concentration in plasma during a dosing interval. (NCT02775240)
Timeframe: Pre-dose, 0.25,0.5,1,1.5,2,3,4,5,6,8,12 hours post-dose on Day 13

InterventionHour (h) (Median)
Treatment B2.00

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax) of Digoxin

Tmax is the time to reach the maximum observed drug concentration in plasma during a dosing interval. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionHour (h) (Median)
Treatment A1.00
Treatment B1.00

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax) of Dextrorphan

Tmax is the time to reach the maximum observed drug concentration in plasma during a dosing interval. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionHour (h) (Median)
Treatment A2.00
Treatment B2.00

[back to top]

Time to Reach Maximum Plasma Concentration (Tmax) of Dextromethorphan

Tmax is the time to reach the maximum observed drug concentration in plasma during a dosing interval. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionHour (h) (Median)
Treatment A3.00
Treatment B3.00

[back to top]

Terminal Half-life (t1/2) of Maribavir

Terminal half-life (t1/2) is the time in hours required for the concentration of the drug to reach half of its original value. (NCT02775240)
Timeframe: Pre-dose, 0.25,0.5,1,1.5,2,3,4,5,6,8,12 hours post-dose on Day 13

InterventionHour (h) (Geometric Mean)
Treatment B4.04

[back to top]

Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) of Dextrorphan

AUC0-infinity is the area under the plasma concentration versus time curve extrapolated to infinity, calculated using the observed value of the last non-zero concentration. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
Treatment A2270
Treatment B2150

[back to top]

Area Under the Plasma Concentration Versus Time Curve Extrapolated to Infinity (AUC0-infinity) of Digoxin

AUC0-infinity is the area under the plasma concentration versus time curve extrapolated to infinity, calculated using the observed value of the last non-zero concentration. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
Treatment A31.6
Treatment B37.3

[back to top]

Terminal Half-life (t1/2) of Digoxin

Terminal half-life (t1/2) is the time in hours required for the concentration of the drug to reach half of its original value. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionHour (h) (Geometric Mean)
Treatment A41.5
Treatment B41.8

[back to top]

Number of Participants With Clinically Significant Changes Reported as TEAE in Physical Examination, Vital Signs, 12-lead ECGs, Hematology, Blood Chemistry and Urinalysis

Clinical significance of the changes observed in the safety parameters to be reported as TEAE was interpreted by the investigator. (NCT02775240)
Timeframe: Baseline up to Day 16

,
InterventionParticipants (Count of Participants)
Physical examinationVital signs12-lead ECGsHematologyBlood ChemistryUrinalysis
Treatment A000000
Treatment B000000

[back to top]

First-order Rate Constant (Lambda z) Associated With the Terminal (Log-linear) Portion of the Curve of Dextrorphan

Lambda z is the first-order rate constant associated with the terminal (log-linear) portion of the plasma concentration versus time curve, determined as the negative slope of the terminal log-linear phase of the curve. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionPer hour (/h) (Geometric Mean)
Treatment A0.16
Treatment B0.17

[back to top]

First-order Rate Constant (Lambda z) Associated With the Terminal (Log-linear) Portion of the Curve of Digoxin

Lambda z is the first-order rate constant associated with the terminal (log-linear) portion of the plasma concentration versus time curve, determined as the negative slope of the terminal log-linear phase of the curve. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionPer hour (/h) (Geometric Mean)
Treatment A0.02
Treatment B0.02

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Dextromethorphan

Cmax is the maximum observed plasma concentration of dextromethorphan. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram per milliliter (ng/mL) (Geometric Mean)
Treatment A1.14
Treatment B1.14

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Dextrorphan

Cmax is the maximum observed plasma concentration of dextrorphan, the metabolite of dextromethorphan. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram per milliliter (ng/mL) (Geometric Mean)
Treatment A433
Treatment B401

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Digoxin

Cmax is the maximum observed plasma concentration of digoxin. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionNanogram per milliliter (ng/mL) (Geometric Mean)
Treatment A1.94
Treatment B2.35

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Maribavir

Cmax is the maximum observed plasma concentration of maribavir. (NCT02775240)
Timeframe: Pre-dose, 0.25,0.5,1,1.5,2,3,4,5,6,8,12 hours post-dose on Day 13

InterventionMicrogram per milliliter (mcg/mL) (Geometric Mean)
Treatment B17.6

[back to top]

Parent/Metabolite Ratio of Area Under the Plasma Concentration Versus Time Curve From the Time of Dosing to the Last Measurable Concentration (AUClast) for Dextromethorphan Over AUClast for Dextrorphan (AUClast Parent/Metabolite Ratio)

AUClast parent/metabolite ratio is the ratio of AUClast for dextromethorphan over AUClast for dextrorphan. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionRatio of AUClast (Geometric Mean)
Treatment A0.003
Treatment B0.003

[back to top]

Pre-dose Concentration (C0) of Maribavir

C0 is the lowest concentration reached by a drug before the next dose is administered. (NCT02775240)
Timeframe: Pre-dose on Day 13

InterventionMicrogram per milliliter (mcg/mL) (Geometric Mean)
Treatment B2.64

[back to top]

Terminal Half-life (t1/2) of Dextrorphan

Terminal half-life (t1/2) is the time in hours required for the concentration of the drug to reach half of its original value. (NCT02775240)
Timeframe: Pre-dose,0.25,0.5,1,1.5,2,3,4,5,6,8,12,24,48,72 hours post-dose on Day 1 for Treatment A and Day 13 for Treatment B

InterventionHour (h) (Geometric Mean)
Treatment A4.42
Treatment B4.16

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Dextromethorphan

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve from Zero to Infinity (AUC[0-∞]) of CYP450 Substrate (Dextromethorphan) (NCT02993471)
Timeframe: Predose, 1, 2, 4, 6, 8, 10, 24, 48, and 72 hours postdose

Interventionng*h/mL (Geometric Mean)
30 mg Dextromethorphan11.7
160 mg Ixekizumab + 30 mg Dextromethorphan12.6
80 mg Ixekizumab Q2W + 30 mg Dextromethorphan8.53

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Omeprazole and Its Metabolite 5-Hydroxyomeprazole

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate (Omeprazole and its metabolite 5-Hydroxyomeprazole) (NCT02993471)
Timeframe: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, and 48 hours postdose

,,
Interventionng/mL (Geometric Mean)
Omeprazole5-Hydroxyomeprazole
160 mg Ixekizumab + 20 mg Omeprazole340143
20 mg Omeprazole333148
80 mg Ixekizumab Q2W + 20 mg Omeprazole368137

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Omeprazole and Its Metabolite 5-Hydroxyomeprazole

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve from Zero to Infinity (AUC[0-∞]) of CYP450 Substrate (Omeprazole and its metabolite 5-Hydroxyomeprazole) (NCT02993471)
Timeframe: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, and 48 hours postdose

,,
Interventionng*h/mL (Geometric Mean)
Omeprazole5-Hydroxyomeprazole
160 mg Ixekizumab + 20 mg Omeprazole829475
20 mg Omeprazole1060519
80 mg Ixekizumab Q2W + 20 mg Omeprazole913455

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of Cytochrome P450 (CYP450) Substrate-Midazolam

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of Cytochrome P450 (CYP450) Substrate (Midazolam) (NCT02993471)
Timeframe: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, and 24 hours postdose

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
1 mg Midazolam4.56
160 mg Ixekizumab + 1 mg Midazolam4.92
80 mg Ixekizumab Q2W (Once Every Two Weeks) + 1 mg Midazolam4.83

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Warfarin

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate (Warfarin) (NCT02993471)
Timeframe: Predose, 1, 2, 4, 6, 8, 10, 24, 48, 72, and 96 hours postdose

Interventionng/mL (Geometric Mean)
10 mg Warfarin510
160 mg Ixekizumab + 10 mg Warfarin525
80 mg Ixekizumab Q2W + 10 mg Warfarin510

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Dextromethorphan

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate (Dextromethorphan) (NCT02993471)
Timeframe: Predose, 1, 2, 4, 6, 8, 10, 24, 48, and 72 hours postdose

Interventionng/mL (Geometric Mean)
30 mg Dextromethorphan0.691
160 mg Ixekizumab + 30 mg Dextromethorphan0.878
80 mg Ixekizumab Q2W + 30 mg Dextromethorphan0.658

[back to top]

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate-Caffeine

Pharmacokinetics (PK): Maximum Observed Drug Concentration (Cmax) of CYP450 Substrate (Caffeine) (NCT02993471)
Timeframe: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, and 48 hours postdose

Interventionng/mL (Geometric Mean)
100 mg Caffeine2230
160 mg Ixekizumab + 100 mg Caffeine2220
80 mg Ixekizumab Q2W + 100 mg Caffeine2240

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Warfarin

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve from Zero to Infinity (AUC[0-∞]) of CYP450 Substrate (Warfarin) (NCT02993471)
Timeframe: Predose, 1, 2, 4, 6, 8, 10, 24, 48, 72, and 96 hours postdose

Interventionng*h/mL (Geometric Mean)
10 mg Warfarin17600
160 mg Ixekizumab + 10 mg Warfarin17700
80 mg Ixekizumab Q2W + 10 mg Warfarin16200

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to Infinity (AUC[0-∞]) of CYP450 Substrate-Midazolam

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve from Zero to Infinity (AUC[0-∞]) of CYP450 Substrate (Midazolam) (NCT02993471)
Timeframe: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, and 24 hours postdose

Interventionnanogram*hour per milliliter (ng*h/mL) (Geometric Mean)
1 mg Midazolam16.6
160 mg Ixekizumab + 1 mg Midazolam15.9
80 mg Ixekizumab Q2W + 1 mg Midazolam15.4

[back to top]

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve From Zero to 48 Hours (AUC[0-48h]) of CYP450 Substrate-Caffeine

Pharmacokinetics (PK): Area Under the Concentration Versus Time Curve from Zero to 48 hours (AUC[0-48h]) of CYP450 Substrate (Caffeine) (NCT02993471)
Timeframe: Predose, 0.5, 1, 2, 3, 4, 6, 8, 12, 24, and 48 hours postdose

Interventionng*h/mL (Geometric Mean)
100 mg Caffeine25000
160 mg Ixekizumab + 100 mg Caffeine22400
80 mg Ixekizumab Q2W + 100 mg Caffeine22400

[back to top]

Small Intestine Transit Time

Small intestinal transit time was calculated by determining the arrival time of the radiolabeled investigational drug formulation at the cecum or colon region from scintigraphic imaging and subtracting the gastric emptying value. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Median)
Treatment Group A184.0
Treatment Group B139.5

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 90 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 90 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 90 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A8.84
Treatment Group B2.45

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 75 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 75 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 75 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A14.73
Treatment Group B3.28

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 60 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 60 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 60 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A20.07
Treatment Group B4.26

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 120 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 120 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 120 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A4.34
Treatment Group B0.99

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 45 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 45 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 45 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A24.79
Treatment Group B7.21

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 30 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 30 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 30 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A33.24
Treatment Group B20.87

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 240 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 240 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 240 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A0.00
Treatment Group B0.00

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 180 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 180 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 180 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A0.64
Treatment Group B0.00

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 15 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 15 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 15 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A67.00
Treatment Group B49.77

[back to top]

Percentage of Radiolabeled Drug Remaining in the Stomach After 105 Minutes of Administration

Percentage of radiolabeled drug remaining in the stomach was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 105 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 105 minutes post dose on Day 1

Interventionpercentage of radiolabeled drug (Mean)
Treatment Group A6.06
Treatment Group B1.72

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 180 Minutes

Area under the gastric emptying curve from time 0 to 180 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 180 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 180 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A56.69
Treatment Group B32.61

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 15 Minutes

Area under the gastric emptying curve from time 0 to 15 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 15 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 15 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A19.38
Treatment Group B16.96

[back to top]

Mean Time to Onset of Gastric Emptying

Mean time to onset of gastric emptying in participants who did not vomit shortly (within 60 minutes) after study drug administration was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 microcurie [mcCi] isotope-technetium-99m-diethylene-triamine-pentaacetate [DTPA]). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. Regions of interest (ROI) included the stomach, proximal small intestine, distal small intestine and colon. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Mean)
Treatment Group A1.107
Treatment Group B8.534

[back to top]

Mean Time to Complete Gastric Emptying

Mean time to complete gastric emptying in participants who did not vomit shortly (within 60 minutes) after study drug administration was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m-DTPA). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. ROI included the stomach, proximal small intestine, distal small intestine and colon. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Mean)
Treatment Group A121.2
Treatment Group B65.3

[back to top]

Mean Time for Gastric Emptying by Measuring 90 Percent Values

Mean time to gastric emptying by 90 percent (GE90%) was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. ROI included the stomach, proximal small intestine, distal small intestine and colon. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Mean)
Treatment Group A84.929
Treatment Group B36.214

[back to top]

Mean Time for Gastric Emptying by Measuring 50 Percent Values

Mean time to gastric emptying by 50 percent (GE50%) was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. ROI included the stomach, proximal small intestine, distal small intestine and colon. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Mean)
Treatment Group A22.643
Treatment Group B16.134

[back to top]

Mean Time for Gastric Emptying by Measuring 25 Percent Values

Mean time to gastric emptying by 25 percent (GE25%) was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. ROI included the stomach, proximal small intestine, distal small intestine and colon. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Mean)
Treatment Group A12.614
Treatment Group B12.171

[back to top]

Gastric Emptying Half-Life

Gastric emptying half-life was defined as the time required by the stomach to empty 50% of the ingested meal and was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionminutes (Median)
Treatment Group A25.00
Treatment Group B5.00

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 90 Minutes

Area under the gastric emptying curve from time 0 to 90 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 90 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 90 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A51.32
Treatment Group B31.29

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 75 Minutes

Area under the gastric emptying curve from time 0 to 75 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 75 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 75 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A48.71
Treatment Group B30.61

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 60 Minutes

Area under the gastric emptying curve from time 0 to 60 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 60 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 60 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A43.69
Treatment Group B29.60

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 45 Minutes

Area under the gastric emptying curve from time 0 to 45 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 45 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 45 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A38.36
Treatment Group B28.19

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 120 Minutes

Area under the gastric emptying curve from time 0 to 120 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 120 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 120 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A54.49
Treatment Group B32.15

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 105 Minutes

Area under the gastric emptying curve from time 0 to 105 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 105 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 105 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A53.16
Treatment Group B31.81

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 240 Minutes

Area under the gastric emptying curve from time 0 to 240 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 240 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 240 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A56.80
Treatment Group B32.61

[back to top]

Total Area Under the Gastric Emptying Curve

Total area under the gastric emptying curve was evaluated by scintigraphic imaging, performed after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA). Data images were analyzed in a time-lapse format and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: Predose until 10 hours post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A56.80
Treatment Group B32.61

[back to top]

Area Under the Gastric Emptying Curve From Time 0 to 30 Minutes

Area under the gastric emptying curve from time 0 to 30 was evaluated by scintigraphic imaging, performed immediately after ingestion of the investigational drug formulation (radiolabeled with not more than 108 mcCi isotope-technetium-99m DTPA) and after 30 minutes of drug ingestion. Data images were analyzed and corrected for radioactive decay and background radiation. (NCT03415243)
Timeframe: 30 minutes post dose on Day 1

Interventionpercentage dose*hour (Mean)
Treatment Group A31.16
Treatment Group B25.11

[back to top]

Number of Participants With Clinically Significant Change in Laboratory Test Values

Haematological, biochemistry, urinalysis and virological parameters were analyzed. Clinical significance was judged by the investigator based upon the out of range values of standard range set for each parameter. (NCT03415243)
Timeframe: From baseline up to Day 1

InterventionParticipants (Count of Participants)
Treatment Group A0
Treatment Group B0

[back to top]

Percentage of Participants Who Experienced a More Than 50% Reduction in Expired Carbon Monoxide (CO) Levels

A biochemical marker of smoking intensity. (NCT03471767)
Timeframe: Baseline (V1), 3-Week Follow-Up Visit (V4)

Interventionpercentage of participants (Number)
AXS-0552.0
Bupropion SR30.4

[back to top]

Urinary Levels of Dextromethorphan

Measured via Urinary Dextromethorphan testing. (NCT03471767)
Timeframe: 3-Week Follow-Up Visit (V4)

Interventionng/mL (Mean)
AXS-051902.1

[back to top]

Change in Smoking Behavior

7-day point prevalence smoking abstinence. Measured by composite self-report diaries and biochemically confirmed via expired CO and salivary cotinine. (NCT03471767)
Timeframe: 3-Week Follow-Up Visit (V4), 4-Week Follow-Up Visit (V5)

InterventionParticipants (Count of Participants)
AXS-0516
Bupropion SR12

[back to top]

Change in Smoking Intensity

Smoking intensity refers to the number of cigarettes smoked per day. (NCT03471767)
Timeframe: Baseline (V1), 3-Week Follow-Up Visit (V4)

Interventionchange in cigarettes per day (Mean)
AXS-05-8.49
Bupropion SR-6.79

[back to top]

Medication Adherence

Medication adherence is measured by composite self-reported diaries. (NCT03471767)
Timeframe: Baseline (V1), 3-Week Follow-Up Visit (V4)

Interventionpercentage of adherence (Mean)
AXS-0597.08
Bupropion SR96.60

[back to top]

Medication Tolerance by Self-Reported Side Effects

Number of participants who scored 3 or higher on a 7-point Likert scale ranking severity of side effects (1-2 mild; 3-5 moderate; 6-7 severe). (NCT03471767)
Timeframe: Baseline (V1), 3-Week Follow-Up Visit (V4)

InterventionParticipants (Count of Participants)
AXS-057
Bupropion SR5

[back to top]

Medication Tolerance by Serious Adverse Events

Measured by FDA reporting guidelines on adverse event or serious adverse event designation. (NCT03471767)
Timeframe: Baseline (V1), 4-Week Follow-Up Visit (V5)

InterventionParticipants (Count of Participants)
AXS-050
Bupropion SR0

[back to top]

Mean Pain Scores Via Numeric Rating Scale (NRS-11)

"Marginal mean pain scores via Numeric Rating Scale (NRS-11) over 24 hours. The scale is from 0 to 10, where 0 represents no pain and 10 represents the worst pain possible." (NCT03480009)
Timeframe: Marginal mean pain scores over 24 hours

Interventionscore on a scale (NRS-11) (Mean)
Dextromethorphan, Opted for Narcotic Prescription2.93
Placebo, Opted for Narcotic Prescription2.96
Dextromethorphan, Declined Narcotic Prescription2.70
Placebo, Declined Narcotic Prescription2.41

[back to top]

Analgesic Usage During Medication Abortion

Analgesic usage by study arm for women who received dextromethorphan vs. placebo as adjunct to routine pain management during medication abortion; missing data are for participants who did not take the specified pain medication. (NCT03480009)
Timeframe: Over 24 hours

,,
Interventionmg (Median)
ibuprofen9acetaminophenoxycodone
Dextromethorphan, Opted for Narcotic Prescription800100010
Placebo, Declined Narcotic Prescription100097515
Placebo, Opted for Narcotic Prescription600130015

[back to top]

Analgesic Usage During Medication Abortion

Analgesic usage by study arm for women who received dextromethorphan vs. placebo as adjunct to routine pain management during medication abortion; missing data are for participants who did not take the specified pain medication. (NCT03480009)
Timeframe: Over 24 hours

Interventionmg (Median)
ibuprofen9acetaminophen
Dextromethorphan, Declined Narcotic Prescription800800

[back to top]

Worst Pain Measurement Via Numeric Rating Scale (NRS-11)

"Self-reported pain measurement via text-messaging system during first 24 hours after misoprostol administration. The scale is from 0 to 10, where 0 represents no pain and 10 represents the worst pain possible." (NCT03480009)
Timeframe: Over 24 hours starting from misoprostol administration

Interventionscore on a scale (Median)
Dextromethorphan, Opted for Narcotic Prescription8.0
Placebo, Opted for Narcotic Prescription7.0
Dextromethorphan, Declined Narcotic Prescription7.5
Placebo, Declined Narcotic Prescription6.5

[back to top]

Daily Self-reported Physical Activity

Self-reported daily activity, rated from 0 - 100 where 0 is less daily activity and 100 is the most amount of daily activity. Participants' daily scores from the last 4 weeks of each condition (placebo and dextromethorphan) were summed and then averaged across the 4-week time period. (NCT03538054)
Timeframe: Daily over 4 weeks

Interventionscore/day (Mean)
Dextromethorphan46.92
Placebo50.10

[back to top]

Daily Self-reported Pain Severity

Daily self-reported widespread pain severity, rated on a 0 - 100 scale (0 = no pain and 100 = worst pain possible). Participants' daily scores from the last 4 weeks of each condition (placebo and dextromethorphan) were summed and then averaged across the 4-week time period. (NCT03538054)
Timeframe: Daily over 4 weeks

Interventionscore/day (Mean)
Dextromethorphan41.09
Placebo45.55

[back to top]

Time in PACU

Total time in PACU before patient met discharge criteria. (NCT03714919)
Timeframe: 1-2 hr post-op

Interventionminutes (Median)
Non-opiod Pain Relief56

[back to top]

Number of Participants With Sedation, Nausea/Vomiting, or Hallucinations

Presence of sedation, nausea/vomiting, or hallucinations post-operatively. (NCT03714919)
Timeframe: 2 hours post-op

Interventionparticipants (Number)
SedationNausea/VomitingHallucinations
Non-opiod Pain Relief710

[back to top]

Extubation Time

Amount of time in the PACU before patient is ready to be extubated. (NCT03714919)
Timeframe: 1 hr post-op

Interventionminutes (Median)
Non-opiod Pain Relief23

[back to top]

End of Surgery to Hospital Discharge

Length of time before patient is ready to be discharged home. (NCT03714919)
Timeframe: 2-3 hours post-op

Interventionminutes (Median)
Non-opiod Pain Relief122

[back to top]

Average Pain Score

Visual Analogue Scale (VAS) pain scores (0 being no pain and 10 being worst pain) in post-anesthesia care unit (PACU). (NCT03714919)
Timeframe: 1 hr post-op

Interventionpain score (Mean)
Non-opiod Pain Relief0

[back to top]

Change in Speech Intelligibility

The Sentence Intelligibility Test (SIT) will be performed to assess the change in speaking intelligibility over the 30 day period. The primary outcome of the SIT will be the percentage of sentence intelligibility (%) during oral reading. (NCT03883581)
Timeframe: Baseline; Day 30

InterventionPercent Intelligibility (Mean)
Pre NuedextaPost Nuedexta
ALS Individuals With Bulbar Dysfunction71.5273.06

[back to top]

Change in Patient-reported Outcome: Center for Neurologic Study-Bulbar Function Scale (CNS-BFS)

The CNS-BFS is a validated patient-reported scale that assess self-reported impairments in the domains of speech, salivation and swallowing. Each domain contains 7 questions with ratings ranging from 1-5 with 5 considered the worst. For the speech domain, individuals who are unable to speak are assigned a value of 6 for each item (speech domain ranges from 1-6). Total scores ranging from 21 (no impairment) - 112 (severe impairment in all domains). (NCT03883581)
Timeframe: Baseline; Day 30

InterventionScore (Mean)
Pre NuedextaPost Nuedexta
ALS Individuals With Bulbar Dysfunction56.8754.13

[back to top]

Change in Dynamic Imaging Grade of Swallowing Toxicity

The validated Dynamic Imaging Grade of Swallowing Toxicity (DIGEST) will be performed on all collected videofluoroscopic swallowing studies to assess global swallowing function. The DIGEST total score is determined using the composite of individual airway safety and bolus efficiency subscores (range: 0-4). The DIGEST total is rated on a 5-point ordinal score ranging from 0 (no dysphagia) to 4 (life-threatening dysphagia). (NCT03883581)
Timeframe: Baseline; Day 30

InterventionParticipants (Count of Participants)
Pre Nuedexta DIGEST 0 (Normal)Post Nuedexta DIGEST 0 (Normal)
ALS Individuals With Bulbar Dysfunction37

[back to top]

Change in ALSFRS-R Bulbar Subscale Score

The ALS Functional Rating Scale-Revised Bulbar subscore is an outcome comprised of questions 1-3 on the validated ALSFRS-R scale. These items rate speech, swallowing and salivation functions on a scale from 0-total loss of function to 4- no symptoms for a total score of 0 to 12. (NCT03883581)
Timeframe: Baseline; Day 30

Interventionscore on a scale (Mean)
Pre NuedextaPost Nuedextaa
ALS Individuals With Bulbar Dysfunction7.478.39

[back to top]

Bamboo Passage Reading Duration (in Seconds)

The Bamboo Passage is a 60-word reading passage that is commonly used to measure speech duration. (NCT03883581)
Timeframe: Baseline; Day 30

InterventionSeconds (Mean)
Pre NuedextaPost Nuedexta
ALS Individuals With Bulbar Dysfunction66.1965.33

[back to top]

Incidence of Treatment-emergent AEs (TEAEs) Following Dosing With AXS-05

Types and rates of adverse events (NCT04039022)
Timeframe: Up to 12 months

InterventionParticipants (Count of Participants)
AXS-05505

[back to top]

Midazolam and Dextromethorphan Half-life

Time to reach one-half of the concentration of midazolam and dextromethorphan (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72, 96,120, and 144 hours

,
Interventionh (Geometric Mean)
midazolamdextromethorphan
Drug Cocktail3.856.87
Kratom + Drug Cocktail4.126.84

[back to top]

Mitragynine Area Under the Concentration vs. Time Curve (AUC)

Area under the concentration vs. time curve (AUC) of mitragynine. (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72, 96, and 120 hours

InterventionnM x h (Median)
Kratom Alone388

[back to top]

Mitragynine Cmax

Maximum plasma concentration of mitragynine. (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72, 96, and 120 hours

InterventionnM (Median)
Kratom Alone81.9

[back to top]

Mitragynine Half Life

Time to reach one-half of the concentration of mitragynine. (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72, 96, and 120 hours

Interventionh (Median)
Kratom Alone45.3

[back to top]

Midazolam and Dextromethorphan Cmax

Maximum concentration (Cmax) of midazolam and dextromethorphan (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72, 96,120, and 144 hours

,
InterventionnM (Geometric Mean)
midazolamdextromethorphan
Drug Cocktail21.14.57
Kratom + Drug Cocktail31.64.30

[back to top]

Dextromethorphan Area Under the Concentration vs. Time Curve (AUC)

Area under the plasma concentration time curve (AUC) of dextromethorphan (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72, 96, 120, and 144 hours

InterventionnM*h (Geometric Mean)
Drug Cocktail46.5
Kratom + Drug Cocktail46.2

[back to top]

Midazolam Area Under the Concentration vs. Time Curve (AUC)

Area under the plasma concentration time curve (AUC) of midazolam (NCT04392011)
Timeframe: 0, 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8,12, 24, 48, 72 , 96, 120, and 144 hours

InterventionnM * hr (Geometric Mean)
Drug Cocktail58.3
Kratom + Drug Cocktail80.8

[back to top]