Page last updated: 2024-12-11

levorphanol

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

Description

Levorphanol: A narcotic analgesic that may be habit-forming. It is nearly as effective orally as by injection. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

Cross-References

ID SourceID
PubMed CID5359272
CHEMBL ID592
CHEBI ID6444
SCHEMBL ID24335
MeSH IDM0012437

Synonyms (108)

Synonym
morphinan-3-ol, 17-methyl-
(-)-n-methylmorphinan-3-ol
17-methylmorphinan-3-ol
einecs 206-048-0
racemorphanum [inn-latin]
2h-10, 4a-iminoethanophenanthren-6-ol, 1,3,4,9,10,10a-hexahydro-11-methyl-, dl-
1,3,4,9,10,10a-hexahydro-11-methyl-2h-10,4a-iminoethanophenanthren-6-ol, l-
(+-)-17-methylmorphinan-3-ol
levorfanolo [dcit]
levorphan
ro 1-6794
morphinan-3-ol, 17-methyl-, (+-)-
dea no. 9733
aromarone
racemic dromoran
nu 2206
methorfinan [czech]
orphan
morphinan, 3-hydroxy-n-methyl-, (+-)-
(-)-3-hydroxy-n-methylmorphinan
morphinan, 3-hydroxy-n-methyl, (+)-
morphinan-3-ol, 17-methyl-, (-)-
methorphinan
levorfanol [inn-spanish]
antalgin
racemorphan [inn:ban]
2h-10,4a-(iminoethano)phenanthren-6-ol, 1,3,4,9,10,10a-hexahydro-11-methyl-, (+)-
levorphanolum [inn-latin]
racemorfano [inn-spanish]
dl-3-hydroxy-n-methylmorphinan
ro 1-5431
dea no. 9220
racemorphane [inn-french]
einecs 201-002-6
2h-10,4a-iminoethanophenanthren-6-ol, 1,3,4,9,10,10a-hexahydro-11-methyl-
morphinan, 3-hydroxy-17-methyl-
levodroman
n-methyl-3-hydroxymorphinan
cetarin
2h-10,4a-iminoethanophenanthren-6-ol, 1,3,4,9,10,10a-hexahydro-11-methyl-, l-
hsdb 3349
(+-)-3-hydroxy-n-methylmorphinan
dromoran
77-07-6
C08014
levorphanol
DB00854
levorphanol (inn)
D08123
chembl592 ,
bdbm50017233
racemorphanum
297-90-5
(+-)-n-methylmorphinan-3-ol
methorfinan
racemorfano
unii-v7r79hn3xd
v7r79hn3xd ,
racemorphane
ids-nl-007
chebi:6444 ,
bdbm82427
levorphanol-tartarate
cas_5985-38-6
bdbm50369518
levorphanal
levorphanol dl-form
unii-27618j1n2x
levorphanolum
levorphanol [inn:ban]
levorfanol
levorfanolo
27618j1n2x ,
levorphanol [hsdb]
levorphanol [inn]
levorphanol [mi]
levorphanol [who-dd]
levorphanol [vandf]
racemorphan [inn]
levorphanol dl-form [mi]
(+/-)-n-methyl-3-hydroxymorphinan
ids-nr-003
(+/-)-n-methylmorphinan-3-ol
(+/-)-3-hydroxy-n-methylmorphinan
morphinan-3-ol, 17-methyl-, (+/-)-
nu-2206
(-) 3-hydroxy-n-methylmorphinan
gtpl7595
SCHEMBL24335
9.alpha.,13.alpha.,14.alpha.-morphinan-3-ol, 17-methyl-
17-methylmorphinan-3-ol, (+)- #
d-form of levorphanol
DTXSID3023213 ,
(1r,9r,10r)-17-methyl-17-azatetracyclo[7.5.3.0^{1,10}.0^{2,7}]heptadeca-2(7),3,5-trien-4-ol
bdbm224028
l-morphan
Q2579108
DTXSID301016136
(1r,9r,10r)-17-methyl-17-azatetracyclo[7.5.3.01,10.02,7]heptadeca-2(7),3,5-trien-4-ol
(4br,8ar,9r)-11-methyl-6,7,8,8a,9,10-hexahydro-5h-9,4b-(epiminoethano)phenanthren-3-ol
77-07-6 (free)
(1r,9r,10r)-17-methyl-17-azatetracyclo[7.5.3.0?,??.0?,?]heptadeca-2,4,6-trien-4-ol
dtxcid203213
leuodromoran
2h-10,4a-iminoethanophenanthren-6-ol, 1,3,4,9,10,10a-hexahydro-11- methyl-, l-
levorphanolum (inn-latin)
levorfanol (inn-spanish)
1,3,4,9,10,10a-hexahydro-11-methyl-2h-10,4a-iminoethanophenanthren-6- ol, l-

Research Excerpts

Overview

Levorphanol is a synthetic strong opioid that is a potent N-methyl-d-aspartate receptor antagonist, mu, kappa, and delta opioid receptor agonist, and reuptake inhibitor of serotonin and norepinephrine. Levorphanol was first approved for use in the United States in 1953.

ExcerptReferenceRelevance
"Levorphanol is a potent opioid agonist and NMDA receptor blocker with minimal drug interactions, and there are few reports of its use in cancer patients."( Levorphanol as a Second Line Opioid in Cancer Patients Presenting to an Outpatient Supportive Care Center: An Open-label Study.
Amaram-Davila, J; Arthur, J; Bruera, E; Chavez, P; Dalal, S; De Moraes, AR; Dev, R; Epner, D; Guay, MOD; Haider, A; Hernandez, F; Hui, D; Mallipeddi, T; Nguyen, K; Reddy, A; Shelal, Z; Subbiah, I; Tanco, K; Wu, J, 2023
)
3.8
"Levorphanol is a synthetic strong opioid that is a potent N-methyl-d-aspartate receptor antagonist, mu, kappa, and delta opioid receptor agonist, and reuptake inhibitor of serotonin and norepinephrine."( Levorphanol for Treatment of Intractable Neuropathic Pain in Cancer Patients.
Bruera, E; Mallipeddi, T; Ng, A; Reddy, A, 2018
)
2.64
"Levorphanol is a long-acting opioid analgesic that is an optical isomer of dextrorphan, a metabolite of the over-the-counter cough suppressant dextromethorphan. "( Identifying Levorphanol Ingestion Using Urine Biomarkers in Health Care Patients.
Roberts, A; Watson, AR, 2018
)
2.3
"Levorphanol is a potent analgesic that has been used for decades. "( Pharmacological Characterization of Levorphanol, a G-Protein Biased Opioid Analgesic.
Hunkle, A; Le Rouzic, V; Lu, Z; Majumdar, S; Marrone, GF; Narayan, A; Pan, YX; Pasternak, GW; Xu, J, 2019
)
2.23
"Levorphanol is a potent opioid analgesic that was first approved for use in the United States in 1953. "( Levorphanol use: past, present and future.
Fudin, J; Gudin, J; Nalamachu, S, 2016
)
3.32
"Levorphanol is a widely used opiate analgesic. "( Kappa 3 receptors and levorphanol-induced analgesia.
Ginsberg, K; Pasternak, GW; Pick, CG; Tive, L, 1992
)
2.04

Effects

Levorphanol has no impact on QTc prolongation and considerably less drug-drug interactions as compared to methadone. It has a wide range of activities including mu opioid agonism, delta agonism and N-methyl-d-aspartate receptor antagonism.

ExcerptReferenceRelevance
"Levorphanol has a wide range of activities including mu opioid agonism, delta agonism, kappa1 and kappa3 receptor agonism, N-methyl-d-aspartate receptor antagonism and reuptake inhibition of both norepinephrine and serotonin."( Levorphanol use: past, present and future.
Fudin, J; Gudin, J; Nalamachu, S, 2016
)
2.6
"Levorphanol has no impact on QTc prolongation and considerably less drug-drug interactions as compared to methadone."( Levorphanol versus methadone use: safety considerations.
Haider, A; Reddy, A, 2020
)
2.72
"Levorphanol has a wide range of activities including mu opioid agonism, delta agonism, kappa1 and kappa3 receptor agonism, N-methyl-d-aspartate receptor antagonism and reuptake inhibition of both norepinephrine and serotonin."( Levorphanol use: past, present and future.
Fudin, J; Gudin, J; Nalamachu, S, 2016
)
2.6
"Levorphanol has clinical efficacy in neuropathic pain."( Levorphanol: the forgotten opioid.
Prommer, E, 2007
)
2.5
"Levorphanol has been reported to provide analgesia at doses that suggest it does not act like other pure agonist opioids. "( Can levorphanol be used like methadone for intractable refractory pain?
McNulty, JP, 2007
)
2.34

Treatment

ExcerptReferenceRelevance
"Levorphanol pretreatment reduced the Hill coefficient (HN) of 1.5 to 0.7, suggesting cooperative interaction between the opiate receptor and the enzyme protein."( Opiates inhibit calmodulin activation of a high-affinity Ca2+-stimulated Mg2+-dependent ATPase in synaptic membranes.
Cardenas, HL; Ross, DH, 1987
)
0.99

Toxicity

ExcerptReferenceRelevance
" DX also antagonized morphological and chemical (lactate dehydrogenase efflux) evidence of cortical neuronal cell injury produced by toxic bath exposure to NMDA, quinolinate or glutamate, but did not affect toxic exposure to quisqualate or kainate."( Dextrorphan and levorphanol selectively block N-methyl-D-aspartate receptor-mediated neurotoxicity on cortical neurons.
Choi, DW; Peters, S; Viseskul, V, 1987
)
0.62

Pharmacokinetics

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
)
0.27
" 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
)
0.27

Bioavailability

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
)
0.26
"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
)
0.28
"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
)
0.27
" 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
)
0.27

Dosage Studied

ExcerptRelevanceReference
" Morphine dose-response curves were determined in the offspring at 5 weeks of age."( Prenatal administration of morphine to the rat: tolerance to the analgesic effect of morphine in the offspring.
Holtzman, SG; O'Callaghan, JP, 1976
)
0.26
" The dextrorphan-trained birds generalized to l-cyclorphan at 10 mg/kg; naltrexone did not alter the l-cyclorphan dose-response curve for this effect."( Discriminative stimulus effects of cyclorphan: selective antagonism with naltrexone.
Bertalmio, AJ; Woods, JH, 1992
)
0.28
" CI was always preceded by a period of repetitive dosing of opioids."( I.v. infusion of opioids for cancer pain: clinical review and guidelines for use.
Foley, KM; Inturrisi, CE; Moulin, DE; Portenoy, RK; Rogers, A, 1986
)
0.27
" It had weak PCP-like bioactivity, but could antagonize PCP's action dose-dependently in vitro study with the rabbit ear artery preparation and shifted the dose-response curve of PCP to the right."( Dextrorphan: an antagonist for phencyclidine receptors.
Sun, FY; Xu, XR; Zhang, AZ; Zhang, LM; Zhu, H, 1987
)
0.27
", 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
)
0.27
" After 12 generations of selective breeding, the high antinociceptive response line exhibited about 7 times steeper dose-response curve than did the low antinociceptive response line whereas only small differences were seen with saline alone."( Selective breeding for levorphanol-induced antinociception on the hot-plate assay: commonalities in mechanism of action with morphine, pentazocine, ethylketocyclazocine, U-50488H and clonidine in mice.
Belknap, JK; Danielson, PW; Laursen, SE; Noordewier, B, 1987
)
0.58
" 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
)
0.27
" ICI 154,129 was proconvulsant in the mouse picrotoxin potentiation test; the dose-response curve had a low ceiling and was biphasic."( In vivo studies with ICI 154,129, a putative delta receptor antagonist.
Cowan, A; Gmerek, DE,
)
0.13
"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
)
0.27
"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
)
0.27
" 2 The dose-response curves to naloxone obtained in tissues individually exposed to different opiates showed that their relative potency in increasing sensitivity to naloxone was as follows: levorphan greater than morphine greater than Met-enkephalin greater than nalorphine greater than pentazocine."( Pharmacological characterization of opiate physical dependence in the isolated ileum of the guinea-pig.
Luján, M; Rodríguez, R, 1981
)
0.26
" Dose-response and time-course determinations were performed with various opioids."( Sex-related differences in the antinociceptive effects of opioids: importance of rat genotype, nociceptive stimulus intensity, and efficacy at the mu opioid receptor.
Barrett, AC; Bowman, JR; Cook, CD; Picker, MJ; Roach, EL, 2000
)
0.31
" Tolerance and OIH were inferred individually based on chronic pain ratings, brief pain inventory scores, and results of the brief thermal sensitization model at 5 opioid dosing sessions."( Evolution of Analgesic Tolerance and Opioid-Induced Hyperalgesia Over 6 Months: Double-Blind Randomized Trial Incorporating Experimental Pain Models.
Rowbotham, MC; Wallace, M,
)
0.13
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Drug Classes (1)

ClassDescription
morphinane alkaloidAn isoquinoline alkaloid based on a morphinan skeleton and its substituted derivatives.
[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 (1)

PathwayProteinsCompounds
Levorphanol Action Pathway3111

Protein Targets (26)

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Solute carrier family 22 member 1 Homo sapiens (human)IC50 (µMol)8.00000.21005.553710.0000AID1526751
Glutamate receptor ionotropic, NMDA 2DHomo sapiens (human)IC50 (µMol)2.60000.00401.73519.8000AID143909; AID143911
AcetylcholinesteraseElectrophorus electricus (electric eel)Ki25.00000.00121.25638.9000AID514203
Sigma non-opioid intracellular receptor 1Mus musculus (house mouse)Ki0.00190.00190.20180.8200AID204010
Glutamate receptor ionotropic, NMDA 3BHomo sapiens (human)IC50 (µMol)2.60000.00401.65799.8000AID143909; AID143911
Kappa-type opioid receptorMus musculus (house mouse)Ki0.01000.00030.35942.7500AID148126
Mu-type opioid receptorHomo sapiens (human)IC50 (µMol)0.00010.00010.813310.0000AID150822
Mu-type opioid receptorHomo sapiens (human)Ki0.00020.00000.419710.0000AID148328; AID151001; AID259394; AID290256; AID362196; AID670110
Glutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)IC50 (µMol)4.45000.00071.600310.0000AID144475; AID144477
Delta-type opioid receptorHomo sapiens (human)IC50 (µMol)0.00400.00020.75218.0140AID148090; AID149186
Delta-type opioid receptorHomo sapiens (human)Ki0.00420.00000.59789.9300AID147944; AID148369; AID149778; AID259395; AID290258; AID362197; AID670113
Kappa-type opioid receptorCavia porcellus (domestic guinea pig)Ki5.00260.00000.20186.4240AID147944; AID149121; AID514203
Kappa-type opioid receptorHomo sapiens (human)IC50 (µMol)0.00400.00001.201110.0000AID147865
Kappa-type opioid receptorHomo sapiens (human)Ki0.00230.00000.362410.0000AID148149; AID149978; AID259396; AID290257; AID362198; AID670111
Mu-type opioid receptorMus musculus (house mouse)Ki0.00030.00000.12281.3000AID149172
Mu-type opioid receptorCavia porcellus (domestic guinea pig)IC50 (µMol)0.00060.00020.660310.0000AID148996
Mu-type opioid receptorCavia porcellus (domestic guinea pig)Ki0.00020.00000.27869.0000AID147903
Glutamate receptor ionotropic, NMDA 2A Rattus norvegicus (Norway rat)IC50 (µMol)4.45000.00071.630610.0000AID144475; AID144477
Glutamate receptor ionotropic, NMDA 2BRattus norvegicus (Norway rat)IC50 (µMol)4.45000.00061.525710.0000AID144475; AID144477
Glutamate receptor ionotropic, NMDA 2CRattus norvegicus (Norway rat)IC50 (µMol)4.45000.00071.747210.0000AID144475; AID144477
Glutamate receptor ionotropic, NMDA 1Homo sapiens (human)IC50 (µMol)2.60000.00101.88779.8000AID143909; AID143911
Glutamate receptor ionotropic, NMDA 2AHomo sapiens (human)IC50 (µMol)2.60000.00101.99589.8000AID143909; AID143911
Glutamate receptor ionotropic, NMDA 2BHomo sapiens (human)IC50 (µMol)2.60000.00401.33259.8000AID143909; AID143911
Glutamate receptor ionotropic, NMDA 2CHomo sapiens (human)IC50 (µMol)2.60000.00401.86339.8000AID143909; AID143911
Glutamate receptor ionotropic, NMDA 2DRattus norvegicus (Norway rat)IC50 (µMol)4.45000.00071.741110.0000AID144475; AID144477
Glutamate receptor ionotropic, NMDA 3AHomo sapiens (human)IC50 (µMol)2.60000.00401.65799.8000AID143909; AID143911
Glutamate receptor ionotropic, NMDA 3BRattus norvegicus (Norway rat)IC50 (µMol)4.45000.00071.741110.0000AID144475; AID144477
Glutamate receptor ionotropic, NMDA 3ARattus norvegicus (Norway rat)IC50 (µMol)4.45000.00071.741110.0000AID144475; AID144477
[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)
Mas-related G-protein coupled receptor member X2Homo sapiens (human)EC50 (µMol)17.50000.14003.73818.9000AID1802708; AID1802709
[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)
Delta-type opioid receptorMus musculus (house mouse)Ke0.00060.00010.14726.1080AID148951
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (140)

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)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerMu-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled acetylcholine receptor signaling pathwayMu-type opioid receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayMu-type opioid receptorHomo sapiens (human)
sensory perceptionMu-type opioid receptorHomo sapiens (human)
negative regulation of cell population proliferationMu-type opioid receptorHomo sapiens (human)
sensory perception of painMu-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor signaling pathwayMu-type opioid receptorHomo sapiens (human)
behavioral response to ethanolMu-type opioid receptorHomo sapiens (human)
positive regulation of neurogenesisMu-type opioid receptorHomo sapiens (human)
negative regulation of Wnt protein secretionMu-type opioid receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeMu-type opioid receptorHomo sapiens (human)
calcium ion transmembrane transportMu-type opioid receptorHomo sapiens (human)
cellular response to morphineMu-type opioid receptorHomo sapiens (human)
regulation of cellular response to stressMu-type opioid receptorHomo sapiens (human)
regulation of NMDA receptor activityMu-type opioid receptorHomo sapiens (human)
neuropeptide signaling pathwayMu-type opioid receptorHomo sapiens (human)
immune responseDelta-type opioid receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayDelta-type opioid receptorHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerDelta-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwayDelta-type opioid receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayDelta-type opioid receptorHomo sapiens (human)
adult locomotory behaviorDelta-type opioid receptorHomo sapiens (human)
negative regulation of gene expressionDelta-type opioid receptorHomo sapiens (human)
negative regulation of protein-containing complex assemblyDelta-type opioid receptorHomo sapiens (human)
positive regulation of CREB transcription factor activityDelta-type opioid receptorHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationDelta-type opioid receptorHomo sapiens (human)
response to nicotineDelta-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor signaling pathwayDelta-type opioid receptorHomo sapiens (human)
eating behaviorDelta-type opioid receptorHomo sapiens (human)
regulation of mitochondrial membrane potentialDelta-type opioid receptorHomo sapiens (human)
regulation of calcium ion transportDelta-type opioid receptorHomo sapiens (human)
cellular response to growth factor stimulusDelta-type opioid receptorHomo sapiens (human)
cellular response to hypoxiaDelta-type opioid receptorHomo sapiens (human)
cellular response to toxic substanceDelta-type opioid receptorHomo sapiens (human)
neuropeptide signaling pathwayDelta-type opioid receptorHomo sapiens (human)
immune responseKappa-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting G protein-coupled receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
chemical synaptic transmissionKappa-type opioid receptorHomo sapiens (human)
sensory perceptionKappa-type opioid receptorHomo sapiens (human)
locomotory behaviorKappa-type opioid receptorHomo sapiens (human)
sensory perception of painKappa-type opioid receptorHomo sapiens (human)
adenylate cyclase-inhibiting opioid receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
response to insulinKappa-type opioid receptorHomo sapiens (human)
positive regulation of dopamine secretionKappa-type opioid receptorHomo sapiens (human)
negative regulation of luteinizing hormone secretionKappa-type opioid receptorHomo sapiens (human)
response to nicotineKappa-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor signaling pathwayKappa-type opioid receptorHomo sapiens (human)
maternal behaviorKappa-type opioid receptorHomo sapiens (human)
eating behaviorKappa-type opioid receptorHomo sapiens (human)
response to estrogenKappa-type opioid receptorHomo sapiens (human)
estrous cycleKappa-type opioid receptorHomo sapiens (human)
response to ethanolKappa-type opioid receptorHomo sapiens (human)
regulation of saliva secretionKappa-type opioid receptorHomo sapiens (human)
behavioral response to cocaineKappa-type opioid receptorHomo sapiens (human)
sensory perception of temperature stimulusKappa-type opioid receptorHomo sapiens (human)
defense response to virusKappa-type opioid receptorHomo sapiens (human)
cellular response to lipopolysaccharideKappa-type opioid receptorHomo sapiens (human)
cellular response to glucose stimulusKappa-type opioid receptorHomo sapiens (human)
positive regulation of p38MAPK cascadeKappa-type opioid receptorHomo sapiens (human)
positive regulation of potassium ion transmembrane transportKappa-type opioid receptorHomo sapiens (human)
response to acrylamideKappa-type opioid receptorHomo sapiens (human)
positive regulation of eating behaviorKappa-type opioid receptorHomo sapiens (human)
conditioned place preferenceKappa-type opioid receptorHomo sapiens (human)
neuropeptide signaling pathwayKappa-type opioid receptorHomo 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)
sensory perception of painMas-related G-protein coupled receptor member X2Homo sapiens (human)
sleepMas-related G-protein coupled receptor member X2Homo sapiens (human)
positive regulation of cytokinesisMas-related G-protein coupled receptor member X2Homo sapiens (human)
mast cell degranulationMas-related G-protein coupled receptor member X2Homo sapiens (human)
mast cell activationMas-related G-protein coupled receptor member X2Homo sapiens (human)
G protein-coupled receptor signaling pathwayMas-related G-protein coupled receptor member X2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (51)

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)
G-protein alpha-subunit bindingMu-type opioid receptorHomo sapiens (human)
G protein-coupled receptor activityMu-type opioid receptorHomo sapiens (human)
beta-endorphin receptor activityMu-type opioid receptorHomo sapiens (human)
voltage-gated calcium channel activityMu-type opioid receptorHomo sapiens (human)
protein bindingMu-type opioid receptorHomo sapiens (human)
morphine receptor activityMu-type opioid receptorHomo sapiens (human)
G-protein beta-subunit bindingMu-type opioid receptorHomo sapiens (human)
neuropeptide bindingMu-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor activityDelta-type opioid receptorHomo sapiens (human)
protein bindingDelta-type opioid receptorHomo sapiens (human)
receptor serine/threonine kinase bindingDelta-type opioid receptorHomo sapiens (human)
G protein-coupled enkephalin receptor activityDelta-type opioid receptorHomo sapiens (human)
neuropeptide bindingDelta-type opioid receptorHomo sapiens (human)
G protein-coupled opioid receptor activityKappa-type opioid receptorHomo sapiens (human)
protein bindingKappa-type opioid receptorHomo sapiens (human)
receptor serine/threonine kinase bindingKappa-type opioid receptorHomo sapiens (human)
dynorphin receptor activityKappa-type opioid receptorHomo sapiens (human)
neuropeptide bindingKappa-type opioid receptorHomo 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 receptor activityMas-related G-protein coupled receptor member X2Homo sapiens (human)
neuropeptide bindingMas-related G-protein coupled receptor member X2Homo sapiens (human)
mast cell secretagogue receptor activityMas-related G-protein coupled receptor member X2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (47)

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)
plasma membraneKappa-type opioid receptorMus musculus (house mouse)
endosomeMu-type opioid receptorHomo sapiens (human)
endoplasmic reticulumMu-type opioid receptorHomo sapiens (human)
Golgi apparatusMu-type opioid receptorHomo sapiens (human)
plasma membraneMu-type opioid receptorHomo sapiens (human)
axonMu-type opioid receptorHomo sapiens (human)
dendriteMu-type opioid receptorHomo sapiens (human)
perikaryonMu-type opioid receptorHomo sapiens (human)
synapseMu-type opioid receptorHomo sapiens (human)
plasma membraneMu-type opioid receptorHomo sapiens (human)
neuron projectionMu-type opioid receptorHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)
plasma membraneGlutamate receptor ionotropic, NMDA 1 Rattus norvegicus (Norway rat)
plasma membraneDelta-type opioid receptorHomo sapiens (human)
synaptic vesicle membraneDelta-type opioid receptorHomo sapiens (human)
dendrite membraneDelta-type opioid receptorHomo sapiens (human)
presynaptic membraneDelta-type opioid receptorHomo sapiens (human)
axon terminusDelta-type opioid receptorHomo sapiens (human)
spine apparatusDelta-type opioid receptorHomo sapiens (human)
postsynaptic density membraneDelta-type opioid receptorHomo sapiens (human)
neuronal dense core vesicleDelta-type opioid receptorHomo sapiens (human)
plasma membraneDelta-type opioid receptorHomo sapiens (human)
neuron projectionDelta-type opioid receptorHomo sapiens (human)
nucleoplasmKappa-type opioid receptorHomo sapiens (human)
mitochondrionKappa-type opioid receptorHomo sapiens (human)
cytosolKappa-type opioid receptorHomo sapiens (human)
plasma membraneKappa-type opioid receptorHomo sapiens (human)
membraneKappa-type opioid receptorHomo sapiens (human)
sarcoplasmic reticulumKappa-type opioid receptorHomo sapiens (human)
T-tubuleKappa-type opioid receptorHomo sapiens (human)
dendriteKappa-type opioid receptorHomo sapiens (human)
synaptic vesicle membraneKappa-type opioid receptorHomo sapiens (human)
presynaptic membraneKappa-type opioid receptorHomo sapiens (human)
perikaryonKappa-type opioid receptorHomo sapiens (human)
axon terminusKappa-type opioid receptorHomo sapiens (human)
postsynaptic membraneKappa-type opioid receptorHomo sapiens (human)
plasma membraneKappa-type opioid receptorHomo sapiens (human)
neuron projectionKappa-type opioid receptorHomo sapiens (human)
plasma membraneMu-type opioid receptorMus musculus (house mouse)
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)
membraneMas-related G-protein coupled receptor member X2Homo sapiens (human)
plasma membraneMas-related G-protein coupled receptor member X2Homo 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 (109)

Assay IDTitleYearJournalArticle
AID1346364Human mu receptor (Opioid receptors)2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
The power of visual imagery in drug design. Isopavines as a new class of morphinomimetics and their human opioid receptor binding activity.
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.
AID148036Binding affinity towards Opioid receptor mu 1 in guinea pig brain membranes using [3H]DAMGO as radioligand2001Bioorganic & medicinal chemistry letters, Oct-22, Volume: 11, Issue:20
Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan.
AID1145608Drug absorption in anesthetized rat colon at pH 6.81977Journal of medicinal chemistry, Jan, Volume: 20, Issue:1
Use of distribution coefficients in quantitative structure-activity relationships.
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.
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.
AID184526Effect was expressed as mortality after injection of kainic acid (10 mg/kg) and the drug at the dose of 40 mg/Kg after 48 hr2001Bioorganic & medicinal chemistry letters, Jul-09, Volume: 11, Issue:13
Anticonvulsant effects of new morphinan derivatives.
AID148951Inhibition of naloxone activity in mouse vas deferens assay for Opioid receptor delta 11982Journal of medicinal chemistry, Dec, Volume: 25, Issue:12
Synthesis and pharmacological characterization in vitro of cyclic enkephalin analogues: effect of conformational constraints on opiate receptor selectivity.
AID362197Displacement of [3H]Naltindole from human delta opioid receptor expressed in CHO cell membrane2008Bioorganic & medicinal chemistry letters, Aug-15, Volume: 18, Issue:16
Synthesis and pharmacological evaluation of hydrophobic esters and ethers of butorphanol at opioid receptors.
AID148126Binding affinity for mouse opioid receptor kappa1989Journal of medicinal chemistry, Sep, Volume: 32, Issue:9
Benzofuro[2,3-c]pyridin-6-ols: synthesis, affinity for opioid-receptor subtypes, and antinociceptive activity.
AID148149Inhibitory activity against Opioid receptor kappa 1 in chinese Hamster Ovary (CHO) cell membranes was determined using [3H]U-69593 radioligand2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of novel dimeric morphinan ligands for kappa and micro opioid receptors.
AID149867Inhibitory potency against Opioid receptor mu 1 in the guinea pig ileum assay1982Journal of medicinal chemistry, Dec, Volume: 25, Issue:12
Synthesis and pharmacological characterization in vitro of cyclic enkephalin analogues: effect of conformational constraints on opiate receptor selectivity.
AID149121Binding affinity against opioid receptor kappa 1 using [3H]-U-69,593 as radioligand in guinea pig brain membranes.2004Journal of medicinal chemistry, Jan-01, Volume: 47, Issue:1
10-Ketomorphinan and 3-substituted-3-desoxymorphinan analogues as mixed kappa and micro opioid ligands: synthesis and biological evaluation of their binding affinity at opioid receptors.
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.
AID112925Analgesic activity in mice after subcutaneous administration in tail-flicking assay; 1.13-1.311985Journal of medicinal chemistry, Sep, Volume: 28, Issue:9
Acylmorphinans. A novel class of potent analgesic agents.
AID151914Binding constant for the antagonist state was measured for its ability to displace [3H]naloxone from opioid mu 1 receptor buffered homogenate of rat brain membranes1988Journal of medicinal chemistry, Mar, Volume: 31, Issue:3
Structure-activity relationships for drugs binding to the agonist and antagonist states of the primary morphine receptor.
AID670115Selectivity index, ratio of inhibition of human delta opioid receptor to inhibition of human mu opioid receptor2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Synthesis, binding affinity, and functional in vitro activity of 3-benzylaminomorphinan and 3-benzylaminomorphine ligands at opioid receptors.
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.
AID112915Analgesic activity in mice after oral administration in writhing assay; 0.70-2.801985Journal of medicinal chemistry, Sep, Volume: 28, Issue:9
Acylmorphinans. A novel class of potent analgesic agents.
AID624620Specific activity of expressed human recombinant UGT2B7H2000Annual review of pharmacology and toxicology, , Volume: 40Human UDP-glucuronosyltransferases: metabolism, expression, and disease.
AID290260Selectivity for kappa opioid receptor over delta opioid receptor2007Bioorganic & medicinal chemistry letters, Mar-15, Volume: 17, Issue:6
High-affinity carbamate analogues of morphinan at opioid receptors.
AID624621Specific activity of expressed human recombinant UGT2B7Y2000Annual review of pharmacology and toxicology, , Volume: 40Human UDP-glucuronosyltransferases: metabolism, expression, and disease.
AID1135662Displacement of [3H]naloxone from opioid receptor (unknown origin) in brain homogenate measured after four washes1978Journal of medicinal chemistry, Jul, Volume: 21, Issue:7
6beta-[N,N-Bis(2-chloroethyl)amino]-17-(cyclopropylmethyl)-4,5alpha-epoxy-3,14-dihydroxymorphinan(chlornaltrexamine) a potent opioid receptor alkylating agent with ultralong narcotic antagonist actitivty.
AID149272Binding affinity towards Opioid receptor kappa 1 in guinea pig brain membranes using [3H]U-69593 as radioligand2001Bioorganic & medicinal chemistry letters, Oct-22, Volume: 11, Issue:20
Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan.
AID1123188Displacement of [3H]-naloxone from opioid receptor in rat brain homogenate at 1x10'-8 M preincubated for 5 mins followed by washout measured after 15 mins by liquid scintillation counting analysis relative to control1979Journal of medicinal chemistry, Feb, Volume: 22, Issue:2
Synthesis and pharmacologic characterization of an alkylating analogue (chlornaltrexamine) of naltrexone with ultralong-lasting narcotic antagonist properties.
AID290259Selectivity for kappa opioid receptor over mu opioid receptor expressed in CHO cells2007Bioorganic & medicinal chemistry letters, Mar-15, Volume: 17, Issue:6
High-affinity carbamate analogues of morphinan at opioid receptors.
AID149778Binding affinity against opioid receptor delta 1 using [3H]naltrindole as radioligand in guinea pig brain membranes.2004Journal of medicinal chemistry, Jan-01, Volume: 47, Issue:1
10-Ketomorphinan and 3-substituted-3-desoxymorphinan analogues as mixed kappa and micro opioid ligands: synthesis and biological evaluation of their binding affinity at opioid receptors.
AID170083The compound was examined for circling behavior (marginal activity) using automated videotracking system 30 min after intraperitoneal injection at the dose of 40 mg/Kg in 12 rats2001Bioorganic & medicinal chemistry letters, Jul-09, Volume: 11, Issue:13
Anticonvulsant effects of new morphinan derivatives.
AID233831Selective inhibitory activity was determined against kappa & mu opioid receptor in chinese Hamster Ovary (CHO) cell membranes2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of novel dimeric morphinan ligands for kappa and micro opioid receptors.
AID149868Inhibitory potency against Opioid receptor mu 1 in mouse vas deferens assay1982Journal of medicinal chemistry, Dec, Volume: 25, Issue:12
Synthesis and pharmacological characterization in vitro of cyclic enkephalin analogues: effect of conformational constraints on opiate receptor selectivity.
AID143909Inhibition of [3H]1 binding to dextromethorpin binding site of guinea pig microsomal pellet P3 N-methyl-D-aspartate glutamate receptor1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID144472Binding affinity against glycine binding site associated with N-methyl-D-aspartate glutamate receptor from rat synaptic plasma membrane(SPM) determined using [3H]glycine as radioligand.;NA = No effect1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID362198Displacement of [3H]U69593 from human kappa opioid receptor expressed in CHO cell membrane2008Bioorganic & medicinal chemistry letters, Aug-15, Volume: 18, Issue:16
Synthesis and pharmacological evaluation of hydrophobic esters and ethers of butorphanol at opioid receptors.
AID229388Relative affinity for delta and kappa opioid receptors2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
2-aminothiazole-derived opioids. Bioisosteric replacement of phenols.
AID362201Selectivity ratio of Ki for human mu opioid receptor to Ki for human kappa opioid receptor2008Bioorganic & medicinal chemistry letters, Aug-15, Volume: 18, Issue:16
Synthesis and pharmacological evaluation of hydrophobic esters and ethers of butorphanol at opioid receptors.
AID235164Selectivity ratio expressed as Ki of delta opioid receptor to that of mu opioid receptor.2004Journal of medicinal chemistry, Jan-01, Volume: 47, Issue:1
10-Ketomorphinan and 3-substituted-3-desoxymorphinan analogues as mixed kappa and micro opioid ligands: synthesis and biological evaluation of their binding affinity at opioid receptors.
AID235163Selectivity ratio expressed as Ki of delta opioid receptor to that of kappa opioid receptor.2004Journal of medicinal chemistry, Jan-01, Volume: 47, Issue:1
10-Ketomorphinan and 3-substituted-3-desoxymorphinan analogues as mixed kappa and micro opioid ligands: synthesis and biological evaluation of their binding affinity at opioid receptors.
AID128507Analgesic activity was determined by writhing test after subcutaneous administration in mice1982Journal of medicinal chemistry, Oct, Volume: 25, Issue:10
Synthesis and pharmacology of metabolically stable tert-butyl ethers of morphine and levorphanol.
AID188507Anticonvulsant activity in rat supramaximal electroshock (MES) test expressed as % rats protected at a dose of 2.5-10 mg/kg.1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID170082The compound was examined for circling behavior (marginal activity) using automated videotracking system 30 min after intraperitoneal injection at the dose of 20 mg/Kg in 12 rats2001Bioorganic & medicinal chemistry letters, Jul-09, Volume: 11, Issue:13
Anticonvulsant effects of new morphinan derivatives.
AID362196Displacement of [3H]DAMGO from human mu opioid receptor expressed in CHO cell membrane2008Bioorganic & medicinal chemistry letters, Aug-15, Volume: 18, Issue:16
Synthesis and pharmacological evaluation of hydrophobic esters and ethers of butorphanol at opioid receptors.
AID670116Selectivity index, ratio of inhibition of human kappa opioid receptor to inhibition of human mu opioid receptor2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Synthesis, binding affinity, and functional in vitro activity of 3-benzylaminomorphinan and 3-benzylaminomorphine ligands at opioid receptors.
AID670113Displacement of [3H]-naltrindole from human delta opioid receptor expressed in CHO cells after 3 hrs by scintillation counting2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Synthesis, binding affinity, and functional in vitro activity of 3-benzylaminomorphinan and 3-benzylaminomorphine ligands at opioid receptors.
AID204010Binding affinity for mouse sigma opioid receptor1989Journal of medicinal chemistry, Sep, Volume: 32, Issue:9
Benzofuro[2,3-c]pyridin-6-ols: synthesis, affinity for opioid-receptor subtypes, and antinociceptive activity.
AID147865In vitro binding affinity against cloned human Opioid receptor kappa 1 expressed in HEK 293S cells2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
The power of visual imagery in drug design. Isopavines as a new class of morphinomimetics and their human opioid receptor binding activity.
AID259395Displacement of [3H]naltrindole from human delta opioid receptor expressed in CHO cells2006Journal of medicinal chemistry, Jan-12, Volume: 49, Issue:1
Synthesis and preliminary in vitro investigation of bivalent ligands containing homo- and heterodimeric pharmacophores at mu, delta, and kappa opioid receptors.
AID148369Inhibitory activity against Opioid receptor delta 1 in chinese Hamster Ovary (CHO) cell membranes was determined using [3H]naltrindole radioligand2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of novel dimeric morphinan ligands for kappa and micro opioid receptors.
AID178286Anticonvulsant activity in rat supramaximal electroshock (MES) test within a dose range of 15-60 mg/kg administered subcutaneously1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID144477Binding affinity against PCP binding site associated with N-methyl-D-aspartate glutamate receptor from rat synaptic plasma membrane(SPM) determined using [3H]TCP as radioligand.1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID112912Analgesic activity in mice after oral administration in tail-flicking assay; 7.22-10.251985Journal of medicinal chemistry, Sep, Volume: 28, Issue:9
Acylmorphinans. A novel class of potent analgesic agents.
AID362199Selectivity ratio of Ki for human kappa opioid receptor to Ki for human delta opioid receptor2008Bioorganic & medicinal chemistry letters, Aug-15, Volume: 18, Issue:16
Synthesis and pharmacological evaluation of hydrophobic esters and ethers of butorphanol at opioid receptors.
AID290257Displacement of [3H]U-69593 from human kappa opioid receptors expressed in CHO cell membrane2007Bioorganic & medicinal chemistry letters, Mar-15, Volume: 17, Issue:6
High-affinity carbamate analogues of morphinan at opioid receptors.
AID1145609Octanol-aqueous phase partition coefficient, log P of the compound at pH 7.101977Journal of medicinal chemistry, Jan, Volume: 20, Issue:1
Use of distribution coefficients in quantitative structure-activity relationships.
AID112932Analgesic activity in mice after subcutaneous administration in writhing assay; 0.06-0.221985Journal of medicinal chemistry, Sep, Volume: 28, Issue:9
Acylmorphinans. A novel class of potent analgesic agents.
AID23717Partition coefficient (logP)1988Journal of medicinal chemistry, Mar, Volume: 31, Issue:3
Structure-activity relationships for drugs binding to the agonist and antagonist states of the primary morphine receptor.
AID235330Selectivity ratio of kappa opioid receptor to that of mu opioid receptor in guinea pig brain membranes2001Bioorganic & medicinal chemistry letters, Oct-22, Volume: 11, Issue:20
Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan.
AID149978Inhibition of [3H]U-69593 binding to Opioid receptor kappa 1 of Chinese hamster ovary membrane2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
2-aminothiazole-derived opioids. Bioisosteric replacement of phenols.
AID150244Concentration required to inhibit [3H]naltrexone binding to Opioid receptors1982Journal of medicinal chemistry, Oct, Volume: 25, Issue:10
Synthesis and pharmacology of metabolically stable tert-butyl ethers of morphine and levorphanol.
AID1123189Displacement of [3H]-naloxone from opioid receptor in rat brain homogenate at 1x10'-8 M preincubated for 5 mins measured after second washout by liquid scintillation counting analysis relative to control1979Journal of medicinal chemistry, Feb, Volume: 22, Issue:2
Synthesis and pharmacologic characterization of an alkylating analogue (chlornaltrexamine) of naltrexone with ultralong-lasting narcotic antagonist properties.
AID147893Sensitivity to naloxone against Opioid receptor mu 1 in the guinea pig ileum assay1982Journal of medicinal chemistry, Dec, Volume: 25, Issue:12
Synthesis and pharmacological characterization in vitro of cyclic enkephalin analogues: effect of conformational constraints on opiate receptor selectivity.
AID151001Inhibitory activity against Opioid receptor mu 1 in chinese Hamster Ovary (CHO) cells membranes was determined using [3H]-DAMGO radioligand2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of novel dimeric morphinan ligands for kappa and micro opioid receptors.
AID290258Displacement of [3H]naltrindole from human delta opioid receptors expressed in CHO cell membrane2007Bioorganic & medicinal chemistry letters, Mar-15, Volume: 17, Issue:6
High-affinity carbamate analogues of morphinan at opioid receptors.
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.
AID1150953Displacement of [3H]etorphine from opioid receptor in guinea pig brain homogenate by scintillation counting analysis1976Journal of medicinal chemistry, Jun, Volume: 19, Issue:6
Stereospecific interaction of the quaternized opiate, N-methyllevorphanol, with opiate receptors.
AID205268Inhibition of binding of Batrachotoxinin [3H]BTX-B to high affinity sites on voltage dependent sodium channels in a vesicular preparation from guinea pig cerebral cortex at 10 uM1985Journal of medicinal chemistry, Mar, Volume: 28, Issue:3
[3H]Batrachotoxinin A 20 alpha-benzoate binding to voltage-sensitive sodium channels: a rapid and quantitative assay for local anesthetic activity in a variety of drugs.
AID128490Analgesic activity 30 min after subcutaneous administration in mouse by using PQW test; NT=Not tested1989Journal of medicinal chemistry, Sep, Volume: 32, Issue:9
Benzofuro[2,3-c]pyridin-6-ols: synthesis, affinity for opioid-receptor subtypes, and antinociceptive activity.
AID148996Inhibitory activity against opioid receptor mu of guinea pig brain using [3H]DAGO radioligand1989Journal of medicinal chemistry, Jun, Volume: 32, Issue:6
Synthesis, opioid receptor binding profile, and antinociceptive activity of 1-azaspiro[4.5]decan-10-yl amides.
AID205267Inhibition of binding of Batrachotoxinin [3H]BTX-B to high affinity sites on voltage dependent sodium channels in a vesicular preparation from guinea pig cerebral cortex1985Journal of medicinal chemistry, Mar, Volume: 28, Issue:3
[3H]Batrachotoxinin A 20 alpha-benzoate binding to voltage-sensitive sodium channels: a rapid and quantitative assay for local anesthetic activity in a variety of drugs.
AID149186Inhibitory activity against opioid receptor delta of guinea pig was determined by using [3H]DADLE radioligand1989Journal of medicinal chemistry, Jun, Volume: 32, Issue:6
Synthesis, opioid receptor binding profile, and antinociceptive activity of 1-azaspiro[4.5]decan-10-yl amides.
AID259394Displacement of [3H]DAMGO from human mu opioid receptor expressed in CHO cells2006Journal of medicinal chemistry, Jan-12, Volume: 49, Issue:1
Synthesis and preliminary in vitro investigation of bivalent ligands containing homo- and heterodimeric pharmacophores at mu, delta, and kappa opioid receptors.
AID144475Binding affinity against dextromethorpin binding site of N-methyl-D-aspartate glutamate receptor from rat brain using [3H]11992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID149904Binding affinity towards Opioid receptor delta 1 in guinea pig brain membranes using [3H]naltrindole as radioligand2001Bioorganic & medicinal chemistry letters, Oct-22, Volume: 11, Issue:20
Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan.
AID1145607Octanol-aqueous phase distribution coefficient, log D of the compound1977Journal of medicinal chemistry, Jan, Volume: 20, Issue:1
Use of distribution coefficients in quantitative structure-activity relationships.
AID290256Displacement of [3H]DAMGO from human mu opioid receptors expressed in CHO cell membrane2007Bioorganic & medicinal chemistry letters, Mar-15, Volume: 17, Issue:6
High-affinity carbamate analogues of morphinan at opioid receptors.
AID196182The compound was tested for anticonvulsive activity expressed as seizure score after 4 hr of injection of kainic acid (10 mg/kg) and the drug at the dose of 40 mg/Kg in 15 rats2001Bioorganic & medicinal chemistry letters, Jul-09, Volume: 11, Issue:13
Anticonvulsant effects of new morphinan derivatives.
AID150822In vitro binding affinity against cloned human Opioid receptor mu 1 expressed in HEK 293S cells2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
The power of visual imagery in drug design. Isopavines as a new class of morphinomimetics and their human opioid receptor binding activity.
AID196181The compound was tested for anticonvulsive activity expressed as seizure score after 4 hr of injection of kainic acid (10 mg/kg) and the drug at the dose of 20 mg/Kg in 15 rats2001Bioorganic & medicinal chemistry letters, Jul-09, Volume: 11, Issue:13
Anticonvulsant effects of new morphinan derivatives.
AID235329Selectivity ratio of kappa opioid receptor to that of delta opioid receptor in guinea pig brain membranes2001Bioorganic & medicinal chemistry letters, Oct-22, Volume: 11, Issue:20
Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan.
AID1123191Displacement of [3H]-naloxone from opioid receptor in rat brain homogenate at 1x10'-8 M preincubated for 5 mins measured after fourth washout by liquid scintillation counting analysis relative to control1979Journal of medicinal chemistry, Feb, Volume: 22, Issue:2
Synthesis and pharmacologic characterization of an alkylating analogue (chlornaltrexamine) of naltrexone with ultralong-lasting narcotic antagonist properties.
AID1123190Displacement of [3H]-naloxone from opioid receptor in rat brain homogenate at 1x10'-8 M preincubated for 5 mins measured after third washout by liquid scintillation counting analysis relative to control1979Journal of medicinal chemistry, Feb, Volume: 22, Issue:2
Synthesis and pharmacologic characterization of an alkylating analogue (chlornaltrexamine) of naltrexone with ultralong-lasting narcotic antagonist properties.
AID259398Selectivity for human kappa opioid receptor over delta opioid receptor2006Journal of medicinal chemistry, Jan-12, Volume: 49, Issue:1
Synthesis and preliminary in vitro investigation of bivalent ligands containing homo- and heterodimeric pharmacophores at mu, delta, and kappa opioid receptors.
AID14593350% Inhibition of stereospecific [3H]-naltrexone (10e-9 M) binding towards opiate receptor in rat brain homogenate1985Journal of medicinal chemistry, Sep, Volume: 28, Issue:9
Acylmorphinans. A novel class of potent analgesic agents.
AID184524Effect was expressed as mortality after injection of kainic acid (10 mg/kg) and the drug at the dose of 20 mg/Kg after 48 hr out of 19 animals2001Bioorganic & medicinal chemistry letters, Jul-09, Volume: 11, Issue:13
Anticonvulsant effects of new morphinan derivatives.
AID229399Relative affinity for mu and kappa opioid receptors2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
2-aminothiazole-derived opioids. Bioisosteric replacement of phenols.
AID178287Anticonvulsant activity in rat supramaximal electroshock (MES) test within a dose range of 15-60 mg/kg.1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID149172Binding affinity for mouse opioid receptor mu1989Journal of medicinal chemistry, Sep, Volume: 32, Issue:9
Benzofuro[2,3-c]pyridin-6-ols: synthesis, affinity for opioid-receptor subtypes, and antinociceptive activity.
AID670111Displacement of [3H]-U69,593 from human kappa opioid receptor expressed in CHO cells after 60 mins by scintillation counting2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Synthesis, binding affinity, and functional in vitro activity of 3-benzylaminomorphinan and 3-benzylaminomorphine ligands at opioid receptors.
AID362200Selectivity ratio of Ki for human mu opioid receptor to Ki for human delta opioid receptor2008Bioorganic & medicinal chemistry letters, Aug-15, Volume: 18, Issue:16
Synthesis and pharmacological evaluation of hydrophobic esters and ethers of butorphanol at opioid receptors.
AID1135664Displacement of [3H]naloxone from opioid receptor (unknown origin) in brain homogenate at 1 uM to 10 nM1978Journal of medicinal chemistry, Jul, Volume: 21, Issue:7
6beta-[N,N-Bis(2-chloroethyl)amino]-17-(cyclopropylmethyl)-4,5alpha-epoxy-3,14-dihydroxymorphinan(chlornaltrexamine) a potent opioid receptor alkylating agent with ultralong narcotic antagonist actitivty.
AID259397Selectivity for human kappa opioid receptor over mu opioid receptor2006Journal of medicinal chemistry, Jan-12, Volume: 49, Issue:1
Synthesis and preliminary in vitro investigation of bivalent ligands containing homo- and heterodimeric pharmacophores at mu, delta, and kappa opioid receptors.
AID149216IC50 ratio measured as inhibitory potencies in mouse vas deferens and guinea pig ileum assays1982Journal of medicinal chemistry, Dec, Volume: 25, Issue:12
Synthesis and pharmacological characterization in vitro of cyclic enkephalin analogues: effect of conformational constraints on opiate receptor selectivity.
AID148090In vitro binding affinity against cloned human Opioid receptor delta 1 expressed in HEK 293S cells2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
The power of visual imagery in drug design. Isopavines as a new class of morphinomimetics and their human opioid receptor binding activity.
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.
AID147944Inhibition of [3H]naltrindole binding to opioid receptor delta 1 of Chinese hamster ovary membrane2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
2-aminothiazole-derived opioids. Bioisosteric replacement of phenols.
AID148328Inhibition of [3H]-DAMGO binding to Opioid receptor mu 1 of Chinese hamster ovary membrane2004Journal of medicinal chemistry, Apr-08, Volume: 47, Issue:8
2-aminothiazole-derived opioids. Bioisosteric replacement of phenols.
AID514203Inhibition of electric eel AChE by Ellman's method2010Bioorganic & medicinal chemistry, Jul-15, Volume: 18, Issue:14
Morphinans and isoquinolines: acetylcholinesterase inhibition, pharmacophore modeling, and interaction with opioid receptors.
AID151584Binding constant for the agonist state was measured for its ability to displace [3H]naloxone from opioid mu 1 receptor buffered homogenate of rat brain membranes1988Journal of medicinal chemistry, Mar, Volume: 31, Issue:3
Structure-activity relationships for drugs binding to the agonist and antagonist states of the primary morphine receptor.
AID147903Binding affinity against opioid receptor mu 1 using [3H]DAMGO as radioligand in guinea pig brain membranes.2004Journal of medicinal chemistry, Jan-01, Volume: 47, Issue:1
10-Ketomorphinan and 3-substituted-3-desoxymorphinan analogues as mixed kappa and micro opioid ligands: synthesis and biological evaluation of their binding affinity at opioid receptors.
AID233830Selective inhibitory activity was determined against kappa & delta opioid receptor in chinese Hamster Ovary (CHO) cell membranes2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of novel dimeric morphinan ligands for kappa and micro opioid receptors.
AID128505Analgesic activity was determined by tail flick assay after subcutaneous administration in mice1982Journal of medicinal chemistry, Oct, Volume: 25, Issue:10
Synthesis and pharmacology of metabolically stable tert-butyl ethers of morphine and levorphanol.
AID670110Displacement of [3H]-DAMGO from human mu opioid receptor expressed in CHO cells after 60 mins by scintillation counting2012Journal of medicinal chemistry, Apr-26, Volume: 55, Issue:8
Synthesis, binding affinity, and functional in vitro activity of 3-benzylaminomorphinan and 3-benzylaminomorphine ligands at opioid receptors.
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.
AID143911Binding affinity against dextromethorpin binding site associated with N-methyl-D-aspartate glutamate receptor from guinea pig mitochondriall pellet P2 determined using [3H]1 as radioligand.1992Journal of medicinal chemistry, Oct-30, Volume: 35, Issue:22
Synthesis and evaluation of 3-substituted 17-methylmorphinan analogs as potential anticonvulsant agents.
AID259396Displacement of [3H]U69593 from human kappa opioid receptor expressed in CHO cells2006Journal of medicinal chemistry, Jan-12, Volume: 49, Issue:1
Synthesis and preliminary in vitro investigation of bivalent ligands containing homo- and heterodimeric pharmacophores at mu, delta, and kappa opioid receptors.
AID151006Selectivity of mu IC50 to that of delta IC50 was calculated2003Journal of medicinal chemistry, Jan-02, Volume: 46, Issue:1
The power of visual imagery in drug design. Isopavines as a new class of morphinomimetics and their human opioid receptor binding activity.
AID624616Specific activity of expressed human recombinant UGT2B152000Annual review of pharmacology and toxicology, , Volume: 40Human UDP-glucuronosyltransferases: metabolism, expression, and disease.
AID1145611Dissociation constant, pKa of the compound by titrimetric analysis1977Journal of medicinal chemistry, Jan, Volume: 20, Issue:1
Use of distribution coefficients in quantitative structure-activity relationships.
AID1802708Intracellular Calcium Mobilization Assay from Article 10.1038/nchembio.2334: \\In silico design of novel probes for the atypical opioid receptor MRGPRX2.\\2017Nature chemical biology, 05, Volume: 13, Issue:5
In silico design of novel probes for the atypical opioid receptor MRGPRX2.
AID1802709PRESTO-Tango Assay from Article 10.1038/nchembio.2334: \\In silico design of novel probes for the atypical opioid receptor MRGPRX2.\\2017Nature chemical biology, 05, Volume: 13, Issue:5
In silico design of novel probes for the atypical opioid receptor MRGPRX2.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (617)

TimeframeStudies, This Drug (%)All Drugs %
pre-1990498 (80.71)18.7374
1990's62 (10.05)18.2507
2000's37 (6.00)29.6817
2010's14 (2.27)24.3611
2020's6 (0.97)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 55.48

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 Index55.48 (24.57)
Research Supply Index6.49 (2.92)
Research Growth Index4.21 (4.65)
Search Engine Demand Index93.94 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (55.48)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials26 (4.10%)5.53%
Reviews20 (3.15%)6.00%
Case Studies12 (1.89%)4.05%
Observational0 (0.00%)0.25%
Other576 (90.85%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (2)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Evolution of Analgesic Tolerance During Long Term Treatment of Chronic Pain With Opioids [NCT00275249]60 participants Interventional2001-03-31Completed
Levorphanol as a Second Line Opioid in Cancer Patients Undergoing Opioid Rotation: An Open Label Study [NCT03579446]Early Phase 143 participants (Actual)Interventional2018-11-29Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]