Page last updated: 2024-12-10

silodosin

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

Description

silodosin: an alpha(1a)-adrenoceptor-selective antagonist; structure given in first source [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

Cross-References

ID SourceID
PubMed CID5312125
CHEMBL ID24778
CHEBI ID135929
SCHEMBL ID136973
MeSH IDM0505454

Synonyms (86)

Synonym
HY-10122
gtpl493
rapaflo
urorec
kad-3213
silodosin
silodyx
kso-0400
kmd-3213
urief
kmd3213
160970-54-7
silodosin (jp17/inn)
rapaflo (tn)
D01965
kmd 3213
urief (tn)
CHEBI:135929
silodal
CHEMBL24778 ,
rapilif
bdbm50160154
1-(3-hydroxy-propyl)-5-((r)-2-{2-[2-(2,2,2-trifluoro-ethoxy)-phenoxy]-ethylamino}-propyl)-2,3-dihydro-1h-indole-7-carboxylic acid amide
AKOS005145899
1-(3-hydroxypropyl)-5-(2-(2-(2-(2,2,2-trifluoroethoxy)phenoxy)ethylamino)propyl)indoline-7-carboxamide
1-(3-hydroxypropyl)-5-[(2r)-2-[2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethylamino]propyl]-2,3-dihydroindole-7-carboxamide
1-(3-hydroxypropyl)-5-[(2r)-2-[[2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl]amino]propyl]-2,3-dihydro-1h-indole-7-carboxamide; 1-(3-hydroxypropyl)-5-[(2r)-2-({2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl}amino)propyl]-2,3-dihydro-1h-indole-7-carboxamide; (-)-1
A810210
kad 3213
rapflo
silodosin [inn:ban]
unii-cuz39luy82
(-)-1-(3-hydroxypropyl)-5-((2r)-2-((2-(2-(2,2,2-trifluoroethoxy)phenoxy)ethyl)amino)propyl)-2,3-dihydro-1h-indole-7-carboxamide
cuz39luy82 ,
2,3-dihydro-1-(3-hydroxypropyl)-5-[(2r)-2-[[2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl]amino]propyl]-1h-indole-7-carboxamide
silodosin [vandf]
silodosin [jan]
silodosin [mi]
silodosin [orange book]
silodosin [inn]
silodosin [who-dd]
2,3-dihydro-1-(3-hydroxypropyl)-5-((2r)-2-((2-(2-(2,2,2-trifluoroethoxy)phenoxy)ethyl)amino)propyl)-1h-indole-7-carboxamide
silodosin [ema epar]
silodosin [mart.]
CS-0284
S1613
DB06207
CCG-221202
smr004701206
MLS006010022
(-)-1-(3-hydroxypropyl)-5-[(2r)-2-[[2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl]amino]propyl]-2,3-di-hydro-1h-indole-7-carboxamide
SCHEMBL136973
AM20090780
1h-indole-7-carboxamide, 2,3-dihydro-1-(3-hydroxypropyl)-5-[(2r)-2-[[2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl]amino]propyl]-
BS-1011
(r)-5-[2-[[2-[2-(2,2,2-trifluoro-ethoxy)phenoxy]ethyl]amino]propyl]-1-(3-hydroxypropyl)-2,3-dihydro-1h-indole-7-carboxamide
1-(3-hydroxypropyl)-5-[(2r)-2-({2-[2-(2,2,2-trifluoroethoxy)phenoxy]-ethyl}amino)propyl]-2,3-dihydro-1h-indole-7-carboxamide
Q-102517
AC-22605
DTXSID40167045 ,
SR-01000944157-1
sr-01000944157
mfcd00930170
silodosin (rapaflo)
HMS3715B06
silodosin, >=98% (hplc)
1-(3-hydroxypropyl)-5-[(2r)-2-({2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl}amino)propyl]-2,3-dihydro-1h-indole-7-carboxamide
SW219765-1
kad 3213;kmd 3213
Q411770
( c)-1-(3-hydroxypropyl)-5-[(2r)-2-[[2-[2-(2,2,2-trifluoroethoxy)phenoxy]ethyl]amino]propyl]-2,3-di-hydro-1h-indole-7-carboxamide
(r)-1-(3-hydroxypropyl)-5-(2-(2-(2-(2,2,2-trifluoroethoxy)phenoxy)ethylamino)propyl)indoline-7-carboxamide
BCP02143
HMS3884O21
NCGC00345882-03
I1128
EN300-6481367
urief, rapaflo
Z2235802137
silodosinum
silodosine
dtxcid3089536
silodosin (mart.)
1-(3-hydroxypropyl)-5-((2r)-2-((2-(2-(2,2,2-trifluoroethoxy)phenoxy)ethyl)amino)propyl)-2,3-dihydro-1h-indole-7-carboxamide
silodosina
g04ca04

Research Excerpts

Overview

Silodosin is a highly selective alpha1A-adrenoceptor antagonist approved for the treatment of the signs and symptoms of benign prostatic hyperplasia. It is specific to the lower urinary tract and may have fewer side effects than other alpha-blockers.

ExcerptReferenceRelevance
"Silodosin (SLD) is an FDA approved α1A-adrenoceptor blocker."( Validated Chromatographic and Spectrofluorimetric Methods for Analysis of Silodosin: A Comparative Study with Application of RP-HPLC in the Kinetic Investigation of Silodosin Degradation.
Abdelkawy, M; Boltia, SA; Mohamed, TA; Mostafa, NN, 2020
)
1.51
"Silodosin is a novel, more selective alpha-blocker, which is specific to the lower urinary tract and may have fewer side effects than other alpha-blockers."( Silodosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia.
Dahm, P; Jung, JH; Kim, J; Kim, MH; MacDonald, R; Reddy, B, 2017
)
2.62
"Silodosin is a highly selective alpha1A-adrenoceptor antagonist approved for the treatment of the signs and symptoms of benign prostatic hyperplasia. "( [Profile of silodosin].
Montorsi, F,
)
1.95
"Silodosin is an effective treatment for male lower urinary tract symptoms suggestive of benign prostatic hyperplasia. "( A pooled analysis of individual patient data from registrational trials of silodosin in the treatment of non-neurogenic male lower urinary tract symptoms (LUTS) suggestive of benign prostatic hyperplasia (BPH).
Chapple, CR; Montorsi, F; Novara, G, 2014
)
2.08
"Silodosin is a highly selective α1A-adrenoceptor antagonist indicated for the treatment of the signs and symptoms of benign prostatic hyperplasia (BPH). "( Silodosin: a review of its use in the treatment of the signs and symptoms of benign prostatic hyperplasia.
Keating, GM, 2015
)
3.3
"Silodosin (SLD) is a novel α1-adrenoceptor antagonist which has shown promising clinical efficacy and safety in patients with benign prostatic hyperplasia (BPH). "( In vivo metabolic investigation of silodosin using UHPLC-QTOF-MS/MS and in silico toxicological screening of its metabolites.
Baikadi, S; Borkar, RM; Satheeshkumar, N; Srinivas, R; Vishnuvardhan, C, 2016
)
2.15
"Silodosin is a new alpha(1)-adrenergic receptor antagonist that is selective for the alpha(1A)-adrenergic receptor. "( Silodosin: a selective alpha1A-adrenergic receptor antagonist for the treatment of benign prostatic hyperplasia.
Benzeroual, KE; Schilit, S, 2009
)
3.24
"Silodosin is a new selective therapy with a high pharmacologic selectivity for the α(1A)-adrenoreceptor."( Silodosin therapy for lower urinary tract symptoms in men with suspected benign prostatic hyperplasia: results of an international, randomized, double-blind, placebo- and active-controlled clinical trial performed in Europe.
Chapple, CR; Fernández Fernández, E; Koldewijn, E; Montorsi, F; Tammela, TL; Wirth, M, 2011
)
3.25
"Silodosin is an effective and well-tolerated treatment for the relief of both voiding and storage symptoms in patients with lower urinary tract symptoms suggestive of bladder outlet obstruction thought to be associated with benign prostatic hyperplasia. "( Silodosin therapy for lower urinary tract symptoms in men with suspected benign prostatic hyperplasia: results of an international, randomized, double-blind, placebo- and active-controlled clinical trial performed in Europe.
Chapple, CR; Fernández Fernández, E; Koldewijn, E; Montorsi, F; Tammela, TL; Wirth, M, 2011
)
3.25
"Silodosin is an α-adrenoceptor antagonist with high selectivity for α(1A)- relative to α(1B)- adrenoceptors. "( Silodosin: treatment of the signs and symptoms of benign prostatic hyperplasia.
Curran, MP, 2011
)
3.25
"Silodosin is a new α(1)-adrenergic receptor antagonist that is selective for the α(1A)-adrenergic receptor."( Safety and efficacy of silodosin for the treatment of benign prostatic hyperplasia.
Homma, Y; Kawabe, K; Kudoh, J; Yoshida, M, 2011
)
1.4
"Silodosin is an α(1) -adrenoceptor antagonist developed by Kissei Pharmaceutical, and has a specific selectivity for the α(1A-) adrenoceptor subtype."( New clinical evidence of silodosin, an α(1A) selective adrenoceptor antagonist, in the treatment for lower urinary tract symptoms.
Homma, Y; Kawabe, K; Kudoh, J; Yoshida, M, 2012
)
1.4
"Silodosin is a new uroselective alpha-blocker with high pharmacological selectivity for the (1A) adrenoceptor. "( Silodosin : a new subtype selective alpha-1 antagonist for the treatment of lower urinary tract symptoms in patients with benign prostatic hyperplasia.
Chapple, CR; Cruz, F; Desgrandchamps, F; Llorente, C; Montorsi, F; Osman, NI, 2012
)
3.26
"Silodosin is a rapidly efficacious and safe agent in the treatment of LUTS/BPH in men. "( Silodosin : a new subtype selective alpha-1 antagonist for the treatment of lower urinary tract symptoms in patients with benign prostatic hyperplasia.
Chapple, CR; Cruz, F; Desgrandchamps, F; Llorente, C; Montorsi, F; Osman, NI, 2012
)
3.26
"Silodosin is an effective and well-tolerated treatment for both voiding and storage symptoms in patients with LUTS associated with BPH. "( A meta-analysis of efficacy and safety of the new α1A-adrenoceptor-selective antagonist silodosin for treating lower urinary tract symptoms associated with BPH.
Dong, Q; Liu, LR; Wei, Q; Wu, YJ, 2013
)
2.05
"Silodosin is a novel drug that is highly selective to subtype alpha 1A and, since 2006, has been used in Japan for treating benign prostatic hyperplasia. "( Silodosin versus naftopidil in Japanese patients with lower urinary tract symptoms associated with benign prostatic hyperplasia: a randomized multicenter study.
Fujisawa, M; Haraguchi, T; Minayoshi, K; Miyake, H; Miyazaki, J; Morishita, S; Shigemura, K; Shirakawa, T; Tanaka, K; Yamada, Y, 2013
)
3.28
"Silodosin is a new selective therapy with a high pharmacologic selectivity for the a (1A)-adrenoreceptor."( [Silodosin therapy for lower urinary tract symptoms in men with suspected benign prostatic hyperplasia: results of an international, randomized, double-blind, placebo- and active-controlled clinical trial performed in Europe].
Chapple, ChR; Fernandez Fernandez, E; Koldewijn, E; Montorsi, F; Tammela, TL; Wirth, M,
)
2.48
"Silodosin is an effective and well-tolerated treatment for the relief of both voiding and storage symptoms in patients with lower urinary tract symptoms suggestive of bladder outlet obstruction thought to be associated with benign prostatic hyperplasia. "( [Silodosin therapy for lower urinary tract symptoms in men with suspected benign prostatic hyperplasia: results of an international, randomized, double-blind, placebo- and active-controlled clinical trial performed in Europe].
Chapple, ChR; Fernandez Fernandez, E; Koldewijn, E; Montorsi, F; Tammela, TL; Wirth, M,
)
2.48
"Silodosin was found to be a dual substrate for CYP3A4 and p-glycoprotein."( [Pharmacokinetics and disposition of silodosin (KMD-3213)].
Abe, Y; Adachi, Y; Harada, H; Kanazawa, T; Kanbe, H; Kobayashi, K; Kojima, Y; Matsubara, Y; Midgley, I; Momose, Y; Terakado, S, 2006
)
1.33
"Silodosin is a novel selective alpha(1A)-adrenoceptor (AR) antagonist generated by Kissei Pharmaceutical Co. "( Silodosin, a novel selective alpha 1A-adrenoceptor selective antagonist for the treatment of benign prostatic hyperplasia.
Homma, Y; Kawabe, K; Yoshida, M, 2007
)
3.23

Effects

Silodosin has a good cardiovascular safety profile and can be considered an option in patients with cardiovascular co-morbidities.

Silodosin has a good cardiovascular safety profile and can be considered an option in patients with cardiovascular co-morbidities. It can be used in all patients suffering from BPH and having indications to the administration of alpha-blockers.

ExcerptReferenceRelevance
"Silodosin has a good cardiovascular safety profile and can be considered an option in patients with cardiovascular co-morbidities."( Silodosin in the management of lower urinary tract symptoms as a result of benign prostatic hyperplasia: who are the best candidates.
Briganti, A; Capitanio, U; Montorsi, F; Salonia, A, 2013
)
2.55
"Silodosin has a greater impact on improving PI-induced lower urinary tract symptoms than the other two agents."( Comparison of prophylactic naftopidil, tamsulosin, and silodosin for 125I brachytherapy-induced lower urinary tract symptoms in patients with prostate cancer: randomized controlled trial.
Baba, S; Fujita, T; Hayakawa, K; Ishiyama, H; Kimura, M; Kitano, M; Kotani, S; Matsumoto, K; Minamida, S; Satoh, T; Tabata, K; Tsumura, H, 2011
)
2.06
"Silodosin has been demonstrated as a predisposing factor, strongly correlated with IFIS development. "( Silodosin as a predisposing factor of intraoperative floppy iris syndrome (IFIS): an observational propensity score-matching cohort study.
Christou, CD; Kourouklidou, M; Mataftsi, A; Oustoglou, E; Tzamalis, A; Ziakas, N, 2022
)
3.61
"Silodosin has higher apoptosis effect than other alpha-blockers in prostate. "( Evaluation of apoptosis indexes in currently used oral alpha-blockers in prostate: a pilot study.
Akin, Y; Buyukfirat, E; Ciftci, H; Demir, M; Gulum, M; Terim, KAK; Yeni, E,
)
1.57
"Silodosin has a good cardiovascular safety profile and can be considered an option in patients with cardiovascular co-morbidities."( Silodosin in the management of lower urinary tract symptoms as a result of benign prostatic hyperplasia: who are the best candidates.
Briganti, A; Capitanio, U; Montorsi, F; Salonia, A, 2013
)
2.55
"Silodosin has rapid onset of effect (3-4 days after the start of treatment); it can be used in all patients suffering from BPH and having indications to the administration of alpha-blockers, and does not require correction of doses of antihypertensive drugs in the case of combined appointment."( [Experience of the use of drug urorec: efficacy and safety].
Derevianko, TI; Kadiev, RM,
)
0.85
"Silodosin has greater affinity for the alpha(1A)-adrenergic receptor than for the alpha(1B)-adrenergic receptor (by a factor of 583), minimizing the propensity for blood pressure-related adverse effects mediated by alpha(1B) blockade."( Silodosin: a selective alpha1A-adrenergic receptor antagonist for the treatment of benign prostatic hyperplasia.
Benzeroual, KE; Schilit, S, 2009
)
2.52
"Silodosin has a greater impact on improving PI-induced lower urinary tract symptoms than the other two agents."( Comparison of prophylactic naftopidil, tamsulosin, and silodosin for 125I brachytherapy-induced lower urinary tract symptoms in patients with prostate cancer: randomized controlled trial.
Baba, S; Fujita, T; Hayakawa, K; Ishiyama, H; Kimura, M; Kitano, M; Kotani, S; Matsumoto, K; Minamida, S; Satoh, T; Tabata, K; Tsumura, H, 2011
)
2.06

Actions

Silodosin improved lower urinary tract symptoms by improving bladder storage function and relieving benign prostatic obstruction. It can inhibit the progression of prostatic hyperplasia through a recovery of prostatics blood flow.

ExcerptReferenceRelevance
"Silodosin can inhibit the progression of prostatic hyperplasia through a recovery of prostatic blood flow."( Effect of Silodosin, an Alpha1A-Adrenoceptor Antagonist, on Ventral Prostatic Hyperplasia in the Spontaneously Hypertensive Rat.
Higashi, Y; Honda, M; Inoue, K; Martin, DT; Nakamura, K; Saito, M; Shimizu, S; Shimizu, T; Tsounapi, P, 2015
)
2.26
"Silodosin improved lower urinary tract symptoms in three out of four patients, including diurnal voiding and storage symptoms, nocturia, and quality of life. "( Effectiveness and safety of silodosin in the treatment of lower urinary tract symptoms in patients with benign prostatic hyperplasia: A European phase IV clinical study (SiRE study).
Chapple, C; Cruz, F; Desgrandchamps, F; Gandaglia, G; Llorente, C; Montorsi, F, 2016
)
2.17
"Silodosin improved lower urinary tract symptoms by improving bladder storage function and relieving benign prostatic obstruction."( Efficacy of silodosin for relieving benign prostatic obstruction: prospective pressure flow study.
Funahashi, Y; Gotoh, M; Hattori, R; Komatsu, T; Matsukawa, Y; Sassa, N, 2009
)
2.17
"Silodosin improved lower urinary tract symptoms by improving bladder storage function and relieving benign prostatic obstruction."( Efficacy of silodosin for relieving benign prostatic obstruction: prospective pressure flow study.
Funahashi, Y; Gotoh, M; Hattori, R; Komatsu, T; Matsukawa, Y; Sassa, N, 2013
)
2.21

Treatment

Treatment with silodosin and the higher dose of tadalafil improved the urodynamic parameters, bladder blood flow and bladder tissue levels of malondialdehyde in the spontaneously hypertensive rats without affecting the blood pressure.

ExcerptReferenceRelevance
"Silodosin treatment produced a significantly higher improvement rate compared with naftopidil (P = 0.0002)."( Silodosin versus naftopidil in the treatment of premature ejaculation: A prospective multicenter trial.
Amano, T; Araki, T; Haga, K; Honma, I; Kondou, N; Matsukawa, M; Otani, T; Sato, Y; Tobe, M; Uchida, K, 2017
)
2.62
"The silodosin-treated group showed a smaller number of NVCs in CMG measurements and lower SAAs of both Aδ- and C-fibers than the vehicle-treated group during bladder-filling. "( Inhibitory effects of silodosin on the bladder mechanosensitive afferent activities and their relation with bladder myogenic contractions in male rats with bladder outlet obstruction.
Aizawa, N; Fujimura, T; Fukuhara, H; Homma, Y; Igawa, Y; Kume, H; Watanabe, D, 2018
)
1.35
"Silodosin treatment reduced the expression/activity of ELK1 in these cells as well as the viability of AR-positive LNCaP and C4-2 cells and the migration of both AR-positive and AR-negative cells, but not the viability of AR-negative or ELK1-negative cells."( Silodosin inhibits prostate cancer cell growth via ELK1 inactivation and enhances the cytotoxic activity of gemcitabine.
Aljarah, AK; Han, B; Ide, H; Inoue, S; Kashiwagi, E; Kawahara, T; Li, Y; Miyamoto, H; Patterson, JD; Shareef, HK; Zheng, Y, 2016
)
2.6
"With silodosin treatment, LUTS could be managed effectively for more than a year in at least 44% of the patients."( Urodynamic effects of silodosin, a new alpha 1A-adrenoceptor selective antagonist, for the treatment of benign prostatic hyperplasia.
Kamai, T; Mizuno, T; Tatsumiya, K; Watanabe, M; Yamanishi, T; Yoshida, K, 2010
)
1.13
"Silodosin-treated patients were stratified by absence or presence of 'retrograde ejaculation' (RE)."( Symptomatic and urodynamic responses in patients with reduced or no seminal emission during silodosin treatment for LUTS and BPH.
Kaplan, SA; Lepor, H; Roehrborn, CG; Volinn, W, 2011
)
1.31
"Treatment with silodosin and the higher dose of tadalafil improved the urodynamic parameters, bladder blood flow and bladder tissue levels of malondialdehyde in the spontaneously hypertensive rats without affecting the blood pressure and bladder tissue levels of interleukin-6 and tumor necrosis factor-alpha."( Effects of silodosin and tadalafil on bladder dysfunction in spontaneously hypertensive rats: Possible role of bladder blood flow.
Fujieda, M; Higashi, Y; Kurabayashi, A; Nagao, Y; Saito, M; Shimizu, S; Shimizu, T; Tsuda, M, 2020
)
1.29
"Treatment with silodosin or tadalafil might improve hypertension-related bladder overactivity, as shown in spontaneously hypertensive rats through an improvement in the bladder blood flow and bladder tissue levels of oxidative stress."( Effects of silodosin and tadalafil on bladder dysfunction in spontaneously hypertensive rats: Possible role of bladder blood flow.
Fujieda, M; Higashi, Y; Kurabayashi, A; Nagao, Y; Saito, M; Shimizu, S; Shimizu, T; Tsuda, M, 2020
)
1.3
"Treatment with silodosin proved to be safe and effective, as demonstrated by the increased stone expulsion rate, the reduced expulsion time, and the reduced analgesics consumption. ."( Efficacy of Silodosin in Expulsive Therapy for Distal Ureteral Stones: A Randomized Double-blinded Controlled Trial.
Chang, CH; Tsai, PC; Wang, CJ, 2016
)
1.17
"Treatment with silodosin produced rapid improvement in urinary symptoms that was sustained for 12 weeks. "( Rapid efficacy of the highly selective alpha1A-adrenoceptor antagonist silodosin in men with signs and symptoms of benign prostatic hyperplasia: pooled results of 2 phase 3 studies.
Gittelman, MC; Hill, LA; Hoel, G; Marks, LS; Volinn, W, 2009
)
0.94
"Treatment with silodosin normalized the decreased BBF, and treatment with naftopidil increased the BBF in a dose-dependent manner in the SHR group."( Characterization of silodosin and naftopidil in the treatment of bladder dysfunction in the spontaneously hypertensive rat.
Dimitriadis, F; Kinoshita, Y; Ohmasa, F; Oikawa, R; Saito, M; Satoh, K; Shimizu, S; Tsounapi, P, 2013
)
1.05
"Treatment with silodosin produced rapid improvement in urinary symptoms that was sustained for 12 weeks. "( Rapid efficacy of the highly selective α(1A)-adrenoceptor antagonist silodosin in men with signs and symptoms of benign prostatic hyperplasia: pooled results of 2 phase 3 studies.
Gittelman, MC; Hill, LA; Hoel, G; Marks, LS; Volinn, W, 2013
)
0.98

Toxicity

Silodosin was found to be safe and effective in the treatment of nocturia in patients with BPH. It was associated with significantly higher adverse event (AE) rates, compared with placebo, in all patient subgroups.

ExcerptReferenceRelevance
" In conclusion, silodosin exhibited adequate safety margins between the clinically recommended dose and those at which toxic effects or safety pharmacological changes were detected."( [Toxicity profile of silodosin (KMD-3213)].
Hayashi, M; Kasahara, H; Kobayashi, K; Kuroda, J; Murakami, M; Muto, S; Shibata, N; Souma, S; Tamura, T; Yokoi, R, 2006
)
1
"We retrospectively evaluated patients who received silodosin for lower urinary tract symptoms suggestive of benign prostatic hyperplasia (BPH/LUTS) in our hospital, to investigate its efficacy, adverse events and continuance rate."( [Investigation of adverse events and the continuance rate of silodosin in all patients who received silodosin for lower urinary tract symptoms suggestive of benign prostatic hyperplasia].
Furuya, R; Furuya, S; Hisasue, S; Kobayashi, K; Masumori, N; Mutoh, M; Ogura, H; Tsukamoto, T, 2010
)
0.85
" Adverse events were observed in 56 of the 195 cases (28."( [Investigation of adverse events and the continuance rate of silodosin in all patients who received silodosin for lower urinary tract symptoms suggestive of benign prostatic hyperplasia].
Furuya, R; Furuya, S; Hisasue, S; Kobayashi, K; Masumori, N; Mutoh, M; Ogura, H; Tsukamoto, T, 2010
)
0.6
" Since silodosin has greater affinity for the α(1A)-adrenergic receptor than for the α(1B)-adrenergic receptor, it minimizes the propensity for blood pressure-related adverse effects caused by α(1B)-adrenergic receptor blockade."( Safety and efficacy of silodosin for the treatment of benign prostatic hyperplasia.
Homma, Y; Kawabe, K; Kudoh, J; Yoshida, M, 2011
)
1.14
" The safety observations showed that mild adverse events, including postural hypotension, dizziness, and headache, were observed."( Safety and pharmacokinetic studies of silodosin, a new α1A-adrenoceptor selective antagonist, in healthy Chinese male subjects.
Cui, YM; Liu, YW; Meng, L; Sun, PH; Zhao, X; Zhou, Y, 2011
)
0.64
"This meta-analysis proves silodosin to be an effective and safe treatment for the relief of lower urinary tract symptom associated with BPH."( The efficacy and safety of silodosin in treating BPH: a systematic review and meta-analysis.
Cui, Y; Zhang, Y; Zong, H, 2012
)
0.98
" Primary outcome was International Prostate Symptom Score (IPSS) and IPSS subsores; secondary outcomes were peak urinary flow rate (Q(max)), quality of life (QoL) and primary adverse events (AEs) included retrograde ejaculation, dizziness and headache."( A meta-analysis of efficacy and safety of the new α1A-adrenoceptor-selective antagonist silodosin for treating lower urinary tract symptoms associated with BPH.
Dong, Q; Liu, LR; Wei, Q; Wu, YJ, 2013
)
0.61
" There were no adverse effects related to the use of silodosin."( Prospective randomized placebo-controlled study to assess the safety and efficacy of silodosin in the management of acute urinary retention.
Ganesamoni, R; Kumar, S; Singh, SK; Tiwari, DP, 2013
)
0.86
" Adverse drug reactions, vital signs, and laboratory tests were recorded."( Safety and efficacy of 8-mg once-daily vs 4-mg twice-daily silodosin in patients with lower urinary tract symptoms suggestive of benign prostatic hyperplasia (SILVER Study): a 12-week, double-blind, randomized, parallel, multicenter study.
Choo, MS; Kim, DK; Kim, JC; Kim, JH; Kim, KD; Kim, SW; Kwon, DD; Lee, JG; Lee, JZ; Lee, KS; Na, YG; Paick, JS; Park, WH; Song, M; Yang, SK, 2014
)
0.65
" The 2 groups had similar frequencies of adverse drug reactions."( Safety and efficacy of 8-mg once-daily vs 4-mg twice-daily silodosin in patients with lower urinary tract symptoms suggestive of benign prostatic hyperplasia (SILVER Study): a 12-week, double-blind, randomized, parallel, multicenter study.
Choo, MS; Kim, DK; Kim, JC; Kim, JH; Kim, KD; Kim, SW; Kwon, DD; Lee, JG; Lee, JZ; Lee, KS; Na, YG; Paick, JS; Park, WH; Song, M; Yang, SK, 2014
)
0.65
" The 2 groups had similar adverse drug reaction profiles."( Safety and efficacy of 8-mg once-daily vs 4-mg twice-daily silodosin in patients with lower urinary tract symptoms suggestive of benign prostatic hyperplasia (SILVER Study): a 12-week, double-blind, randomized, parallel, multicenter study.
Choo, MS; Kim, DK; Kim, JC; Kim, JH; Kim, KD; Kim, SW; Kwon, DD; Lee, JG; Lee, JZ; Lee, KS; Na, YG; Paick, JS; Park, WH; Song, M; Yang, SK, 2014
)
0.65
" Silodosin was associated with significantly higher adverse event (AE) rates, compared with placebo, in all patient subgroups, with retrograde ejaculation being the most common."( Individual patient data from registrational trials of silodosin in the treatment of non-neurogenic male lower urinary tract symptoms (LUTS) associated with benign prostatic hyperplasia (BPH): subgroup analyses of efficacy and safety data.
Chapple, CR; Montorsi, F; Novara, G, 2015
)
1.58
" The main adverse event was ejaculation disorder."( A 12-Week, Open Label, Multi-Center Study to Evaluate the Clinical Efficacy and Safety of Silodosin on Voiding Dysfunction in Patients with Neurogenic Bladder.
Jung, HC; Kim, DY; Kim, JS; Moon, KH; Oh, TH; Park, CH, 2015
)
0.64
"This study indicates that silodosin was significantly effective, well tolerated and safe in patients who have voiding dysfunction associated with neurogenic bladder."( A 12-Week, Open Label, Multi-Center Study to Evaluate the Clinical Efficacy and Safety of Silodosin on Voiding Dysfunction in Patients with Neurogenic Bladder.
Jung, HC; Kim, DY; Kim, JS; Moon, KH; Oh, TH; Park, CH, 2015
)
0.94
" Adverse events occurred more frequently with silodosin than with tamsulosin; however, none of the adverse events required treatment discontinuation."( Randomized Crossover Comparison of the Short-Term Efficacy and Safety of Single Half-Dose Silodosin and Tamsulosin Hydrochoride in Men With Lower Urinary Tract Symptoms Secondary to Benign Prostatic Hyperplasia.
Arai, Y; Chiba, K; Horiuchi, S; Moriyama, S; Noro, A; Saito, K; Takeshita, H; Washino, S, 2016
)
0.91
" Treatment-emergent adverse events were recorded."( Effectiveness and safety of silodosin in the treatment of lower urinary tract symptoms in patients with benign prostatic hyperplasia: A European phase IV clinical study (SiRE study).
Chapple, C; Cruz, F; Desgrandchamps, F; Gandaglia, G; Llorente, C; Montorsi, F, 2016
)
0.73
" The most common treatment-emergent adverse event was ejaculation failure (185 patients; 17."( Effectiveness and safety of silodosin in the treatment of lower urinary tract symptoms in patients with benign prostatic hyperplasia: A European phase IV clinical study (SiRE study).
Chapple, C; Cruz, F; Desgrandchamps, F; Gandaglia, G; Llorente, C; Montorsi, F, 2016
)
0.73
" In conclusion, a combination of dutasteride and silodosin therapy may be effective and safe for patients with AUR due to BPH."( Efficacy and Safety of Silodosin and Dutasteride Combination Therapy in Acute Urinary Retention due to Benign Prostatic Hyperplasia: A Single-Arm Prospective Study.
Hagiwara, K; Hashimoto, Y; Hatakeyama, S; Imai, A; Iwamura, H; Koie, T; Ohyama, C; Yoneyama, T, 2016
)
1
"0006), few adverse effects were observed."( The efficacy and safety of silodosin for the treatment of ureteral stones: a systematic review and meta-analysis.
Cui, Y; Wu, J; Yang, D; Yuan, H, 2016
)
0.73
"This meta-analysis indicates silodosin is an effective and safe treatment option for ureteral stones with a low occurrence of side effects."( The efficacy and safety of silodosin for the treatment of ureteral stones: a systematic review and meta-analysis.
Cui, Y; Wu, J; Yang, D; Yuan, H, 2016
)
1.02
" Safety assessments included the rate of adverse events and adverse drug reactions."( Evaluating the Efficacy and Safety of Silodosin on Nocturia in Patients With Benign Prostatic Hyperplasia: A Multicenter, Prospective, Open-label, Single-arm, Phase IV Trial.
Cho, KJ; Choi, JB; Kim, DK; Kim, JC; Kim, YT; Lee, JZ; Song, YS, 2018
)
0.75
" The most common adverse drug reaction was an ejaculatory disorder (7."( Evaluating the Efficacy and Safety of Silodosin on Nocturia in Patients With Benign Prostatic Hyperplasia: A Multicenter, Prospective, Open-label, Single-arm, Phase IV Trial.
Cho, KJ; Choi, JB; Kim, DK; Kim, JC; Kim, YT; Lee, JZ; Song, YS, 2018
)
0.75
"Silodosin was found to be safe and effective in the treatment of nocturia in patients with BPH."( Evaluating the Efficacy and Safety of Silodosin on Nocturia in Patients With Benign Prostatic Hyperplasia: A Multicenter, Prospective, Open-label, Single-arm, Phase IV Trial.
Cho, KJ; Choi, JB; Kim, DK; Kim, JC; Kim, YT; Lee, JZ; Song, YS, 2018
)
2.19
" However, 1-adrenoreceptors are known to be present in other tissues and this fact could be a reason of potential risk of adverse events, associated with changes in peripheral vascular tone, such as orthostatic hypotension, syncopal states, dizziness, etc."( [Efficiency and safety of silodosin for treatment of lower urinary tract symptoms, associated of benign prostatic hyperplasia].
Krivoborodov, GG; Tur, EI, 2019
)
0.81

Pharmacokinetics

ExcerptReferenceRelevance
" The relationship between plasma silodosin concentration, its pharmacokinetic parameters, polymorphic alleles (UGT2B7, UGT1A8, MDR1, ALDH, ADH, CYP3A4, and CYP3A5), and other enzymes related to silodosin metabolism were assessed for each subject."( The influence of UGT2B7, UGT1A8, MDR1, ALDH, ADH, CYP3A4 and CYP3A5 genetic polymorphisms on the pharmacokinetics of silodosin in healthy Chinese volunteers.
Cui, Y; Wang, Z; Xiang, Q; Zhao, X; Zhou, Y, 2013
)
0.88

Bioavailability

ExcerptReferenceRelevance
" Absolute bioavailability values in rat, dog and human were about 9, 25 and 32%, respectively."( [Pharmacokinetics and disposition of silodosin (KMD-3213)].
Abe, Y; Adachi, Y; Harada, H; Kanazawa, T; Kanbe, H; Kobayashi, K; Kojima, Y; Matsubara, Y; Midgley, I; Momose, Y; Terakado, S, 2006
)
0.61

Dosage Studied

This article reviews the pharmacology, pharmacokinetics, clinical efficacy, adverse effects, drug interactions, and dosage and administration of silodosin in adult male patients with BPH. The purpose of this study was to evaluate the efficacy of the use of silodeosin (Urorec®) at dosage 8 mg once a day for relief of obstructive symptoms of BPH in men over 40 years old.

ExcerptRelevanceReference
" Extent and time course of the KMD-3213 effect reduction in the phenylephrine-induced increase in intraurethral pressure were closely associated with those in prostatic [(3)H]KMD-3213 binding after continuous infusion of the corresponding dosage of the radioligand."( Relationship between prostatic alpha(1)-adrenoceptor binding and reduction in intraurethral pressure following continuous infusion of KMD-3213 in rats.
Akiyama, K; Katayama, S; Kimura, R; Nakajima, M; Oki, T; Okura, T; Tatemichi, S; Yamada, S, 2002
)
0.31
" In conclusion, the prediction of alpha(1)-adrenoceptor occupancy in the human prostate by alpha(1)-adrenoceptor antagonists may provide the rationale for the optimum dosage regimen of these drugs in the therapy of BPH."( Prediction of alpha1-adrenoceptor occupancy in the human prostate from plasma concentrations of silodosin, tamsulosin and terazosin to treat urinary obstruction in benign prostatic hyperplasia.
Kagawa, Y; Kato, Y; Kawabe, K; Okura, T; Yamada, S, 2007
)
0.56
"This article reviews the pharmacology, pharmacokinetics, clinical efficacy, adverse effects, drug interactions, and dosage and administration of silodosin in adult male patients with BPH."( Silodosin: a selective alpha1A-adrenergic receptor antagonist for the treatment of benign prostatic hyperplasia.
Benzeroual, KE; Schilit, S, 2009
)
2
" Therapeutic doses of silodosin are safe for men with mild-to-moderate liver dysfunction; dosage adjustment is recommended in those with moderate renal impairment."( Silodosin for the treatment of benign prostatic hyperplasia: pharmacology and cardiovascular tolerability.
Hill, LA; Lepor, H, 2010
)
2.12
" It is concluded that silodosin is safe and tolerated in healthy Chinese male subjects at the dosing levels used in this study."( Safety and pharmacokinetic studies of silodosin, a new α1A-adrenoceptor selective antagonist, in healthy Chinese male subjects.
Cui, YM; Liu, YW; Meng, L; Sun, PH; Zhao, X; Zhou, Y, 2011
)
0.96
" Comparison of DR(10) values (the dose required to shift the dose-response curve 10-fold to the right) in both tissues showed that the inhibitory effect of silodosin was significantly more potent in the salivary gland than in the urethra (18-fold), but tamsulosin (2."( Effects of α1-adrenoceptor antagonists on phenylephrine-induced salivary secretion and intraurethral pressure elevation in anesthetized rats.
Hatanaka, T; Noguchi, Y; Ohtake, A; Sasamata, M; Sato, S; Suzuki, M; Ueshima, K; Yanai-Inamura, H, 2012
)
0.58
"To analyse data supporting the use of silodosin, a highly selective once-daily dosing α1-ARs blocker, in different daily clinical practice scenarios."( Silodosin in the management of lower urinary tract symptoms as a result of benign prostatic hyperplasia: who are the best candidates.
Briganti, A; Capitanio, U; Montorsi, F; Salonia, A, 2013
)
2.1
" We sought to assess the effects of a 3-month course of silodosin 8 mg daily dosing on sexual functioning, mainly including ejaculation and orgasm, in a cohort of 100 consecutive sexually active men in the real-life setting."( Effects of silodosin on sexual function - realistic picture from the everyday clinical practice.
Boeri, L; Briganti, A; Capogrosso, P; Damiano, R; Dehò, F; Montorsi, F; Salonia, A; Serino, A; Ventimiglia, E, 2015
)
1.05
"A novel, sensitive and selective spectrofluorimetric method has been developed and validated for determination of silodosine (SLD) in its dosage form and human plasma."( Selective Spectrofluorimetric Method with Enhanced Sensitivity for Determination of Silodosine in Dosage Form and Human Plasma. Application to Stability Studies and Content Uniformity Testing.
Derayea, SM; Hammad, MA; Mohamed, AA; Mohamed, AI; Omar, MA, 2017
)
0.89
" We also found that increasing value of total payment and increasing frequency of payments were both independently associated with increased odds of prescribing with a dose-response effect."( The Receipt of Industry Payments is Associated With Prescribing Promoted Alpha-blockers and Overactive Bladder Medications.
Chang, SL; Dupree, JM; Kirk, PS; Modi, PK; Singer, EA; Wang, Y, 2018
)
0.48
" The purpose of this study was to evaluate the efficacy of the use of silodosin (Urorec®) at dosage 8 mg once a day for relief of obstructive symptoms of BPH in men over 40 years old."( EFFICACY OF SILODOSIN (UROREC®) IN THE TREATMENT OF BENIGN PROSTATIC HYPERPLASIA: THE MAXIMUM DECREASE OF BLADDER OUTLET OBSTRUCTION INDEX.
Frolov, R; Khamzin, A, 2018
)
1.09
"Three analytical methods were developed for the determination of SLD based on RP-HPLC-PDAD, HPTLC, and 1DSFS in bulk and capsule dosage form."( Validated Chromatographic and Spectrofluorimetric Methods for Analysis of Silodosin: A Comparative Study with Application of RP-HPLC in the Kinetic Investigation of Silodosin Degradation.
Abdelkawy, M; Boltia, SA; Mohamed, TA; Mostafa, NN, 2020
)
0.79
" Eco-friendly and progressive spectrophotometric methods were firstly developed in this work, for the simultaneous determination of Dutasteride (DUT) and Silodosin (SLD) in their newly-marketed dosage form."( Fingerprinting and iso-absorptive resolution techniques for the spectrally overlapping Dutasteride and Silodosin mixture: Content uniformity testing along with greenness profile assessment.
Abdel-Moety, EM; Marzouk, HM; Rezk, MR; Rostom, Y; Wadie, M, 2022
)
1.13
" The work was also extended to verify content uniformity of dosage units in accordance with USP recommendations."( State-of-the-art mathematically induced filtration approaches for smart spectrophotometric assessment of silodosin and solifenacin mixture in their new challenging formulation: Multi-tool greenness and whiteness evaluation.
Abdel-Moety, EM; Marzouk, HM; Rezk, MR; Rostom, Y; Wadie, M, 2024
)
1.66
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Drug Classes (1)

ClassDescription
indolecarboxamide
[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]

Protein Targets (13)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Spike glycoproteinSevere acute respiratory syndrome-related coronavirusPotency39.81070.009610.525035.4813AID1479145
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Alpha-1A adrenergic receptorOryctolagus cuniculus (rabbit)Ki0.00000.00000.02040.2512AID36753
Potassium voltage-gated channel subfamily E member 1Homo sapiens (human)IC50 (µMol)251.18900.12004.048010.0000AID1207384
Alpha-1B adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)0.09010.00021.874210.0000AID1320520
Alpha-1D adrenergic receptorHomo sapiens (human)IC50 (µMol)0.01680.00020.75688.8970AID1301978; AID1320516
Alpha-1D adrenergic receptorHomo sapiens (human)Ki0.00200.00000.360910.0000AID35468
Alpha-1A adrenergic receptorHomo sapiens (human)IC50 (µMol)0.00190.00020.56145.4000AID1301976; AID1320512
Alpha-1A adrenergic receptorHomo sapiens (human)Ki0.00000.00000.272610.0000AID36753
Alpha-1B adrenergic receptorHomo sapiens (human)IC50 (µMol)0.32870.00020.65268.3300AID1301977; AID1320515
Alpha-1B adrenergic receptorHomo sapiens (human)Ki0.02000.00000.471310.0000AID37478
Alpha-1A adrenergic receptorRattus norvegicus (Norway rat)IC50 (µMol)0.00080.00001.819410.0000AID1320519
Potassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)IC50 (µMol)251.18900.12004.048010.0000AID1207384
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)13.75410.00091.901410.0000AID1207478; AID1207538; AID1320524
Voltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)IC50 (µMol)794.32800.00032.25459.6000AID1207572
Sodium channel protein type 5 subunit alphaHomo sapiens (human)IC50 (µMol)63.09570.00033.64849.2000AID1207324
Potassium voltage-gated channel subfamily D member 3Homo sapiens (human)IC50 (µMol)316.22801.40005.35009.3000AID1207444
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (145)

Processvia Protein(s)Taxonomy
epithelial cell maturationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
male gonad developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
vestibular nucleus developmentPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
secretory granule organizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to acidic pHPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cellular response to light stimulusPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell action potential involved in contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of protein targeting to membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
negative regulation of delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
G protein-coupled receptor signaling pathwayAlpha-1D adrenergic receptorHomo sapiens (human)
adenylate cyclase-modulating G protein-coupled receptor signaling pathwayAlpha-1D adrenergic receptorHomo sapiens (human)
positive regulation of cell population proliferationAlpha-1D adrenergic receptorHomo sapiens (human)
neuron-glial cell signalingAlpha-1D adrenergic receptorHomo sapiens (human)
cell-cell signalingAlpha-1D adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayAlpha-1D adrenergic receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayAlpha-1D adrenergic receptorHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationAlpha-1D adrenergic receptorHomo sapiens (human)
positive regulation of vasoconstrictionAlpha-1D adrenergic receptorHomo sapiens (human)
positive regulation of MAPK cascadeAlpha-1D adrenergic receptorHomo sapiens (human)
MAPK cascadeAlpha-1A adrenergic receptorHomo sapiens (human)
negative regulation of heart rate involved in baroreceptor response to increased systemic arterial blood pressureAlpha-1A adrenergic receptorHomo sapiens (human)
norepinephrine-epinephrine vasoconstriction involved in regulation of systemic arterial blood pressureAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of heart rate by epinephrine-norepinephrineAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of the force of heart contraction by epinephrine-norepinephrineAlpha-1A adrenergic receptorHomo sapiens (human)
apoptotic processAlpha-1A adrenergic receptorHomo sapiens (human)
smooth muscle contractionAlpha-1A adrenergic receptorHomo sapiens (human)
signal transductionAlpha-1A adrenergic receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayAlpha-1A adrenergic receptorHomo sapiens (human)
activation of phospholipase C activityAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationAlpha-1A adrenergic receptorHomo sapiens (human)
adult heart developmentAlpha-1A adrenergic receptorHomo sapiens (human)
negative regulation of cell population proliferationAlpha-1A adrenergic receptorHomo sapiens (human)
response to xenobiotic stimulusAlpha-1A adrenergic receptorHomo sapiens (human)
response to hormoneAlpha-1A adrenergic receptorHomo sapiens (human)
negative regulation of autophagyAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of cardiac muscle hypertrophyAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of synaptic transmission, GABAergicAlpha-1A adrenergic receptorHomo sapiens (human)
intracellular signal transductionAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of MAPK cascadeAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of action potentialAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of vasoconstrictionAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of smooth muscle contractionAlpha-1A adrenergic receptorHomo sapiens (human)
calcium ion transport into cytosolAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of cardiac muscle contractionAlpha-1A adrenergic receptorHomo sapiens (human)
cell growth involved in cardiac muscle cell developmentAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeAlpha-1A adrenergic receptorHomo sapiens (human)
positive regulation of protein kinase C signalingAlpha-1A adrenergic receptorHomo sapiens (human)
pilomotor reflexAlpha-1A adrenergic receptorHomo sapiens (human)
neuron-glial cell signalingAlpha-1A adrenergic receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayAlpha-1A adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayAlpha-1A adrenergic receptorHomo sapiens (human)
cell-cell signalingAlpha-1A adrenergic receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayAlpha-1B adrenergic receptorHomo sapiens (human)
adenylate cyclase-modulating G protein-coupled receptor signaling pathwayAlpha-1B adrenergic receptorHomo sapiens (human)
regulation of vasoconstrictionAlpha-1B adrenergic receptorHomo sapiens (human)
intracellular signal transductionAlpha-1B adrenergic receptorHomo sapiens (human)
positive regulation of MAPK cascadeAlpha-1B adrenergic receptorHomo sapiens (human)
regulation of cardiac muscle contractionAlpha-1B adrenergic receptorHomo sapiens (human)
neuron-glial cell signalingAlpha-1B adrenergic receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayAlpha-1B adrenergic receptorHomo sapiens (human)
cell-cell signalingAlpha-1B adrenergic receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayAlpha-1B adrenergic receptorHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationAlpha-1B adrenergic receptorHomo sapiens (human)
gastrin-induced gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
glucose metabolic processPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
heart developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
rhythmic behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of blood pressurePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of heart ratePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
iodide transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
erythrocyte differentiationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intracellular chloride ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
response to insulinPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
social behaviorPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
corticosterone secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear morphogenesisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
inner ear developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
intestinal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
detection of mechanical stimulus involved in sensory perception of soundPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
auditory receptor cell developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of cardiac muscle contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of gastric acid secretionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
stomach developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
renal sodium ion absorptionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to cAMPPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cellular response to epinephrine stimulusPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
adrenergic receptor signaling pathwayPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cardiac muscle cell contractionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cochlea developmentPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during atrial cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
non-motile cilium assemblyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
immune system developmentVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
positive regulation of cytosolic calcium ion concentrationVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
heart developmentVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of cardiac muscle contraction by regulation of the release of sequestered calcium ionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
embryonic forelimb morphogenesisVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
camera-type eye developmentVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
positive regulation of adenylate cyclase activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
positive regulation of muscle contractionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion transport into cytosolVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
cardiac conductionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion transmembrane transport via high voltage-gated calcium channelVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion transmembrane transportVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
cardiac muscle cell action potential involved in contractionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membrane depolarization during cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membrane depolarization during AV node cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
cell communication by electrical coupling involved in cardiac conductionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of heart rate by cardiac conductionVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of ventricular cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membrane depolarization during atrial cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calcium ion import across plasma membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
regulation of heart rateSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac conduction system developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac ventricle developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
brainstem developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of sodium ion transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
response to denervation involved in regulation of muscle adaptationSodium channel protein type 5 subunit alphaHomo sapiens (human)
telencephalon developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
cerebellum developmentSodium channel protein type 5 subunit alphaHomo sapiens (human)
sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
odontogenesis of dentin-containing toothSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
positive regulation of epithelial cell proliferationSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of atrial cardiac muscle cell membrane repolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of ventricular cardiac muscle cell membrane depolarizationSodium channel protein type 5 subunit alphaHomo sapiens (human)
cellular response to calcium ionSodium channel protein type 5 subunit alphaHomo sapiens (human)
cardiac muscle cell action potential involved in contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of cardiac muscle cell contractionSodium channel protein type 5 subunit alphaHomo sapiens (human)
ventricular cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during Purkinje myocyte cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
AV node cell to bundle of His cell communicationSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of heart rate by cardiac conductionSodium channel protein type 5 subunit alphaHomo sapiens (human)
membrane depolarization during atrial cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
regulation of sodium ion transmembrane transportSodium channel protein type 5 subunit alphaHomo sapiens (human)
potassium ion transportPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein homooligomerizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
action potentialPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (44)

Processvia Protein(s)Taxonomy
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
potassium channel regulator activityPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
telethonin bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein-containing complex bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
protein bindingAlpha-1D adrenergic receptorHomo sapiens (human)
identical protein bindingAlpha-1D adrenergic receptorHomo sapiens (human)
alpha1-adrenergic receptor activityAlpha-1D adrenergic receptorHomo sapiens (human)
alpha1-adrenergic receptor activityAlpha-1A adrenergic receptorHomo sapiens (human)
protein bindingAlpha-1A adrenergic receptorHomo sapiens (human)
protein heterodimerization activityAlpha-1A adrenergic receptorHomo sapiens (human)
protein bindingAlpha-1B adrenergic receptorHomo sapiens (human)
protein heterodimerization activityAlpha-1B adrenergic receptorHomo sapiens (human)
alpha1-adrenergic receptor activityAlpha-1B adrenergic receptorHomo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
calmodulin bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
phosphatidylinositol-4,5-bisphosphate bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein phosphatase 1 bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
outward rectifier potassium channel activityPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A catalytic subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
protein kinase A regulatory subunit bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in atrial cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
high voltage-gated calcium channel activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activity involved in cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
protein bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
calmodulin bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
high voltage-gated calcium channel activityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
metal ion bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
alpha-actinin bindingVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activity involved in cardiac muscle cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel activity involved in AV node cell action potentialVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated sodium channel activitySodium channel protein type 5 subunit alphaHomo sapiens (human)
protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
calmodulin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
fibroblast growth factor bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
enzyme bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein kinase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
protein domain specific bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ankyrin bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
ubiquitin protein ligase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
transmembrane transporter bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
nitric-oxide synthase bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in cardiac muscle cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in AV node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in bundle of His cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in Purkinje myocyte action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel activity involved in SA node cell action potentialSodium channel protein type 5 subunit alphaHomo sapiens (human)
scaffold protein bindingSodium channel protein type 5 subunit alphaHomo sapiens (human)
A-type (transient outward) potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
transmembrane transporter bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
metal ion bindingPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (46)

Processvia Protein(s)Taxonomy
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
Z discPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily E member 1Homo sapiens (human)
plasma membraneAlpha-1D adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-1D adrenergic receptorHomo sapiens (human)
nucleusAlpha-1A adrenergic receptorHomo sapiens (human)
nucleoplasmAlpha-1A adrenergic receptorHomo sapiens (human)
cytoplasmAlpha-1A adrenergic receptorHomo sapiens (human)
cytosolAlpha-1A adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-1A adrenergic receptorHomo sapiens (human)
caveolaAlpha-1A adrenergic receptorHomo sapiens (human)
nuclear membraneAlpha-1A adrenergic receptorHomo sapiens (human)
intracellular membrane-bounded organelleAlpha-1A adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-1A adrenergic receptorHomo sapiens (human)
nucleusAlpha-1B adrenergic receptorHomo sapiens (human)
cytoplasmAlpha-1B adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-1B adrenergic receptorHomo sapiens (human)
caveolaAlpha-1B adrenergic receptorHomo sapiens (human)
nuclear membraneAlpha-1B adrenergic receptorHomo sapiens (human)
plasma membraneAlpha-1B adrenergic receptorHomo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lysosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
early endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
late endosomePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
endoplasmic reticulumPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
apical plasma membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
transport vesiclePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
cytoplasmic vesicle membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuron projectionPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membrane raftPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
ciliary basePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
lumenal side of membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
basolateral part of cellPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
monoatomic ion channel complexPotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
membranePotassium voltage-gated channel subfamily KQT member 1Homo sapiens (human)
virion membraneSpike glycoproteinSevere acute respiratory syndrome-related coronavirus
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cytoplasmVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
plasma membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
postsynaptic densityVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
Z discVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
dendriteVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
perikaryonVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
postsynaptic density membraneVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
L-type voltage-gated calcium channel complexVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
voltage-gated calcium channel complexVoltage-dependent L-type calcium channel subunit alpha-1CHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
nucleolusSodium channel protein type 5 subunit alphaHomo sapiens (human)
endoplasmic reticulumSodium channel protein type 5 subunit alphaHomo sapiens (human)
plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
caveolaSodium channel protein type 5 subunit alphaHomo sapiens (human)
cell surfaceSodium channel protein type 5 subunit alphaHomo sapiens (human)
intercalated discSodium channel protein type 5 subunit alphaHomo sapiens (human)
membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
lateral plasma membraneSodium channel protein type 5 subunit alphaHomo sapiens (human)
Z discSodium channel protein type 5 subunit alphaHomo sapiens (human)
T-tubuleSodium channel protein type 5 subunit alphaHomo sapiens (human)
sarcolemmaSodium channel protein type 5 subunit alphaHomo sapiens (human)
perinuclear region of cytoplasmSodium channel protein type 5 subunit alphaHomo sapiens (human)
voltage-gated sodium channel complexSodium channel protein type 5 subunit alphaHomo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
sarcolemmaPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
GABA-ergic synapsePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic specialization membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
dendritic spinePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
neuronal cell bodyPotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
postsynaptic membranePotassium voltage-gated channel subfamily D member 3Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (43)

Assay IDTitleYearJournalArticle
AID1301976Antagonist activity at human alpha-1A adrenergic receptor transfected in HEK293 cells assessed as reduction in agonist-induced calcium mobilization after 10 mins2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1301997Plasma protein binding in rat at 2 uM after 16 hrs by UPLC-MS/MS analysis2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID36125Affinity estimate (pA2) for Alpha-1 adrenergic receptor antagonist determined on human prostate; ND=not determined1997Journal of medicinal chemistry, Apr-25, Volume: 40, Issue:9
Pharmacological options in the treatment of benign prostatic hyperplasia.
AID1320518Selectivity ratio of IC50 for human full-length N-terminal SNAP-tagged alpha1D adrenoceptor to IC50 for human full-length N-terminal SNAP-tagged alpha1A adrenoceptor2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320521Selectivity ratio of IC50 for alpha1B adrenoceptor in Sprague-Dawley rat aorta to IC50 for alpha1A adrenoceptor in Sprague-Dawley rat urethra2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1207324Inhibition of fast sodium current (INa) in Chinese Hamster Ovary (CHO) K1 cells transfected with human Nav1.5 measured using IonWorks Quattro automated patch clamp platform
AID239801Binding affinity for alpha 1d adrenoceptor2005Journal of medicinal chemistry, Jul-28, Volume: 48, Issue:15
Range and sensitivity as descriptors of molecular property spaces in dynamic QSAR analyses.
AID1301975Selectivity ratio of IC50 for human alpha-1B adrenergic receptor transfected in HEK293 cells to IC50 for human alpha-1A adrenergic receptor transfected in HEK293 cells2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID239202Binding constant measured against Alpha-1A adrenergic receptor in human prostate; +++:highly active2005Bioorganic & medicinal chemistry letters, Feb-01, Volume: 15, Issue:3
Pharmacophore identification of alpha(1A)-adrenoceptor antagonists.
AID1320522Inhibition of micturition frequency in po dosed Sprague-Dawley rat benign prostatic hyperplasia model administered 20 mins followed by distilled watering loading measured for 2 hrs2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1301980Inhibition of noradrenaline-induced contraction of Sprague-Dawley rat urethra smooth muscles after 20 mins2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1207478Inhibition of rapid delayed inward rectifying potassium current (IKr) in Chinese hamster ovary (CHO) K1 cells stably expressing hERG measured using IonWorks Quattro automated patch clamp platform
AID1320523Increase in voided volume in po dosed Sprague-Dawley rat benign prostatic hyperplasia model administered 20 mins followed by distilled watering loading measured for 2 hrs2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
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.
AID1207384Inhibition of slow delayed inward rectifying potassium current (Iks) in Chinese Hamster Ovary (CHO) cells expressing hKvLQT1/hminK measured using IonWorks Quattro automated patch clamp platform
AID239800Binding affinity for alpha 1b adrenoceptor2005Journal of medicinal chemistry, Jul-28, Volume: 48, Issue:15
Range and sensitivity as descriptors of molecular property spaces in dynamic QSAR analyses.
AID36753Binding affinity was tested on human Alpha-1A adrenergic receptor1997Journal of medicinal chemistry, Apr-25, Volume: 40, Issue:9
Pharmacological options in the treatment of benign prostatic hyperplasia.
AID1207538Inhibition of rapid delayed inward rectifying potassium current (IKr) measured using manual patch clamp assay
AID1320519Antagonist activity at alpha1A adrenoceptor in Sprague-Dawley rat urethra assessed as inhibition of norepinephrine-induced smooth muscle contraction after 20 mins2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID35468Binding affinity was tested on human Alpha-1D adrenergic receptor1997Journal of medicinal chemistry, Apr-25, Volume: 40, Issue:9
Pharmacological options in the treatment of benign prostatic hyperplasia.
AID1301981Inhibition of noradrenaline-induced contraction of Sprague-Dawley rat aorta after 20 mins2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1301983Induction of mean voided volume in po dosed Sprague-Dawley rat benign prostatic hyperplasia model administered for 20 mins measured after 2 hrs2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1207444Inhibition of transient outward potassium current (Ito) current in Chinese Hamster Ovary (CHO) K1 cells expressing human Kv4.3 measured using IonWorks Quattro automated patch clamp platform
AID1301984Reduction of micturition frequency in po dosed Sprague-Dawley rat benign prostatic hyperplasia model administered for 20 mins measured after 2 hrs2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1207572Inhibition of long-lasting type calcium current (hICa) in Chinese Hamster Ovary (CHO) cells expressing hCav1.2 measured using IonWorks Quattro automated patch clamp platform
AID1320513Antagonist activity at human full-length N-terminal SNAP-tagged alpha1A adrenoceptor expressed in HEK293 cells assessed as inhibition of agonist-induced calcium mobilization at 10 uM preincubated for 10 mins followed by agonist addition by fluo-4 AM dye-b2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320524Inhibition of human ERG2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320543Toxicity in patient assessed as orthostatic hypotension2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320512Antagonist activity at human full-length N-terminal SNAP-tagged alpha1A adrenoceptor expressed in HEK293 cells assessed as inhibition of agonist-induced calcium mobilization preincubated for 10 mins followed by agonist addition by fluo-4 AM dye-based assa2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320516Antagonist activity at human full-length N-terminal SNAP-tagged alpha1D adrenoceptor expressed in HEK293 cells assessed as inhibition of agonist-induced calcium mobilization preincubated for 10 mins followed by agonist addition by fluo-4 AM dye-based assa2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
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.
AID239799Binding affinity for alpha 1a adrenoceptor2005Journal of medicinal chemistry, Jul-28, Volume: 48, Issue:15
Range and sensitivity as descriptors of molecular property spaces in dynamic QSAR analyses.
AID1301978Antagonist activity at alpha-1D adrenergic receptor (unknown origin) transfected in HEK293 cells assessed as reduction in agonist-induced calcium mobilization after 10 mins2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1301979Selectivity ratio of IC50 for alpha-1D adrenergic receptor (unknown origin) transfected in HEK293 cells to IC50 for human alpha-1A adrenergic receptor transfected in HEK293 cells2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID37478Binding affinity was tested on human Alpha-1B adrenergic receptor1997Journal of medicinal chemistry, Apr-25, Volume: 40, Issue:9
Pharmacological options in the treatment of benign prostatic hyperplasia.
AID1301982Selectivity ratio of IC50 for Sprague-Dawley rat aorta muscles to IC50 for Sprague-Dawley rat urethra smooth2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1301977Antagonist activity at human alpha-1B adrenergic receptor transfected in HEK293 cells assessed as reduction in agonist-induced calcium mobilization after 10 mins2016Journal of medicinal chemistry, 04-28, Volume: 59, Issue:8
Design, Synthesis, and Biological Evaluation of Indoline and Indole Derivatives as Potent and Selective α1A-Adrenoceptor Antagonists.
AID1320517Selectivity ratio of IC50 for human full-length N-terminal SNAP-tagged alpha1B adrenoceptor to IC50 for human full-length N-terminal SNAP-tagged alpha1A adrenoceptor2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320515Antagonist activity at human full-length N-terminal SNAP-tagged alpha1B adrenoceptor expressed in HEK293 cells assessed as inhibition of agonist-induced calcium mobilization preincubated for 10 mins followed by agonist addition by fluo-4 AM dye-based assa2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1320520Antagonist activity at alpha1B adrenoceptor in Sprague-Dawley rat aorta assessed as inhibition of norepinephrine-induced smooth muscle contraction after 20 mins2016Journal of medicinal chemistry, Oct-27, Volume: 59, Issue:20
Design, Synthesis, and Biological Evaluation of Novel Tetrahydroprotoberberine Derivatives (THPBs) as Selective α
AID1345971Human alpha1D-adrenoceptor (Adrenoceptors)1995Molecular pharmacology, Aug, Volume: 48, Issue:2
KMD-3213, a novel, potent, alpha 1a-adrenoceptor-selective antagonist: characterization using recombinant human alpha 1-adrenoceptors and native tissues.
AID1346000Human alpha1B-adrenoceptor (Adrenoceptors)1995Molecular pharmacology, Aug, Volume: 48, Issue:2
KMD-3213, a novel, potent, alpha 1a-adrenoceptor-selective antagonist: characterization using recombinant human alpha 1-adrenoceptors and native tissues.
AID1345908Human alpha1A-adrenoceptor (Adrenoceptors)1995Molecular pharmacology, Aug, Volume: 48, Issue:2
KMD-3213, a novel, potent, alpha 1a-adrenoceptor-selective antagonist: characterization using recombinant human alpha 1-adrenoceptors and native tissues.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (297)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's10 (3.37)18.2507
2000's60 (20.20)29.6817
2010's188 (63.30)24.3611
2020's39 (13.13)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 103.78

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 Index103.78 (24.57)
Research Supply Index5.97 (2.92)
Research Growth Index5.27 (4.65)
Search Engine Demand Index187.87 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (103.78)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials74 (23.34%)5.53%
Reviews50 (15.77%)6.00%
Case Studies7 (2.21%)4.05%
Observational9 (2.84%)0.25%
Other177 (55.84%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (30)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A 12-week, Open-label, Multi-center Study to Evaluate the Clinical Efficacy and Safety of Silodosin on Voiding Dysfunction Associated With Neurogenic Bladder [NCT01228370]Phase 495 participants (Actual)Interventional2011-04-30Completed
A Randomized, Double-blind, Placebo-controlled, Multicentre Study of KSO-0400 in BPH Patients With LUTS [NCT01222650]Phase 2400 participants InterventionalCompleted
Safety and Efficacy of 8mg Once-daily Versus 4mg Twice-daily Silodosin With Lower Urinary Tract Symptoms Suggestive of BPH ; 12-week, Double-blind, Randomized, Comparison, Multi-center Study [NCT01260129]Phase 4424 participants (Actual)Interventional2010-10-31Completed
A Multi-center, Prospective, Open-label, Single-arm, 12-weeks, Phase IV Trial to Evaluate the Efficacy and Safety of Silodosin on Nocturia for Patients With Benign Prostatic Hyperplasia [NCT02106182]Phase 4135 participants (Actual)Interventional2014-01-02Completed
Effectiveness of Silodosin in Medical Expulsive Therapy for Ureteral Pelvic Stone From 4 to 10 mm. [NCT02090439]Phase 38 participants (Actual)Interventional2014-07-31Terminated(stopped due to Following difficulties in patient recruitment, we were forced to stop the study prematurely.)
A Multi-Center, Randomized, Double-Blind, Placebo Controlled, Parallel Evaluation of the Efficacy and Safety of a New Drug in the Treatment of the Signs and Symptoms of Benign Prostatic Hyperplasia [NCT00224120]Phase 3462 participants (Actual)Interventional2005-05-31Completed
Differential Effect of Silodosin Versus Tamsulosin on Stone Clearance After ESWL [NCT01560091]Phase 30 participants (Actual)Interventional2012-03-31Withdrawn(stopped due to accrual of subjects did not occur as anticipated)
A 12-week, Open Label, Multi-center Study to Investigate the Efficacy and Safety of a α1A Adrenoceptor Selective Antagonist Silodosin on Urinary Disturbance Associated With Benign Prostatic Hyperplasia [NCT01259531]Phase 4100 participants (Actual)Interventional2010-12-31Completed
Prophylactic Versus Therapeutic Alpha-Blockers in Prostate Cancer Patients Undergoing Radical Course Radiation Therapy ± HDR Boost. A GROUQ Phase III Randomized Clinical Trial (PCS-VII). [NCT02220829]Phase 3188 participants (Anticipated)Interventional2016-06-30Recruiting
Comparison of Silodosin Versus Tamsulosin on Passage of Acutely Obstructing Ureteral Calculi (History of Last 4 Weeks) in Medical Expulsive Therapy [NCT05977647]Phase 4240 participants (Anticipated)Interventional2023-10-01Not yet recruiting
A Double-Blind, Placebo-Controlled Phase 2 Study of Silodosin 8 mg Daily for the Treatment of Nocturia in Men With Benign Prostatic Hyperplasia [NCT00793819]Phase 2215 participants (Actual)Interventional2009-01-31Completed
COMPARISON OF SILODOSIN AND TAMSULOSIN IN MEDICAL EXPULSIVE THERAPY OF DISTAL URETERIC CALCULI [NCT05790902]Phase 1142 participants (Actual)Interventional2022-09-01Completed
Efficacy of Silodosin in the Treatment of Symptomatic Benign Prostatic Hyperplasia (BPH) [NCT04107896]Phase 4141 participants (Actual)Interventional2017-08-31Completed
Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation [NCT02581826]Phase 240 participants (Anticipated)Interventional2013-10-31Recruiting
A Multi-Center, Double-Blind, Placebo-Controlled Investigation of Silodosin in the Treatment of Subjects With Moderate to Severe Abacterial Chronic Prostatitis/Chronic Pelvic Pain Syndrome [NCT00740779]Phase 2153 participants (Actual)Interventional2008-09-30Completed
A Multi-Center, Randomized, Double-Blind, Placebo Controlled, Parallel Evaluation of the Efficacy and Safety of a New Drug in the Treatment of the Signs and Symptoms of Benign Prostatic Hyperplasia [NCT00224107]Phase 3461 participants (Actual)Interventional2005-05-31Completed
A Multi-Center, Open-Label Evaluation of the Safety of a New Drug in the Treatment of the Signs and Symptoms of Benign Prostatic Hyperplasia [NCT00224133]Phase 3661 participants (Actual)Interventional2005-09-30Completed
Randomised Controlled Trial on Silodosin Versus Tamsulosin for Medical Expulsive Treatment of Ureteral Stones Size 5-10mm in Chinese [NCT05570084]Phase 380 participants (Anticipated)Interventional2022-05-31Recruiting
Silodosin vs Placebo in the Treatment of Female Lower Urinary Tract Symptoms: A Randomized Controlled Trial [NCT06114979]Phase 3278 participants (Anticipated)Interventional2023-11-01Not yet recruiting
A 4-week, Double-blind, Randomized, Comparative and Multi-center Clinical Trial to Evaluate the Efficacy and Safety of Silodosin in the Treatment of Natural Expulsion in Patients With Ureteral Stones. [NCT01533389]Phase 4114 participants (Actual)Interventional2011-10-31Completed
A Randomized, Double-blind, Placebo-controlled, Phase II Study of Silodosin in Patients With Urinary Calculi [NCT01539265]Phase 2300 participants InterventionalCompleted
Efficacy of Silodosin, Tadalafil Alone Versus Silodosin Plus Tadalafil as Medical Expulsive Therapy for Lower Ureteric Stones: A Prospective Randomized Placebo Controlled Study [NCT05789732]168 participants (Actual)Interventional2022-03-30Completed
Effectiveness and Safety of Silodosin in the Treatment of LUTS in Patients With Benign Prostatic Hyperplasia: a European Phase IV Clinical Study. The Silodosin in Real-life Evaluation Study [NCT01757769]Phase 41,036 participants (Actual)Interventional2011-05-31Completed
Clinical Progression and Costs in Benign Prostatic Hyperplasia Patients Treated With Early Versus Delayed Combination Therapy [NCT01435954]13,551 participants (Actual)Observational2010-08-31Completed
Effect of Perioperative Silodosin on Ureteric Dilatation in Retrograde Intrarenal Surgery [NCT05921370]Phase 360 participants (Anticipated)Interventional2023-04-04Enrolling by invitation
Comparative Study Between DJ Stenting and Sildosin After Uretroscopic Lithotripsy for Lower Ureteric Stones [NCT05823662]70 participants (Actual)Interventional2022-03-02Completed
Evaluation of the Efficacy and Safety of Silodosin vs. Tamsulosin and Placebo in the Treatment of the Signs and Symptoms of BPH. Multicentre, Randomised, Double-Blind, Controlled Trial With an Optional Long-Term, Open-Label Extension Phase. [NCT00359905]Phase 31,228 participants (Actual)Interventional2006-05-31Completed
Comparison of Silodosin vs. Tamsulosin on Spontaneous Passage of Acutely Obstructing Ureteral Calculi in Medical Expulsive Therapy [NCT02369744]Phase 41 participants (Actual)Interventional2013-03-31Terminated(stopped due to due to lack of suitable patient population)
The Efficacy and Safety of Silodosin Singly or Combined With Ningmitai Capsules in the Treatment of Benign Prostatic Hyperplasia (BPH) Complicated With Lower Urinary Tract Symptoms (LUTS) -A Multicenter, Prospective, Randomized, Double-blind, Positive Con [NCT05551221]Phase 4312 participants (Anticipated)Interventional2022-07-18Recruiting
A Double-Blind, Placebo-Controlled Study of Silodosin to Facilitate Urinary Stone Passage [NCT01144949]Phase 2239 participants (Actual)Interventional2010-06-30Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00224107 (2) [back to overview]International Prostate Symptom Score (IPSS)
NCT00224107 (2) [back to overview]Maximum Urine Flow Rate (Qmax)
NCT00224120 (2) [back to overview]Change From Baseline in Maximum Urine Flow Rate (Qmax) at 12 Weeks
NCT00224120 (2) [back to overview]Measuring Change From Baseline in International Prostate Symptom Score (IPSS) at 12 Weeks
NCT00224133 (1) [back to overview]Adverse Events
NCT00740779 (1) [back to overview]National Institutes of Health-Chronic Prostatitis Symptom Index (NIH-CPSI) Total Score.
NCT00793819 (1) [back to overview]Change in Nocturia Episodes
NCT01144949 (6) [back to overview]Change From Baseline in Average Score on the Brief Pain Inventory (Distal Stones)
NCT01144949 (6) [back to overview]Outpatient Narcotic Analgesic Use for Pain Relief
NCT01144949 (6) [back to overview]Spontaneous Stone Passage (All Stones) Without Need for Emergency Department Visits, Hospital Admissions, Surgical Intervention, or Other Interventional Procedures.
NCT01144949 (6) [back to overview]Spontaneous Stone Passage (Distal Stones) Without Need for Emergency Department Visits, Hospital Admissions, Surgical Intervention, or Other Interventional Procedures.
NCT01144949 (6) [back to overview]Time to Spontaneous Stone Passage (All Stones)
NCT01144949 (6) [back to overview]Time to Spontaneous Stone Passage (Distal Stones)

International Prostate Symptom Score (IPSS)

Change from baseline In IPSS at Week 12. IPSS uses a 0 to 35 scale; 0 best, 35 worse symptoms (NCT00224107)
Timeframe: 12 weeks

,
InterventionUnits on a 0 to 35 scale (Mean)
Baseline IPSSChange from Baseline in IPSS at Week 12
Placebo21.4-3.6
Silodosin21.5-6.5

[back to top]

Maximum Urine Flow Rate (Qmax)

Change from baseline in maximum urine flow rate (Qmax)at Week 12 (NCT00224107)
Timeframe: 12 weeks

,
InterventionmL/sec (Mean)
Baseline QmaxChange from Baseline in Qmax at Week 12
Placebo9.01.2
Silodosin9.02.2

[back to top]

Change From Baseline in Maximum Urine Flow Rate (Qmax) at 12 Weeks

(NCT00224120)
Timeframe: Baseline and 12 weeks

,
InterventionmL/min (Mean)
Baseline QmaxChange from baseline in Qmax at Week 12
Placebo8.71.9
Silodosin8.42.9

[back to top]

Measuring Change From Baseline in International Prostate Symptom Score (IPSS) at 12 Weeks

International prostate symptom score: Measuring prostate signs and symptoms asociated with benign prostatic hyperplasia on a 0 to 35 scale; 0 best, 35 worst symptoms (NCT00224120)
Timeframe: Baseline and 12 weeks

,
InterventionUnits on a scale (Mean)
Baseline IPSSChange from baseline in IPSS at Week 12
Placebo21.2-3.4
Silodosin21.2-6.3

[back to top]

Adverse Events

All reported adverse events were recorded. Clinically significant abnormal laboratory values or other clinical findings upon examination were also recorded as adverse events. (NCT00224133)
Timeframe: 9 months

Interventionparticipants (Number)
Patients with at least 1 adverse eventPatients with drug related adverse eventPatients with a serious adverse eventsPatients with a drug related serious adverse eventPatients discontinued due to an adverse eventDiscontinued for drug related adverse event
8 mg Silodosin Per Day With Food4311882908654

[back to top]

National Institutes of Health-Chronic Prostatitis Symptom Index (NIH-CPSI) Total Score.

Change from baseline In NIH-CPSI at Week 12. Three separate domain scores are calculated as pain, urinary symptoms, and quality of life impact. NIH-CPSI total score uses a 0 to 43 scale; 0 best, 43 worse symptoms. (NCT00740779)
Timeframe: 12 weeks

,,
InterventionUnits on a 0 to 43 scale (Mean)
Baseline NIH-CPSI Total ScoreChage from Baseline in NIH-CPSI at Week 12
Placebo27.9-8.5
Silodosin 4 mg26.0-12.1
Silodosin 8 mg26.8-10.2

[back to top]

Change in Nocturia Episodes

(NCT00793819)
Timeframe: 12 weeks

Interventionepisodes (Mean)
Silodosin 8mg-1.2
Placebo-1.0

[back to top]

Change From Baseline in Average Score on the Brief Pain Inventory (Distal Stones)

At each study visit, subjects were given a Brief Pain Inventory (BPI) Questionnaire to complete. The BPI collects subject-reported pain severity scores and assesses impact of pain upon the subject's daily life, on a 10-point scale (with 10 being the greatest severity/impact). Analysis was change from baseline to week 4. (NCT01144949)
Timeframe: 4 weeks

Interventionunits on a scale (Mean)
Silodsosin-2.6
Placebo-1.9

[back to top]

Outpatient Narcotic Analgesic Use for Pain Relief

Narcotic analgesic use was assessed through a subject diary. Analysis was performed on the number of days with analgesic use. (NCT01144949)
Timeframe: 4 weeks

InterventionDays (Mean)
Silodsosin5.8
Placebo5.5

[back to top]

Spontaneous Stone Passage (All Stones) Without Need for Emergency Department Visits, Hospital Admissions, Surgical Intervention, or Other Interventional Procedures.

The primary efficacy variable is the occurrence of spontaneous stone passage within 4 weeks, as determined by radiography. For this outcome measure, analysis includes all ureteral stones, regardless of location in the ureter. (NCT01144949)
Timeframe: 4 weeks

Interventionparticipants (Number)
Silodsosin60
Placebo52

[back to top]

Spontaneous Stone Passage (Distal Stones) Without Need for Emergency Department Visits, Hospital Admissions, Surgical Intervention, or Other Interventional Procedures.

"The primary efficacy variable is the occurrence of spontaneous distal stone passage within 4 weeks, as determined by radiography.~For this outcome measure, analysis includes only those stones located in the distal ureter." (NCT01144949)
Timeframe: 4 weeks

Interventionparticipants (Number)
Silodsosin36
Placebo27

[back to top]

Time to Spontaneous Stone Passage (All Stones)

Time to stone passage for all ureteral stones (regardless of location) is assessed by entries in subject diaries. (NCT01144949)
Timeframe: 4 weeks

Interventiondays (Mean)
Silodsosin22.3
Placebo22.9

[back to top]

Time to Spontaneous Stone Passage (Distal Stones)

Time to stone passage for distally-located stones is assessed by entries in subject diaries. (NCT01144949)
Timeframe: 4 weeks

Interventiondays (Mean)
Silodsosin19.6
Placebo22.0

[back to top]