Skip to main content
The Cochrane Database of Systematic Reviews logoLink to The Cochrane Database of Systematic Reviews
. 2017 Nov 22;2017(11):CD012615. doi: 10.1002/14651858.CD012615.pub2

Silodosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia

Jae Hung Jung 1,2,3,, Jiye Kim 4, Roderick MacDonald 5, Balaji Reddy 6, Myung Ha Kim 7, Philipp Dahm 2,3
Editor: Cochrane Urology Group
PMCID: PMC6486059  PMID: 29161773

Abstract

Background

A variety of alpha‐blockers are used for treating lower urinary tract symptoms (LUTS) in men with benign prostatic hyperplasia (BPH). 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.

Objectives

To assess the effects of silodosin for the treatment of LUTS in men with BPH.

Search methods

We performed a comprehensive search using multiple databases (Cochrane Library, MEDLINE, EMBASE, Scopus, Google Scholar, and Web of Science), trials registries, other sources of grey literature, and conference proceedings with no restrictions on the language of publication or publication status up until 13 June 2017.

Selection criteria

We included all parallel, randomized controlled trials. We also included cross‐over designs.

Data collection and analysis

Two review authors independently classified studies and abstracted data from the included studies. We performed statistical analyses using a random‐effects model and interpreted them according to the Cochrane Handbook for Systematic Reviews of Interventions. We rated the quality of evidence according to the GRADE approach.

Main results

We included 19 unique studies with 4295 randomized participants across four comparisons for short‐term follow‐up. The mean age, prostate volume, and International Prostate Symptom Score were 66.5 years, 38.2 mL, and 19.1, respectively.

Silodosin versus placebo

Based on four studies with a total of 1968 randomized participants, silodosin may reduce urologic symptom scores in an appreciable number of men (mean difference (MD) ‐2.65, 95% confidence interval (CI) ‐3.23 to ‐2.08; low‐quality evidence). Silodosin likely does not result in a clinically important reduction in quality of life (MD ‐0.42, 95% CI ‐0.71 to ‐0.13; moderate‐quality evidence). It may not increase rates of treatment withdrawal for any reason (relative risk (RR) 1.08, 95% CI 0.70 to 1.66; low‐quality evidence). We are uncertain about the effect of silodosin on cardiovascular adverse events (RR 1.28, 95% CI 0.67 to 2.45; very low‐quality evidence). Silodosin likely increases sexual adverse events (RR 26.07, 95% CI 12.36 to 54.97; moderate‐quality evidence); this would result in 180 more sexual adverse events per 1000 men (95% CI 82 more to 388 more).

Silodosin versus tamsulosin

Based on 13 studies with a total of 2129 randomized participants, silodosin may result in little to no difference in urologic symptom scores (MD ‐0.04, 95% CI ‐1.31 to 1.24; low‐quality evidence) and quality of life (MD ‐0.15, 95% CI ‐0.53 to 0.22; low‐quality evidence). We are uncertain about treatment withdrawals for any reason (RR 1.02, 95% CI 0.62 to 1.69; very low‐quality evidence). Silodosin may result in little to no difference in cardiovascular adverse events (RR 0.77, 95% CI 0.53 to 1.12; low‐quality evidence). Silodosin likely increases sexual adverse events (RR 6.05, 95% CI 3.55 to 10.31; moderate‐quality evidence); this would result in 141 more sexual adverse events per 1000 men (95% CI 71 more to 261 more).

Silodosin versus naftopidil

Based on five studies with a total of 763 randomized participants, silodosin may result in little to no differences in urologic symptom scores (MD ‐0.85, 95% CI ‐2.57 to 0.87; low‐quality evidence), quality of life (MD ‐0.17, 95% CI ‐0.60 to 0.27; low‐quality evidence), treatment withdrawal for any reason (RR 1.25, 95% CI 0.81 to 1.93; low‐quality evidence), and cardiovascular adverse events (RR 1.02, 95% CI 0.41 to 2.56; low‐quality evidence). Silodosin likely increases sexual adverse events (RR 5.93, 95% CI 2.16 to 16.29; moderate‐quality evidence); this would result in 74 more sexual adverse events per 1000 men (95% CI 17 more to 231 more).

Silodosin versus alfuzosin

Based on two studies with a total of 155 randomized participants, silodosin may or may not result in a clinically important increase in urologic symptom scores (MD 3.83, 95% CI 0.12 to 7.54; low‐quality evidence). Silodosin likely results in little to no difference in quality of life (MD 0.14, 95% CI ‐0.46 to 0.74; moderate‐quality evidence). We found no event of treatment withdrawal for any reason. Silodosin may not reduce cardiovascular adverse events (RR 0.67, 95% CI 0.36 to 1.24; low‐quality evidence) but likely increases sexual adverse events (RR 37.21, 95% CI 5.32 to 260.07; moderate‐quality evidence); this would result in 217 more sexual adverse events per 1000 men (95% CI 26 more to 1000 more).

Authors' conclusions

Silodosin may reduce urologic symptom scores in an appreciable number of men compared to placebo. Quality of life and treatment withdrawals for any reason appears similar. Its efficacy appears similar to that of other alpha blockers (tamsulosin, naftopidil and alfuzosin) but the rate of sexual side effects is likely higher. Our certainty in the estimates of effect was lowered due to study limitations, inconsistency and imprecision.

Plain language summary

Silodosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia

Review question

Does silodosin improve bothersome urinary symptoms in men with an enlarged prostate?

Background

Prostate enlargement is common in men as they get older and may cause difficulties in urination such as a weak stream, having to get up at night and a feeling of not emptying the bladder completely. Silodosin is a newer medication that may help with these symptoms and may cause fewer unwanted drug effects. We did this review to compare silodosin to placebo (dummy drug) and other medications.

Study characteristics

We included 19 studies with 4295 men. Participants' average age was 66.5 years. All studies included men aged over 40 years and reported that on average these men had a moderate degree of bothersome urinary symptoms.

Key results

Silodosin may improve urinary symptoms compared to placebo. It may have comparable effects on urinary symptoms, quality of life, treatment discontinuation for any reason, and unwanted drug effects compared to other medications. However, silodosin likely increases unwanted sexual side effects compared to placebo and other medications.

Quality of the evidence

The quality of evidence for most outcomes was low. This means that the true effect may be substantially different from what this review shows.

Summary of findings

Summary of findings for the main comparison. Silodosin compared to placebo for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term).

Silodosin compared to placebo for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term)
Participants: men with lower urinary tract symptoms suggesting benign prostatic hyperplasia
Setting: likely outpatients
Intervention: silodosin
Comparator: placebo
Outcomes № of participants
 (studies) Quality of the evidence
 (GRADE) Relative effect
 (95% CI) Anticipated absolute effects* (95% CI)
Risk with placebo Risk difference with silodosin
Urologic symptom scores 
 Assessed with: IPSS
 Scale from: 0 (best: not at all) to 35 (worst: almost always)
 Follow‐up: mean 3 months 1743
 (3 RCTs) ⊕⊕⊝⊝
 Lowa,b The mean change of urologic symptom scores ranged from ‐5.30 to ‐3.50 MD 2.65 lower
 (3.23 lower to 2.08 lower)
QoL 
 Assessed with: IPSS‐QoL
 Scale from: 0 (best: delighted) to 6 (worst: terrible)
 Follow‐up: mean 3 months 820
 (2 RCTs) ⊕⊕⊕⊝
 Moderatea,c The mean change of QoL ranged from ‐1.10 to ‐0.80 MD 0.42 lower
 (0.71 lower to 0.13 lower)
Treatment withdrawal due to any reason 
 Follow‐up: mean 3 months 1703
 (3 RCTs) ⊕⊕⊝⊝
 Lowa,b RR 1.08
 (0.70 to 1.66) Study population
83 per 1000 7 more per 1000
 (25 fewer to 55 more)
Cardiovascular adverse events 
 Follow‐up: mean 3 months 1967
 (4 RCTs) ⊕⊝⊝⊝
 Very lowa,b,d RR 1.28
 (0.67 to 2.45) Study population
42 per 1000 12 more per 1000
 (14 fewer to 61 more)
Assumed baseline riske
61 per 1000 17 more per 1000
 (20 fewer to 88 more)
Sexual adverse events 
 Follow‐up: mean 3 months 1967
 (4 RCTs) ⊕⊕⊝⊝
 Moderatea RR 26.07
 (12.36 to 54.97) Study population
7 per 1000 180 more per 1000
 (82 more to 388 more)
*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
 
 CI: confidence interval; IPSS: International Prostate Symptom Score; MD: mean difference; QoL: quality of life; RCTs: randomized controlled trials; RR: risk ratio
GRADE Working Group grades of evidenceHigh quality: we are very confident that the true effect lies close to that of the estimate of the effect.
 Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
 Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
 Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aDowngraded by one level for study limitations: unclear or high risk of bias for one or more domains among the included studies.
 bDowngraded by one level for imprecision: confidence interval crosses assumed threshold of clinically important difference.
 cNot downgraded for inconsistency despite moderate heterogeneity given that likely not clinically meaningful.
 dDowngraded by one level for inconsistency: substantial heterogeneity among the included studies.
 eEstimates for control event rates for cardiovascular adverse events come from Rosenzweig 1993.

Summary of findings 2. Silodosin compared to tamsulosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term).

Silodosin compared to tamsulosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term)
Participants: men with lower urinary tract symptoms suggesting benign prostatic hyperplasia
Setting: likely outpatients
Intervention: silodosin
Comparator: tamsulosin
Outcomes № of participants
 (studies) Quality of the evidence
 (GRADE) Relative effect
 (95% CI) Anticipated absolute effects* (95% CI)
Risk with tamsulosin Risk difference with silodosin
Urologic symptom scores 
 Assessed with: IPSS
 Scale from: 0 (best: not at all) to 35 (worst: almost always)
 Follow‐up: range 4 weeks to 12 months 1708
 (10 RCTs) ⊕⊕⊝⊝
 Lowa,b The mean change of urologic symptom scores ranged from ‐15.60 to ‐4.60 MD 0.04 lower
 (1.31 lower to 1.24 higher)
QoL 
 Assessed with: IPSS‐QoL
 Scale from: 0 (best: delighted) to 6 (worst: terrible)
 Follow‐up: range 4 weeks to 12 months 1707
 (10 RCTs) ⊕⊕⊝⊝
 Lowa,b The mean change of QoL ranged from ‐3.60 to ‐0.90 MD 0.15 lower
 (0.53 lower to 0.22 higher)
Treatment withdrawal due to any reason 
 Follow‐up: range 4 weeks to 12 months 1573
 (10 RCTs) ⊕⊝⊝⊝
 Very lowa,b,c RR 1.02
 (0.62 to 1.69) Study population
121 per 1000 2 fewer per 1000
 (46 fewer to 84 more)
Cardiovascular adverse events 
 Follow‐up: range 4 weeks to 3 months 1955
 (11 RCTs) ⊕⊕⊝⊝
 Lowa,c RR 0.77
 (0.53 to 1.12) Study population
63 per 1000 14 fewer per 1000
 (29 fewer to 8 more)
Sexual adverse events 
 Follow‐up: range 4 weeks to 3 months 1849
 (10 RCTs) ⊕⊕⊕⊝
 Moderatea RR 6.05
 (3.55 to 10.31) Study population
28 per 1000 141 more per 1000
 (71 more to 261 more)
*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
 
 CI: confidence interval; IPSS: International Prostate Symptom Score; MD: mean difference; QoL: quality of life; RCTs: randomized controlled trials; RR: risk ratio
GRADE Working Group grades of evidenceHigh quality: we are very confident that the true effect lies close to that of the estimate of the effect.
 Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
 Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
 Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aDowngraded by one level for study limitations: unclear or high risk of bias for one or more domains among the included studies.
 bDowngraded by one level for inconsistency: considerable heterogeneity among the included studies.
 cDowngraded by one level for imprecision: confidence interval crosses assumed threshold of clinically important difference.

Summary of findings 3. Silodosin compared to naftopidil for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term).

Silodosin compared to naftopidil for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term)
Participants: men with lower urinary tract symptoms suggesting benign prostatic hyperplasia
Setting: likely outpatients
Intervention: silodosin
Comparator: naftopidil
Outcomes № of participants
 (studies) Quality of the evidence
 (GRADE) Relative effect
 (95% CI) Anticipated absolute effects* (95% CI)
Risk with naftopidil Risk difference with silodosin
Urologic symptom scores 
 Assessed with: IPSS
 Scale from: 0 (best: not at all) to 35 (worst: almost always)
 Follow‐up: range 6 weeks to 3 months 652
 (5 RCTs) ⊕⊕⊝⊝
 Lowa,b The mean change of urologic symptom scores ranged from ‐7.52 to ‐3.56 MD 0.85 lower
 (2.57 lower to 0.87 higher)
QoL 
 Assessed with: IPSS‐QoL
 Scale from: 0 (best: delighted) to 6 (worst: terrible)
 Follow‐up: range 6 weeks to 3 months 652
 (5 RCTs) ⊕⊕⊝⊝
 Lowa,b The mean change of QoL ranged from ‐1.60 to ‐0.95 MD 0.17 lower
 (0.6 lower to 0.27 higher)
Treatment withdrawal due to any reason 
 Follow‐up: range 2 months to 3 months 659
 (4 RCTs) ⊕⊕⊝⊝
 Lowa,c RR 1.25
 (0.81 to 1.93) Study population
102 per 1000 25 more per 1000
 (19 fewer to 94 more)
Cardiovascular adverse events 
 Follow‐up: range 6 weeks to 3 months 808
 (5 RCTs) ⊕⊕⊝⊝
 Lowa,c RR 1.02
 (0.41 to 2.56) Study population
23 per 1000 0 more per 1000
 (13 fewer to 35 more)
Sexual adverse events 
 Follow‐up: range 6 weeks to 3 months 405
 (4 RCTs) ⊕⊕⊕⊝
 Moderatea RR 5.93
 (2.16 to 16.29) Study population
15 per 1000 74 more per 1000
 (17 more to 231 more)
*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
 
 CI: confidence interval; IPSS: International Prostate Symptom Score; MD: mean difference; QoL: quality of life; RCTs: randomized controlled trials; RR: risk ratio
GRADE Working Group grades of evidenceHigh quality: we are very confident that the true effect lies close to that of the estimate of the effect.
 Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
 Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
 Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aDowngraded by one level for study limitations: unclear or high risk of bias for one or more domains among the included studies.
 bDowngraded by one level for inconsistency: considerable heterogeneity among the included studies.
 cDowngraded by one level for imprecision: confidence interval crosses assumed threshold of clinically important difference.

Summary of findings 4. Silodosin compared to alfuzosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term).

Silodosin compared to alfuzosin for the treatment of lower urinary tract symptoms in men with benign prostatic hyperplasia (short term)
Participants: men with lower urinary tract symptoms suggesting benign prostatic hyperplasia
Setting: likely outpatients
Intervention: silodosin
Comparator: alfuzosin
Outcomes № of participants
 (studies) Quality of the evidence
 (GRADE) Relative effect
 (95% CI) Anticipated absolute effects* (95% CI)
Risk with alfuzosin Risk difference with silodosin
Urologic symptom scores 
 Assessed with: IPSS
 Scale from: 0 (best: not at all) to 35 (worst: almost always)
 Follow‐up: mean 3 months 60
 (1 RCT) ⊕⊕⊝⊝
 Lowa,b The mean change of urologic symptom scores was ‐16.93 MD 3.83 higher
 (0.12 higher to 7.54 higher)
QoL 
 Assessed with: IPSS‐QoL
 Scale from: 0 (best: delighted) to 6 (worst: terrible)
 Follow‐up: mean 3 months 60
 (1 RCT) ⊕⊕⊕⊝
 Moderatea The mean change of QoL was ‐4.27 MD 0.14 higher
 (0.46 lower to 0.74 higher)
Treatment withdrawal due to any reason 
 Follow‐up: mean 3 months 60
 (1 RCT) ⊕⊕⊝⊝
 Lowa,c Not estimable Study population
Cardiovascular adverse events 
 Follow‐up: mean 3 months 60
 (1 RCT) ⊕⊕⊝⊝
 Lowa,b RR 0.67
 (0.36 to 1.24) Study population
500 per 1000 165 fewer per 1000
 (320 fewer to 120 more)
Assumed baseline riskd
44 per 1000 15 fewer per 1000
 (11 fewer to 28 more)
Sexual adverse events 
 Follow‐up: mean 3 months 95
 (1 RCT) ⊕⊕⊝⊝
 Moderatea RR 37.21
 (5.32 to 260.07) Study population
21 per 1000 770 more per 1000
 (92 more to 5,512 more)
Assumed baseline riskd
6 per 1000 217 more per 1000
 (26 more to 1000 more)
*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
 
 CI: confidence interval; IPSS: International Prostate Symptom Score; MD: mean difference; QoL: quality of life; RCTs: randomized controlled trials; RR: risk ratio
GRADE Working Group grades of evidenceHigh quality: we are very confident that the true effect lies close to that of the estimate of the effect.
 Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
 Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
 Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aDowngraded by one level for study limitations: unclear or high risk of bias for one or more domains in the included study.
 bDowngraded by one level for imprecision: confidence interval crosses assumed threshold of clinically important difference.
 cDowngraded by one level for imprecision: no event (very rare event).
 dEstimates for control event rates for cardiovascular and sexual adverse events come from Van Kerrebroec 2002.

Background

Description of the condition

Benign prostatic hyperplasia (BPH) is characterized by an unregulated proliferative process of connective tissue, smooth muscle and glandular epithelium within the prostate, and is one of the most common conditions in elderly men (Roehrborn 2008). The prevalence of BPH rises significantly with increased age. Autopsy studies have observed a histological prevalence of 40% and 70% in the sixth and eighth decades of life, respectively (Barry 1997; Egan 2016; Roehrborn 2008). BPH can lead to troublesome lower urinary tract symptoms (LUTS), including storage disturbances (such as daytime urinary urgency and nocturia) or voiding disturbances (such as urinary hesitancy, weak urinary stream, straining to void, and prolonged voiding), or both (AUA Practice Guidelines Committee 2003; EAU 2017; McVary 2011). LUTS secondary to BPH (LUTS/BPH) may negatively impact on public health and reduce quality of life because BPH can progress over time and cause serious consequences, such as acute urinary retention (AUR), urinary tract infection, and upper urinary tract deterioration (Crawford 2006; Kozminski 2015; Martin 2014). In 2000, the direct treatment cost for LUTS/BPH in the USA was more than USD 1 billion. Based on the Hospital Episode Statistics data from the UK, LUTS/BPH is the fifth most expensive disease and costs GBP 1.16 billion each year (Cornu 2010).

Diagnosis

Initial assessment of LUTS/BPH includes patient history, review of current medications, physical exam including a digital rectal examination, urinalysis, a frequency/volume chart, and validated symptom questionnaires such as the International Prostate Symptom Score (IPSS) and BPH Impact Index (BII) (EAU 2017; Juliao 2012; McVary 2011). The IPSS questionnaire is composed of three domains related to storage symptoms (frequency, urgency, and nocturia); four domains related to voiding symptoms (hesitancy, weak stream, intermittence, and incomplete emptying); and one quality of life domain (AUA Practice Guidelines Committee 2003; Barry 1992). Seven symptom domains use a six‐point scale ranging from 0 (none) to 5 (5 or more) (Barry 1992). The quality of life domain is assigned a score from 1 to 6 (ordinal and range from 0 to 6: 0 = delighted, 1 = pleased, 2 = mostly satisfied, 3 = mixed, 4 = mostly dissatisfied, 5 = unhappy, 6 = terrible) (AUA Practice Guidelines Committee 2003; Barry 1992).

Simultaneous with the development of the IPSS, the BII was developed to assess the effect of LUTS/BPH on the men's health. The BII questionnaire is composed of four items: physical discomfort (0 = none, 1 = only a little, 2 = some, 3 = a lot); worry item (0 = none, 1 = only a little, 2 = some, 3 = a lot); bother item (0 = not at all bothersome, 1 = bothers me a little, 2 = bothers me some, 3 = bothers me a lot); the interference with usual activities item (0 = none of the time, 1 = a little of the time, 2 = some of the time, 3 = most of the time, 4 = all of the time) (Barry 1995). In addition, measurement of the maximum flow rate (Qmax) and postvoid residual (PVR) are widely used, noninvasive tests to evaluate joint functioning between bladder and prostate (EAU 2017; McVary 2011). Although diagnostic thresholds of Qmax and PVR have not been determined, low Qmax and large PVR have been associated with an increased risk of symptom deterioration (Crawford 2006; Kozminski 2015). Other optional tests include imaging of the urinary tract and prostate, urodynamic evaluation of bladder function, and cystoscopy to predict the response to medical therapy or determine the need for invasive therapy and therefore, the best approach (EAU 2017; McVary 2011).

Treatment

Treatment decisions are based on symptoms and degree of bother (EAU 2017; McVary 2011). Watchful waiting is a reasonable treatment strategy for men with mild LUTS (IPSS: 0 to 7) or those who do not perceive their symptoms to be particularly bothersome (Netto 1999). Lifestyle interventions such as modifying fluid intake or toileting behavior can be used as conservative treatments in men with mild LUTS (Yap 2009). In men with bothersome, moderate (IPSS: 8 to 19) to severe LUTS (IPSS: 20 to 35), surgical and medical therapies are the treatment options, and the choice of treatment depends on a number of factors such as the severity of disease, risk of progression, patient preference, and morbidity (EAU 2017; McVary 2011).

Recently, the dynamic component of BPH has been elucidated, with a focus on symptoms rather than prostate enlargement, which has led to a shift from surgery to medical treatment (Yoo 2012). Alpha‐adrenergic receptor blockers (ABs), which reduce smooth‐muscle tone in the prostate and bladder neck with/without 5‐alpha reductase inhibitors (5‐ARIs), which reduce prostate volume by inducing epithelial atrophy, are an established treatment in LUTS/BPH and have been widely used as first‐line therapy for decades (McConnell 2003; Milani 2005; Yoo 2012). In particular, ABs that can decrease smooth muscle tone in the prostate and bladder neck have been considered as fundamental pharmacotherapy for men with BPH (Cornu 2010; Milani 2005; Yoo 2012). Prior systematic reviews have shown that ABs can typically reduce the IPSS by 20% to 50% and increase the Qmax by 15% to 45% (MacDonald 2005; Wilt 2006). ABs are the most commonly prescribed category of drug, accounting for about 70% of all medications prescribed in 2008 (Cornu 2010). Adverse effects of ABs include postural hypotension, dizziness, headache, asthenia, syncope, peripheral edema, and retrograde ejaculation, which cause approximately 4% to 10% of men to withdraw from AB treatment (Djavan 1999; Gacci 2014; MacDonald 2005; Schulman 2003; Wilt 2006).

Other medical therapies, such as anticholinergics and desmopressin, have been used with ABs, depending on the main symptoms of patients (Brasure 2016; Dahm 2016; EAU 2017). Alternatively, a phosphodiesterase type 5 inhibitor, tadalafil (5 mg once daily), has been licensed for the treatment of male LUTS in the last few years, and various plant extracts have been proposed for the treatment of male LUTS (EAU 2017; Keehn 2016; Oelke 2012). In men with LUTS/BPH refractory to conservative/medical treatment or in cases of absolute clinical indications (e.g. AUR, recurrent urinary tract infection, bladder stones or diverticula, hematuria, or renal insufficiency), surgical techniques should be considered (EAU 2017; McVary 2011). Transurethral resection of the prostate (TURP) is regarded as the standard surgical procedure for the treatment of LUTS/BPH (EAU 2017; Juliao 2012; McVary 2011). Recently, minimal invasive laser therapies such as holmium laser enucleation of the prostate and photoselective vaporization were introduced as treatment alternatives to TURP (Nair 2016).

Description of the intervention

Silodosin is a new subtype selective AB that was approved in Japan in 2006, and more recently has received approval in the USA and Europe (Kawabe 2006a; Yoshida 2007). An initial randomized, placebo‐controlled trial, which was conducted in Japan showed a greater reduction in IPSS after 12 weeks' treatment compared to both tamsulosin – which is a well established AB for LUTS – and placebo. Furthermore, a clinical benefit in IPSS in the silodosin group over placebo was found from one week onwards (Kawabe 2006a). There were no significant differences in the incidence of cardiovascular adverse events between the silodosin and tamsulosin groups. Abnormal ejaculation was more frequent in the silodosin group than in the tamsulosin group (22.3% versus 1.6%), but only five men (2.9%) discontinued treatment due to abnormal ejaculation (Kawabe 2006a).

How the intervention might work

The A1a adrenergic receptors are a class of G protein‐coupled receptors that consists of three homologous subtypes, including A1a, A1b, and A1d receptors. The A1a receptor subtype predominates in the human prostate, bladder neck, and urethra (Minneman 1994; Schilit 2009). On the other hand, A1b receptor subtypes are mainly expressed in the peripheral vasculature and are important in the regulation of blood pressure. A1d receptor is expressed in the detrusor muscle of the bladder and the sacral region of the spinal cord (Minneman 1994; Schilit 2009). In the initial in vitro study, it was shown that silodosin had a high binding ratio (162:1) for A1a versus A1b receptors, which can be more specific to the lower urinary tract, and may therefore induce a therapeutic effect without cardiovascular side effects, such as dizziness, headache, and orthostatic hypotension related to vasodilation, compared with the other ABs, which have either no specific selectivity for the different adrenergic receptor subtypes (alfuzosin, doxazosin, terazosin) or a limited selectivity for the A1a receptor (tamsulosin) (Osman 2012; Schilit 2009; Yoshida 2007).

Why it is important to do this review

It is unclear whether the high affinity of silodosin to A1a adrenergic receptor actually translates into more clinical benefits and fewer adverse effects in clinical practice (Cui 2012; Novara 2013). While there are existing systematic reviews that compare silodosin to other agents used to treat men with LUTS/BPH, none so far has used the same rigorous methodology as Cochrane Reviews, which include the GRADE approach (Cui 2012; Fusco 2016; Novara 2013). In this era, with the availability of numerous ABs to treat LUTS/BPH, the findings of this Cochrane Review will be relevant to policymakers, healthcare providers and patients.

Objectives

To assess the effects of silodosin for the treatment of LUTS in men with BPH.

Methods

Criteria for considering studies for this review

Types of studies

We included parallel, randomized controlled trials regardless of their publication status or language of publication. We also included cross‐over designs.

Types of participants

We included adult men (aged 40 years and over) with LUTS/BPH. The age limitation was based on the observation that the prevalence of BPH increases in middle‐aged and older men (Barry 1997; Egan 2016), and is infrequent in younger men.

We excluded trials of men with a known neurogenic bladder due to spinal cord injury, multiple sclerosis, or central nervous system disease, and men who have been treated with surgery for BPH already. We included studies in which only a subset of participants were relevant to this review.

Types of interventions

We planned to investigate the following comparisons of experimental intervention versus comparator intervention. Concomitant interventions had to be the same in the experimental and comparator groups to establish fair comparisons.

Experimental interventions
  • Silodosin

  • Silodosin + any 5‐ARIs (if available)

Silodosin at 8 mg/day is the clinically recommended dosage in Japan, Europe, and the USA (Chapple 2011; Kawabe 2006a; Marks 2009). A recent study reported that silodosin at 4 mg/day was also useful for Japanese men with BPH, in order to improve treatment compliance (Seki 2015). We included trials with a dosage of silodosin at 8 mg/day (4 mg twice daily or 8 mg once daily) and 4 mg/day.

Comparator interventions
  • Placebo

  • Other ABs

  • Other ABs + any 5‐ARIs (if available)

Comparisons
  • Silodosin versus placebo

  • Silodosin versus other ABs

  • Silodosin + any 5‐ARIs versus other ABs + any 5‐ARIs (if available)

Types of outcome measures

We did not use the measurement of the outcomes assessed in this review as an eligibility criterion.

Primary outcomes
  • Urologic symptom scores

  • Quality of life

  • Treatment withdrawals for any reason

Secondary outcomes
  • Treatment withdrawals due to adverse events

  • AUR

  • Surgical intervention for LUTS/BPH

  • Cardiovascular adverse events

  • Sexual adverse events

Method and timing of outcome measurement
  • Urologic symptom scores: final value or change from baseline assessed with a validated scale (such as IPSS)

  • Quality of life: final value or change from baseline assessed with a validated scale (such as IPSS‐quality of life or BII scores)

  • Treatment withdrawals for any reason: defined as treatment discontinuation from any cause at any time after participants were randomized to intervention/comparator groups

  • Treatment withdrawals due to adverse events: defined as treatment discontinuation from adverse event at any time after participants were randomized to intervention/comparator groups

  • AUR: events requiring catheterization after intervention

  • Surgical intervention for LUTS/BPH: events requiring other surgical treatment modalities (e.g. TURP) after intervention

  • Cardiovascular adverse events: such as dizziness, headache, orthostatic hypotension, and syncope

  • Sexual adverse events: such as retrograde ejaculation, anejaculation, and decreased libido

We used the clinically important difference for the review outcomes to rate the quality of the evidence in the 'Summary of finding' tables (Jaeschke 1989; Johnston 2013). When the mean difference (MD) or risk ratio (RR) is equal to or larger than the minimal clinically important difference (MCID), we would assume that many participants may have gained a clinically meaningful improvement from treatment; when the MD is at least half of the MCID but less than the MCID, an appreciable number of participants have likely achieved a clinically meaningful improvement; and when the MD is less than one‐half of the MCID, it is unlikely that an appreciable number of participants have achieved a clinically meaningful improvement (Johnston 2010). We considered MCID in the IPSS and BII score to be 3 and 0.5 points, respectively (Barry 1995). We did not find any published information on a MCID for the IPSS‐quality of life question. We used a MCID of 1 to assess efficacy and comparative effectiveness (Brasure 2016). We did not establish thresholds for treatment withdrawals due to adverse events, AUR, surgical intervention for LUTS/BPH, cardiovascular adverse events, and sexual adverse events. We considered the clinically important differences of all listed outcomes above as a relative risk increase of at least 25% (Guyatt 2011a).

We considered outcomes measured up to and including 12 months after randomization as short term, and later than 12 months as long term.

  • Up to 12 months (short term)

  • More than 12 months (long term)

Main outcomes for 'Summary of findings' tables

We present 'Summary of findings' tables, reporting the following outcomes listed according to priority.

  • Urologic symptom scores

  • Quality of life

  • Treatment withdrawals for any reason

  • Cardiovascular adverse events

  • Sexual adverse events

Search methods for identification of studies

We performed a comprehensive search with no restrictions on the language of publication or publication status. We updated searches within three months prior to the anticipated publication of the review.

Electronic searches

We initially searched the following sources from inception of each database to 1 September 2016. The date of last search of
 all databases was 13 June 2017. See Appendix 1.

  • Cochrane Library (via Wiley)

    • Cochrane Database of Systematic Reviews

    • Cochrane Central Register of Controlled Trials (CENTRAL)

    • Database of Abstracts of Reviews of Effects

    • Health Technology Assessment Database

  • MEDLINE (via Pubmed)

  • EMBASE (via Ovid)

  • Scopus

  • Google Scholar

  • Web of Science

We also searched the following trials registers on 1 September 2016 and again on 13 June 2017.

  • ClinicalTrials.gov (clinicaltrials.gov/)

  • World Health Organization International Clinical Trials Registry Platform search portal (apps.who.int/trialsearch/)

  • Grey Literature Report (www.greylit.org/)

Searching other resources

We tried to identify other potentially eligible trials or ancillary publications by searching the reference lists of retrieved included trials, reviews, meta‐analyses and health technology assessment reports. We also contacted study authors of included trials to identify any further studies that we might have missed. We contacted drug/device manufacturers for ongoing or unpublished trials. We searched for unpublished studies by handsearching the abstract proceedings of the annual meetings of the American Urological Association, European Association of Urology, and International Continence Society for the last three years (2014 to 2016) and then updated the search for the most recent year (2017).

Data collection and analysis

Selection of studies

We used reference management software to identify and remove potential duplicate records (EndNote). Two review authors (JHJ, JK) independently scanned the abstract, title, or both, of remaining records retrieved, to determine which studies should be assessed further. Two review authors (JHJ, JK) investigated all potentially relevant records as full text, mapped records to studies, and classified studies as included studies, excluded studies, studies awaiting classification, or ongoing studies, in accordance with the criteria for each provided in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011a). We used Covidence for title/abstract, and full‐text screening. We resolved any discrepancies through consensus or recourse to a third review author (PD). If resolution of a disagreement was not possible, we planned to designate the study as 'awaiting classification' and contact study authors for clarification. We documented reasons for exclusion of studies that may have reasonably been expected to be included in the review in a Characteristics of excluded studies table. We present an adapted PRISMA flow diagram showing the process of study selection (Liberati 2009).

Data extraction and management

We developed a dedicated data abstraction form that we pilot tested ahead of time.

For studies that fulfilled the inclusion criteria, two review authors (JHJ, JK) independently abstracted the following information, which we provided in the Characteristics of included studies table.

  • Study design

  • Study dates

  • Study settings and country

  • Participant inclusion and exclusion criteria (e.g. age, baseline IPSS)

  • Participant details, baseline demographics (e.g. age, ethnic background, IPSS)

  • The number of participants by study and by study arm

  • Details of relevant experimental and comparator interventions such as such as frequency (e.g. once a day or twice a day) and treatment duration (in weeks or months)

  • Definitions of relevant outcomes, and method (e.g. type of instrument such as IPSS) and timing of outcome measurement (e.g. in weeks or months) as well as any relevant subgroups (e.g. based on age)

  • Study funding sources

  • Declarations of interest by primary investigators

We extracted outcome data relevant to this Cochrane Review as needed for calculation of summary statistics and measures of variance. For dichotomous outcomes, we obtained numbers of events and totals for population of a 2 x 2 table, as well as summary statistics with corresponding measures of variance. For continuous outcomes, we obtained means and standard deviations or data necessary to calculate this information.

We resolved any disagreements by discussion, or, if required, by consultation with a third review author (PD).

We provided information (including trial identifier), about potentially relevant, ongoing studies in the table Characteristics of ongoing studies.

We attempted to contact authors of included studies to obtain key missing data as needed.

Dealing with duplicate and companion publications

In the event of duplicate publications, companion documents or multiple reports of a primary study, we maximized yield of information by mapping all publications to unique studies and collating all available data. We used the most complete data‐set aggregated across all known publications. In case of doubt, we gave priority to the publication reporting the longest follow‐up associated with our primary or secondary outcomes.

Assessment of risk of bias in included studies

Two review authors (JHJ, JK) assessed the risk of bias of each included study independently. We resolved disagreements by consensus, or by consultation with a third review author (PD).

We assessed risk of bias using Cochrane's 'Risk of bias' assessment tool (Higgins 2011b) for the following domains.

  • Random sequence generation (selection bias)

  • Allocation concealment (selection bias)

  • Blinding of participants and personnel (performance bias)

  • Blinding of outcome assessment (detection bias)

  • Incomplete outcome data (attrition bias)

  • Selective reporting (reporting bias)

  • Other sources of bias

We judged risk of bias domains as 'low risk', 'high risk', or 'unclear risk' and evaluated individual bias items as described in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011b). We presented a 'Risk of bias' summary figure to illustrate these findings.

For selection bias (random sequence generation and allocation concealment), we evaluated risk of bias at a trial level.

For performance bias (blinding of participants and personnel), we considered that all outcomes were susceptible to performance bias and assessed in one group.

For detection bias (blinding of outcome assessment), we grouped outcomes as susceptible to detection bias (subjective) or not susceptible to detection bias (objective) outcomes.

We defined the following outcomes as subjective outcomes:

  • Urologic symptom scores

  • Quality of life

  • Treatment withdrawals for any reason

  • Treatment withdrawals due to adverse events

  • Cardiovascular adverse events

  • Sexual adverse events

We defined the following outcomes as objective outcomes:

  • AUR

  • Surgical intervention for LUTS/BPH

We initially assessed attrition bias (incomplete outcome data) on a per‐outcome basis but created groups of outcomes based on similar reporting characteristic.

For reporting bias (selective reporting), we evaluated risk of bias on a trial level.

We further summarized the risk of bias across domains for each outcome in each included study, as well as across studies and domains for each outcome, in accordance with the approach for summary assessments of the risk of bias presented in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011b).

Measures of treatment effect

We expressed dichotomous data as RRs with 95% confidence intervals (CIs). We expressed continuous data as MDs with 95% CIs unless different studies used different measures to assess the same outcome, in which case we expressed data as standardized MDs with 95% CIs.

Unit of analysis issues

The unit of analysis was the individual participant. For cross‐over trials or trials with more than two intervention groups, we planned to incorporate these study designs in meta‐analysis in accordance with guidance provided in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011c)

Dealing with missing data

We obtained missing data from study authors and performed intention‐to‐treat analyses if data were available; we otherwise performed available case analyses. We investigated attrition rates, for example, dropouts, losses to follow‐up and withdrawals, and critically appraised issues of missing data. We did not impute missing data.

Assessment of heterogeneity

We identified heterogeneity (inconsistency) through visual inspection of the forest plots to assess the amount of overlap of CIs, and the I2 statistic, which quantifies inconsistency across studies to assess the impact of heterogeneity in the meta‐analysis (Higgins 2002; Higgins 2003); we interpreted the I2 statistic as follows (Deeks 2011).

  • 0% to 40%: may not be important

  • 30% to 60%: may indicate moderate heterogeneity

  • 50% to 90%: may indicate substantial heterogeneity

  • 75% to 100%: considerable heterogeneity

When we found heterogeneity, we attempted to determine possible reasons for it by examining individual study and subgroup characteristics.

Assessment of reporting biases

We attempted to obtain study protocols to assess for selective outcome reporting.

If we included 10 studies or more investigating a particular outcome, we planned to use funnel plots to assess small study effects (Sterne 2011). Several explanations can be offered for the asymmetry of a funnel plot, including true heterogeneity of effect with respect to trial size, poor methodological design (and hence bias of small trials), and publication bias. We therefore interpreted results carefully. However, there were fewer than 10 studies for each comparison.

Data synthesis

We summarized data using a random‐effects model. We interpreted random‐effects meta‐analyses with due consideration of the whole distribution of effects. In addition, we performed statistical analyses according to the statistical guidelines contained in the Cochrane Handbook for Systematic Reviews of Interventions (Deeks 2011). For dichotomous outcomes, we used the Mantel‐Haenszel method; for continuous outcomes, we used the inverse variance method. We used Review Manager 5 software to perform analyses (RevMan 2014).

Subgroup analysis and investigation of heterogeneity

We expected the following characteristics to introduce clinical heterogeneity, and planned to carry out subgroup analyses with investigation of interactions.

  • Severity of baseline symptoms based on IPSS (0 to 7 = mildly symptomatic; 8 to 19 = moderately symptomatic; 20 to 35 = severely symptomatic)

  • Participant age (less than 65 years versus 65 years and older)

These subgroup analyses are based on the following observations.

  • The relationship between changes in IPSS scores and patient global ratings of improvement is influenced by the baseline scores (Barry 1995).

  • Tolerability of other ABs (as the main comparator) may differ by patient age but that this may be less the case for silodosin, which would represent a potential benefit of this agent (Kozminski 2015; Lepor 2007; Schilit 2009).

We planned to perform subgroup analyses limited to the primary outcomes.

Sensitivity analysis

We planned to perform sensitivity analyses limited to the primary outcomes in order to explore the influence of the following factors (when applicable) on effect sizes.

  • Restricting the analysis by taking into account risk of bias, by excluding studies at 'high risk' or 'unclear risk' of bias

'Summary of findings' tables

We presented the quality of the evidence for each outcome according to the GRADE approach, which takes into account five criteria not only related to internal validity (risk of bias, inconsistency, imprecision, publication bias), but also to external validity, such as directness of results (Guyatt 2008). For each comparison, two review authors (JHJ, JK, or RM) independently rated the quality of evidence for each outcome as 'high', 'moderate', 'low', or 'very low' using GRADEpro GDT 2015. We resolved any discrepancies by consensus, or, if needed, by arbitration by a third review author (PD). For each comparison, we presented a summary of the evidence for the main outcomes in a 'Summary of findings' table, which provides key information about the best estimate of the magnitude of the effect in relative terms and absolute differences for each relevant comparison of alternative management strategies; numbers of participants and studies addressing each important outcome; and the rating of the overall confidence in effect estimates for each outcome (Guyatt 2011b; Schünemann 2011).

Results

Description of studies

Results of the search

We identified 645 records through electronic database searching, including 52 records in trials registers. We found no records in the grey literature repository nor through handsearching abstract proceedings of relevant meetings from the last three years. We identified further six records through other sources by searching the reference lists of reviews (Alcántara Montero 2016; Brasure 2016) and one additional protocol for Takeshita 2016. After removal of duplicates, we screened the titles and abstracts of 458 records, and excluded 378 records. We screened 80 full‐text articles, and excluded 23 studies (24 records) that did not meet the inclusion criteria or were not relevant to the question under trial. Two studies are ongoing and 12 studies are awaiting classification. We included a total of 19 studies (42 records) in the review. The flow of literature through the assessment process is shown in the PRISMA flowchart (Figure 1).

1.

1

Flow diagram

Included studies

Details of included studies are presented elsewhere (see Characteristics of included studies; Table 5; Table 6).

1. Baseline characteristics of included studies.
Study name Trial
 period
 (year to
 year) Country Setting Description of participants Intervention(s)
 and
 comparator(s) Duration of
 intervention
 (duration of
 follow‐up) Age Prostate volume IPSS
Chapple 2011 2006‐2007 Europe 72 hospital clinics and inpatient units in 11 countries Men ≥ 50 years with LUTS (defined by a stable IPSS total score ≥ 13 points), bladder outlet obstruction (defined by a Qmax between 4 and 15 mL/s, with a minimum voided volume of ≥ 125 mL) Silodosin 8 mg once daily 12 weeks 65.8 ± 7.70 NR 19.1 ± 4.2
Tamsulosin 0.4 mg once daily 65.9 ± 7.41 18.9 ± 4.3
Placebo once daily 66.0 ± 7.37 19.3 ± 4.3
Jung 2012 NR South Korea NR Sexually active men with BPH Silodosin 8 mg once daily 4 weeks NR NR NR
Tamsulosin 0.2 mg once daily
Alfuzosin 10 mg once daily
Kawabe 2006a NR Japan 88 centers/outpatient Men ≥ 50 years with LUTS (IPSS of ≥ 8, an associated QoL score of ≥ 3) and prostate volume of ≥ 20 mL Silodosin 4 mg twice daily 12 weeks 65.4 ± 7.0 36.0 ± 16.9 17.1 ± 5.7
Tamsulosin 0.2 mg once daily 65.6 ± 7.0 35.7 ± 14.4 17.0 ± 5.7
Placebo twice daily 65.0 ± 6.9 35.2 ± 16.0 17.1 ± 6.1
Manjunatha 2016a 2013‐2014 India Tertiary care hospital Men ≥ 45 years with symptomatic BPH with LUTS (IPSS of ≥ 8, QoL of ≥ 3, and Qmax of < 15 mL/s, but > 4 mL/s with a voided volume of > 100 mL) Silodosin 8 mg once daily 12 weeks 64.00 ± 11.14 40.57 ± 16.45 15.93 ± 6.03
Tamsulosin 0.4 mg once daily 63.60 ± 9.05 40.33 ± 21.55 21.63 ± 7.63
Alfuzosin 10 mg once daily 63.43 ± 8.91 44.43 ± 27.72 19.2 ± 9.6
Marks 2009 2005‐2006 USA Multicenter Men ≥ 50 years with IPSS ≥ 13, Qmax 4 mL/s‐15 mL/s and a PVR < 250 mL Silodosin 8 mg once daily 12 weeks 64.6 ± 8.1 NR 21.3 ± 5.1
Placebo once daily 64.7 ± 8.1 21.3 ± 4.9
Masuda 2012 2009‐2011 Japan NR Men ≥ 50 years with prostate estimated volume of > 20 mL, IPSS ≥ 8, QoL score ≥ 3 points Silodosin 2 mg‐4 mg twice/d for 2 weeks, followed by 4 mg twice/d for 4 weeks 6 weeks (before cross‐over)/total 12 weeks 66.5 ± 5.6 38.8 ± 13.1 18.6 ± 5.5
Naftopidil 50 mg‐75 mg once/d for 2 weeks, followed by 75 mg once/d for 4 weeks 68.5 ± 5.7 45.7 ± 17.8 17.6 ± 5.0
Matsukawa 2016 2012‐2013 Japan 52 urologists participated at a total of 44 investigational sites/outpatients Men with LUTS (IPSS ≥ 8, IPSS QoL score 3) and prostate volume ≥ 20 mL Silodosin 4 mg/d for 4 weeks, followed by 8 mg/d for 8 weeks 12 weeks 70.6 ± 7.8 39.6 ± 16.7 18.8 ± 6.2
Naftopidil 50 mg/d for 4 weeks, followed by 75 mg/d for 8 weeks 70.3 ± 7.8 38.6 ± 14.8 18.9 ± 6.1
Miyakita 2010 2006‐2007 Japan Multicenter Men with IPSS ≥ 8 points; QoL score ≥ 3 points; prostate volume measured by ultrasonographic method ≥ 20 mL; void volume ≥ 100 mL; and maximal urinary flow rate (Qmax) < 15 mL/s Silodosin 4 mg twice daily 4 weeks (before cross‐over)/total 8 weeks 68.2 ± 8.6 41.3 ± 25.3 16.6 ± 5.2
Tamsulosin 0.2 mg once daily 70.1 ± 8.9 37.8 ± 16.3 18.2 ± 5.8
Natarajan 2015 2013‐2015 India Tertiary hospital Men > 50 years with bothersome LUTS from BPH and IPSS > 7 Silodosin 8 mg once daily 12 weeks 61 ‐ 62 NR NR
Tamsulosin 0.4 mg once daily
NCT00793819 2009 NR NR Men ≥ 50 years, with symptoms of moderate‐severe BPH and nocturia (≥ 2 episodes/night) Silodosin 8 mg daily 12 weeks 64.6 ± 8.03 NR NR
Placebo once daily 64.2 ± 8.92
Pande 2014 2012‐2013 India Tertiary care hospital/ outpatient Men > 50 years with bothersome LUTS from BPH and IPSS > 7 Silodosin 8 mg once daily 12 weeks 61.4 ± 7.88 42.0 ± 19.96 18.4 ± 3.32
Tamsulosin 0.4 mg once daily 62.6 ± 7.55 35.6 ± 9.56 18.4 ± 3.94
Shirakawa 2013 2007‐2011 Japan Kobe University School or other collaborating institutions Men with LUTS (total IPSS ≥ 8, QoL index ≥ 3) and prostate volume ≥ 20 mL Silodosin 4 mg twice daily 8 weeks 70.98 ± 6.69 38.24 ± 12.94 17.53 ± 5.4
Naftopidil 50 mg once daily 70.50 ± 6.58 39.39 ± 25.96 17.56 ± 6.7
Takeshita 2016 2011‐2014 Japan Four community‐based hospitals Men aged ≥ 50 years with LUTS/BPH, an IPSS of ≥ 8, QoL score of ≥ 3, and ultrasound‐estimated prostatic volume of ≥ 20 mL Silodosin 4 mg once daily 4 weeks (before cross‐over)/total 8 weeks 69.6 ± 5.4 38.7 ± 11.6 17.1 ± 7.3
Tamsulosin 0.2 mg once daily 69.4 ± 7.0 47.3 ± 30.4 15.2 ± 7.0
Watanabe 2011 2008‐2009 Japan Three institutions Men with LUTS associated with BPH and had an IPSS ≥ 8 and an IPSS‐QoL score ≥ 2 Silodosin 4 mg twice daily 4 weeks (before cross‐over)/total 8 weeks 69.3 ± 8.3 36.6 ± 18.3 16.4 ± 5.0
Tamsulosin 0.2 mg once daily 69.9 ± 8.4 35.1 ± 13.0 18.1 ± 6.2
Yamaguchi 2013 2007‐2010 Japan Nihon University School of Medicine Men with BPH, ≥ 50 years with significant LUTS (IPSS ≥ 8, QoL score ≥ 3) Silodosin 8 mg/ day 12 weeks 69.3 ± 7.8 33.2 ± 21.2 16.9 ± 5.5
Naftopidil 75 mg/ day 70.0 ± 7.0 39.5 ± 18.0 18.9 ± 7.0
Yamanishi 2011 NR NR NR Men with LUTS (IPSS total score ≥ 8, Qmax < 15 mL/s) and prostate volume > 20 mL Silodosin 4 mg twice daily 12 months 71.3 ± 8.2 42.0 ± 23.7 18.8 ± 7.3
Tamsulosin 0.2 ‐ 0.4 mg daily 72.2 ± 7.6 41.2 ± 23.0 17.8 ± 6.4
Yokoyama 2011 NR Japan Kawasaki Medical School Men aged 50–‐80 years and with IPSS ≥ 8 Silodosin 4 mg twice daily 12 weeks 70.2 ± 0.9 33.3 ± 2.3 18.7 ± 0.7
Tamsulosin 0.2 mg once daily 71.5 ± 1.1 32.5 ± 2.0 18.0 ± 1.1
Naftopidil 50 mg once daily 69.1 ± 1.2 35.0 ± 3.1 17.4 ± 0.8
Yokoyama 2012 2008‐2010 Japan Single center Men aged 50 years who had a total IPSS ≥ 8 and a QoL index ≥ 3 Silodosin 4 mg twice daily 3 months (before cross‐over)/1 month wash‐out/3 months (after cross‐over)/total 7 months 68.9 ± 5.6 35.0 ± 18.4 19.3 ± 4.9
Tamsulosin 0.2 mg once daily 70.0 ± 6.8 36.1 ± 15.5 21.1 ± 6.8
Yu 2011 2007‐2008 Taiwan Nine medical centers Men aged ≥ 40 years with an IPSS of ≥ 13 and prostate volume of ≥ 20 mL Silodosin 4 mg twice daily 12 weeks 67.5 ± 9.3 44.8 ± 24.2 19.3 ± 4.5
Tamsulosin 0.2 mg and one placebo 65.0 ± 8.8 38.2 ± 16.7 19.8 ± 4.5

BPH: benign prostatic hyperplasia; IPSS: International Prostate Symptom Score; LUTS: lower urinary tract symptoms; NR: not reported; PVR: postvoid residual; Qmax: maximum flow rate; QoL: quality of life

2. Participants in included studies.
Study name Intervention(s) and comparator(s) Screened/eligible (N) randomized (N) Analysed (N) Finishing trial (N (%))
Chapple 2011 Silodosin 8 mg 1228/955 381 346 356 (93.4)
Tamsulosin 0.4 mg 384 347 364 (94.7)
Placebo 190 168 172 (90.5)
Total 955 861 892 (93.4)
Jung 2012 Silodosin 8 mg NR/138 48 48 48 (100.0)
Tamsulosin 0.2 mg 43 43 43 (100.0)
Alfuzosin 10 mg 47 47 47 (100.0)
Total 138 138 138 (100.0)
Kawabe 2006a Silodosin 8 mg NR/457 176 175 175 (99.4)
Tamsulosin 0.2 mg 192 192 192 (100.0)
Placebo 89 89 89 (100.0)
Total 457 456 456 (99.7)
Manjunatha 2016a Silodosin 8 mg NR/90 30 30 30 (100.0)
Tamsulosin 0.4 mg 30 30 30 (100.0)
Alfuzosin 10 mg 30 30 30 (100.0)
Total 90 90 90 (100.0)
Marks 2009 Silodosin 8 mg 2849/923 466 466 413 (88.6)
Placebo 457 457 419 (91.6)
Total 923 923 832 (90.1)
Masuda 2012 Silodosin 4 mg or 8 mg NR/92 44 30/83a 30 (68.1)
Naftopidil 50 mg or 75 mg 48 34/79a 34 (70.8)
Total 92 64/162a 64 (69.5)
Matsukawa 2016 Silodosin 4 mg followed by 8 mg NR/350 175 157 157 (89.7)
Naftopidil 50 mg followed by 75 mg 175 157 157 (89.7)
Total 350 314 314 (89.7)
Miyakita 2010 Silodosin 8 mg NR/97 46 34 34 (73.9)
Tamsulosin 0.2 mg 51 31 31 (60.7)
Total 97 65 65 (67.0)
Natarajan 2015 Silodosin 8 mg NR/57 28 NR NR
Tamsulosin 0.4 mg 29 NR NR
Total 57 NR NR
NCT00793819 Silodosin 8 mg 215/209 111 111 97 (87.3)
Placebo 98 98 89 (90.8)
Total 209 209 186 (88.9)
Pande 2014 Silodosin 8 mg 102/61 32 26 26 (81.2)
Tamsulosin 0.4 mg 29 27 27 (93.1)
Total 61 53 53 (86.8)
Shirakawa 2013 Silodosin 8 mg NR/121 61 56/59a 56 (91.8)
Naftopidil 50 mg 60 56/57a 56 (93.3)
Total 121 112/116a 112 (92.5)
Takeshita 2016 Silodosin 4 mg NR/34 18 16 16 (88.8)
Tamsulosin 0.2 mg 16 14 14 (87.5)
Total 34 30 30 (88.2)
Watanabe 2011 Silodosin 4 mg NR/102 51 42/88a 42 (82.3)
Tamsulosin 0.2 mg 51 42/91a 42 (82.3)
Total 102 84/179a 42 (82.3)
Yamaguchi 2013 Silodosin 8 mg 109/109 58 53 53 (91.3)
Naftopidil 75 mg 51 44 44 (86.2)
Total 109 97 97 (88.9)
Yamanishi 2011 Silodosin 8 mg NR/149 75 NR NR
Tamsulosin 0.2 ‐ 0.4 mg 74 NR NR
Total 149 NR NR
Yokoyama 2011 Silodosin 8 mg 136/136 45 41 41 (91.1)
Tamsulosin 0.2 mg 45 39 39 (86.6)
Naftopidil 50 mg 46 42 42 (91.3)
Total 136 122 122 (89.7)
Yokoyama 2012 Silodosin 8 mg NR/46 23 23 23 (100.0)
Tamsulosin 0.2 mg 23 23 23 (100.0)
Total 46 46 46 (100.0)
Yu 2011 Silodosin 8 mg NR/209 105 87 87 (82.8)
Tamsulosin 0.2 mg 104 83 83 (79.8)
Total 209 170 170 (81.3)
Grand total Interventions: silodosin   1955   1684b
Compartors: placebo 834 769 (92.2)
Compartors: tamsulosin 1049 888b
Compartors: naftopidil 380 333 (87.6)
Comparator: alfuzosin 77 77 (100.0)
Overall 4295 3751b

N: number; NR: not reported

a Efficacy analysis/safety analysis.
 b The number of participants who finished trials were not reported in two included studies (Natarajan 2015; Yamanishi 2011).

Source of data

We included 16 published studies, two abstract proceedings (Jung 2012; Yamanishi 2011), and one record (NCT00793819) from a trials register. Of 16 published studies, we found two studies (Pande 2014; Yokoyama 2011) by searching the reference lists of an existing review (Brasure 2016) and the remaining studies were identified through our electronic database search. Eighteen studies were published in English and one was published in Japanese (Masuda 2012), which was translated into English using Google translator. We attempted to contact all corresponding authors of included trials to obtain additional information on study methodology and results, and received replies from nine (Chapple 2011; Jung 2012; Masuda 2012; Pande 2014; Takeshita 2016; Yamaguchi 2013; Yamanishi 2011; Yokoyama 2011; Yokoyama 2012; see Appendix 2).

Study design and settings

We included 14 parallel, randomized controlled trials (Chapple 2011; Jung 2012; Kawabe 2006a; Manjunatha 2016a; Marks 2009; Matsukawa 2016; Natarajan 2015; NCT00793819; Pande 2014; Shirakawa 2013; Yamaguchi 2013; Yamanishi 2011; Yokoyama 2011; Yu 2011) and five cross‐over trials (Masuda 2012; Miyakita 2010; Takeshita 2016; Watanabe 2011; Yokoyama 2012). Five of 19 studies were reported as 'double‐blinded'. The participants and investigators were blinded in three studies (Marks 2009; NCT00793819; Yu 2011) and participants and personnel were blinded in one study (Chapple 2011). One study was reported to be 'double‐blinded' but it was not clear who was blinded (Kawabe 2006a). Four studies were open‐label trials (Jung 2012; Manjunatha 2016a; Matsukawa 2016; Shirakawa 2013). The remaining five trials had no information regarding blinding. There were three trials with run‐in periods (Chapple 2011; Marks 2009; Yu 2011).

All studies were likely conducted in an outpatient clinic setting. Three studies explicitly stated that the trial was conducted in an outpatient clinic setting (Kawabe 2006a; Matsukawa 2016; Pande 2014). Most of the included studies (Jung 2012; Kawabe 2006a; Manjunatha 2016a; Masuda 2012; Matsukawa 2016; Miyakita 2010; Natarajan 2015; Pande 2014; Shirakawa 2013; Takeshita 2016; Watanabe 2011; Yamaguchi 2013; Yokoyama 2011; Yokoyama 2012; Yu 2011) were performed in Asia, except two studies, which were performed in Europe (Chapple 2011) and the USA (Marks 2009). Eight trials (Chapple 2011; Kawabe 2006a; Marks 2009; Masuda 2012; Matsukawa 2016; Miyakita 2010; Takeshita 2016; Watanabe 2011) were multicenter trials. The studies were performed from 2005 to 2015.

Participants

We included a total of 4295 randomized participants (silodosin 1955, placebo 834, tamsulosin 1049, naftopidil 380, alfuzosin 77), of which a total of 3751 finished the trials (silodosin 1684, placebo 769, tamsulosin 888, naftopidil 333, alfuzosin 77). However, two studies (Natarajan 2015; Yamanishi 2011), that compared silodosin to tamsulosin, did not report the number of participants who completed the trial in each group. All studies included men aged over 40 years. The mean age, prostate volume, prostate specific antigen (PSA), International Prostate Symptom Score (IPSS), and maximum flow rate (Qmax) of the participants were 66.5 years, 38.2 mL, 3.14 ng/mL, 19.1, and 9.5 mL/s, respectively.

Most of the studies included participants with lower urinary tract symptoms (LUTS) of a total IPSS more than 8 (Kawabe 2006a; Manjunatha 2016a; Masuda 2012; Matsukawa 2016; Miyakita 2010; Natarajan 2015; Pande 2014; Shirakawa 2013; Takeshita 2016; Watanabe 2011; Yamaguchi 2013; Yamanishi 2011; Yokoyama 2011; Yokoyama 2012). Participants with IPSS more than 13 were included in three studies (Chapple 2011; Marks 2009; Yu 2011). Two studies (abstract Jung 2012, protocol NCT00793819) did not specify the inclusion criteria for LUTS in detail. Eight out of 19 studies included participants with a prostate volume greater than 20 mL (Kawabe 2006a; Masuda 2012; Matsukawa 2016; Miyakita 2010; Shirakawa 2013; Takeshita 2016; Yamanishi 2011; Yu 2011). Nine studies used 15mL/s or less in Qmax as an inclusion criterion (Chapple 2011; Kawabe 2006a; Manjunatha 2016a; Marks 2009; Matsukawa 2016; Miyakita 2010; Shirakawa 2013; Yamanishi 2011; Yu 2011). Major exclusion criteria included LUTS from any cause other than benign prostatic hyperplasia (BPH), prior treatment with other BPH medical therapy, recent acute urinary retention (AUR), raised prostate‐specific antigen (PSA) level suspicious of prostate cancer, a history of prostate cancer or prior prostate‐related surgery. Chapple 2011 excluded men with improvement in the IPSS total score of 25% or more in the run‐in period and Marks 2009 excluded 615 participants due to adverse events, investigator recommendation, lack of efficacy, loss‐to‐follow‐up, protocol violation, voluntary withdrawal, or other issues during the run‐in period.

Intervention(s) and comparator(s)

Silodosin was administered as an oral dose of 8 mg (8 mg once daily or 4 mg twice daily) in all studies except one (Takeshita 2016; 4 mg once daily). Two studies titrated the drug dose (4 mg followed by 8 mg) during the study period (Masuda 2012; Matsukawa 2016).

Four agents, namely placebo, tamsulosin, naftopidil, and alfuzosin were used as comparators. All comparators were administrated orally. Placebo was administrated in accordance with frequencies of silodosin (Chapple 2011; Kawabe 2006a; Marks 2009; NCT00793819). Tamsulosin was administered as an oral dose of 0.2 mg (Jung 2012; Kawabe 2006a; Miyakita 2010; Takeshita 2016; Watanabe 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) or 0.4 mg once daily (Chapple 2011; Manjunatha 2016a; Natarajan 2015; Pande 2014). One study did not specify the exact dose of tamsulosin (Yamanishi 2011). Naftopidil was administered as an oral dose of 50 mg (Shirakawa 2013; Yokoyama 2011) or 75 mg once daily (Yamaguchi 2013). Two studies titrated the drug dose (50 mg followed by 75 mg) during the study period (Masuda 2012; Matsukawa 2016). Alfuzosin was administered as an oral dose of 10 mg once daily (Jung 2012; Manjunatha 2016a).

The duration of intervention was 12 weeks in 11 (of 19) studies (Chapple 2011; Kawabe 2006a; Manjunatha 2016a; Marks 2009; Matsukawa 2016; Natarajan 2015; Pande 2014; NCT00793819; Yamaguchi 2013; Yokoyama 2011; Yu 2011). Jung 2012 and Shirakawa 2013 followed the participants for four weeks and eight weeks only, respectively. Yamanishi 2011 reported 12‐month follow‐up data. For cross‐over trials, three (Miyakita 2010; Takeshita 2016; Watanabe 2011), one (Masuda 2012), and one study (Yokoyama 2012) reported 4 weeks', 6 weeks', and 12 weeks' follow‐up outcomes before the cross‐over, respectively.

Comparisons

We included four comparisons in this review: four studies compared silodosin to placebo (Chapple 2011; Kawabe 2006a; Marks 2009; NCT00793819). Thirteen (Chapple 2011; Jung 2012; Kawabe 2006a; Manjunatha 2016a; Miyakita 2010; Natarajan 2015; Pande 2014; Takeshita 2016; Watanabe 2011; Yamanishi 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) compared silodosin to tamsulosin, five (Masuda 2012; Matsukawa 2016; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) compared silodosin to naftopidil, and two studies (Jung 2012; Manjunatha 2016a) compared silodosin to alfuzosin. All studies used silodosin, placebo, or other ABs as monotherapy. There were no trials that compared combination therapy with silodosin or any 5‐ARIs to combination therapy with other ABs and any 5‐ARIs.

Outcomes

We identified all primary outcomes in each of the included studies for four comparisons. No trial reported on our predefined secondary outcomes of AUR or surgical intervention for LUTS/BPH. However, we were able to obtain additional information by contact with the study authors (Chapple 2011; Pande 2014; Yamaguchi 2013; Yokoyama 2011) which we used for the data analysis across three comparisons. Given the nature of cross‐over design studies, we assumed they were not applicable to the outcomes of AUR and surgical intervention for LUTS/BPH for the analysis. Other secondary outcomes were reported in at least one of the included studies.

Funding sources and conflicts of interest

Four reported no funding source (Jung 2012; Kawabe 2006a; Manjunatha 2016a; Yokoyama 2012), and five were supported by pharmaceutical companies (Chapple 2011; Marks 2009; NCT00793819; Pande 2014; Yu 2011). The remaining did not mention a funding source.

Nine studies reported no conflicts of interest (Kawabe 2006a; Manjunatha 2016a; Pande 2014; Shirakawa 2013; Takeshita 2016; Watanabe 2011; Yamaguchi 2013; Yokoyama 2011; Yokoyama 2012), and four reported having relationships with pharmaceutical companies (Chapple 2011; Marks 2009; NCT00793819; Yu 2011). The remaining did not mention conflicts of interest.

Excluded studies

We excluded 23 studies (24 records) out of 58 studies (80 records) after evaluation of the full‐text publications. Seven studies were reviews (Abramowicz 2009; Alcántara Montero 2016; Curran 2011; Montorsi 2010; Montorsi 2013; Prescrire Int 2011; Prescrire Int 2012). Six studies had the wrong comparator (Cakiroglu 2016; JPRN‐UMIN000007917; Matsukawa 2012; Matsukawa 2017; Yoshida 2017; Yoshihisa 2012). Each of four studies had the wrong study design (Araki 2013; Boeri 2016; Chapple 2009; Kawabe 2006b) and wrong study outcome (Kobayashi 2008; Kobayashi 2009; Manjunatha 2016b; Zhou 2011), respectively. Two studies had the wrong population (Michel 2011; Roehrborn 2009). Details of excluded studies are presented elsewhere (see Characteristics of excluded studies).

Studies awaiting classification and ongoing trials

We found 12 studies awaiting classification (CTRI/2010/091/000526; Devana 2014; Jha 2015; JPRN‐UMIN000003125; JPRN‐UMIN000005151; JPRN‐UMIN000008538; JPRN‐UMIN000011556; Mandal 2013; Manohar 2014; Miyamae 2011; NCT01222650; Pawar 2015) that did not provide usable outcome data and two ongoing studies (JPRN‐UMIN000003609; CTRI/2013/10/004112). Details of studies awaiting classification and ongoing trials are presented elsewhere (see Characteristics of studies awaiting classification; Characteristics of ongoing studies).

Risk of bias in included studies

See Figure 2; Figure 3.

2.

2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies

3.

3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study

Allocation

Random sequence generation
Allocation concealment

Blinding

Blinding of participants and personnel
Blinding of outcome assessment
  • Subjective outcomes: we rated three studies as low risk of bias for subjective outcomes (Marks 2009; NCT00793819; Yu 2011). We judged nine studies (Jung 2012; Manjunatha 2016a; Matsukawa 2016; Pande 2014; Shirakawa 2013; Takeshita 2016; Watanabe 2011; Yamaguchi 2013; Yokoyama 2012) as high risk of bias and the remaining as unclear risk of bias for subjective outcomes. (Subjective outcomes: urologic symptom scores, quality of life, treatment withdrawal for any reason, treatment withdrawal due to adverse events, cardiovascular adverse events, and sexual adverse events.)

  • Objective outcomes: we rated all studies as low risk of bias for objective outcomes because objective outcomes are not likely to be affected by lack of blinding. (Objective outcomes: AUR and surgical intervention for LUTS/BPH.)

Incomplete outcome data

Selective reporting

Other potential sources of bias

Effects of interventions

See: Table 1; Table 2; Table 3; Table 4

Details are presented elsewhere (see Table 1; Table 2; Table 3; Table 4).

1. Silodosin versus placebo

1.1. Urologic symptom scores

We included three randomized controlled trials (RCTs) (Chapple 2011; Kawabe 2006a; Marks 2009) with 1743 participants in the analysis (silodosin 1012, placebo 731). Silodosin may reduce urologic symptom scores in an appreciable number of men (MD ‐2.65, 95% CI ‐3.23 to ‐2.08; I2 = 0%). We rated the quality of the evidence as low, downgrading for study limitations and imprecision (Analysis 1.1).

1.1. Analysis.

1.1

Comparison 1 Silodosin versus placebo, Outcome 1 Urologic symptom scores (short term).

1.2. Quality of life

We included two RCTs (Chapple 2011; Kawabe 2006a) with 820 participants in the analysis (silodosin 546, placebo 274). Silodosin likely results in little or no difference in quality of life (MD ‐0.42, 95% CI ‐0.71 to ‐0.13; I2 = 57%). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 1.2).

1.2. Analysis.

1.2

Comparison 1 Silodosin versus placebo, Outcome 2 Quality of life (short term).

1.3. Treatment withdrawal for any reason

We included three RCTs (Chapple 2011; Marks 2009; NCT00793819) with 1703 participants in the analysis (silodosin 958, placebo 745). Silodosin may result in little or no difference in treatment withdrawal for any reason (RR 1.08, 95% CI 0.70 to 1.66; I2 = 42%). We rated the quality of the evidence as low, downgrading for study limitations and imprecision (Analysis 1.3).

1.3. Analysis.

1.3

Comparison 1 Silodosin versus placebo, Outcome 3 Treatment withdrawal due to any reason (short term).

1.4. Treatment withdrawal due to adverse events

We included four RCTs (Chapple 2011; Kawabe 2006a; Marks 2009; NCT00793819) with 1967 participants in the analysis (silodosin 1133, placebo 834). Silodosin likely increases treatment withdrawal due to adverse events (RR 2.29, 95% CI 1.41 to 3.72; I2 = 0%). Assuming a baseline risk of treatment withdrawal due to adverse events to be 13.0% (Preston 2000), silodosin would result in 168 more treatment withdrawals per 1000 men (95% CI 53 more to 354 more). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 1.4).

1.4. Analysis.

1.4

Comparison 1 Silodosin versus placebo, Outcome 4 Treatment withdrawal due to adverse events (short term).

1.5. AUR

We included two RCTs (Chapple 2011; NCT00793819) with 780 participants in the analysis (silodosin 492, placebo 288). We found no event for AUR in either study group.

1.6. Surgical intervention for LUTS/BPH

We included two RCTs (Chapple 2011; NCT00793819) with 780 participants in the analysis (silodosin 492, placebo 288). We found no event for surgical intervention for LUTS/BPH in either study group.

1.7. Cardiovascular adverse events

We included four RCTs (Chapple 2011; Kawabe 2006a; Marks 2009; NCT00793819) with 1967 participants in the analysis (silodosin 1133, placebo 834). We are uncertain about the effect of silodosin on cardiovascular adverse events (RR 1.28, 95% CI 0.67 to 2.45; I2 = 55%). We rated the quality of the evidence as very low, downgrading for study limitations, inconsistency, and imprecision (Analysis 1.5).

1.5. Analysis.

1.5

Comparison 1 Silodosin versus placebo, Outcome 5 Cardiovascular adverse events (short term).

1.8. Sexual adverse events

We included four RCTs (Chapple 2011; Kawabe 2006a; Marks 2009; NCT00793819) with 1967 participants in the analysis (silodosin 1133, placebo 834). Silodosin likely increases sexual adverse events (RR 26.07, 95% CI 12.36 to 54.97; I2 = 0%). Silodosin would result in 180 more sexual adverse events per 1000 men (95% CI 82 more to 388 more). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 1.6).

1.6. Analysis.

1.6

Comparison 1 Silodosin versus placebo, Outcome 6 Sexual adverse events (short term).

Subgroup analysis

We were unable to perform any predefined subgroup analyses because there were no relevant data in the included studies. While we found pooled analyses (Novara 2015) from two included studies (Chapple 2011; Marks 2009) that evaluated the efficacy and safety of silodosin versus placebo, we were unable to replicate these analyses ourselves.

Sensitivity analysis

We rated three RCTs (Chapple 2011; Marks 2009; NCT00793819) as low risk of bias and reanalyzed the data focused on these studies alone.

  • Urologic symptom scores: we found an MD of ‐2.60 (95% CI ‐3.28 to ‐1.92) with the studies rated as low risk of bias (Chapple 2011; Marks 2009) versus an MD of ‐2.65 (95% CI ‐3.23 to ‐2.08) with all included studies (Chapple 2011; Kawabe 2006a; Marks 2009).

  • Quality of life: we found an MD of ‐0.30 (95% CI ‐0.51 to ‐0.09) with the study rated as low risk of bias (Chapple 2011) versus an MD of ‐0.42 (95% CI ‐0.71 to ‐0.13) with all included studies (Chapple 2011; Kawabe 2006a).

  • Treatment withdrawal for any reason: we rated all included studies as low risk of bias and we were unable to perform sensitivity analyses for this outcome.

2. Silodosin versus tamsulosin

2.1. Urologic symptom scores

We included 10 RCTs (Chapple 2011; Kawabe 2006a; Manjunatha 2016a; Miyakita 2010; Pande 2014; Takeshita 2016; Yamanishi 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) with 1708 participants in the analysis (silodosin 859, tamsulosin 849). We used the final value in one study (Pande 2014) and change from baseline in the remaining studies. Silodosin may result in little to no difference in urologic symptom scores (MD ‐0.04, 95% CI ‐1.31 to 1.24; I2 = 87%). We rated the quality of the evidence as low, downgrading for study limitations and inconsistency (Analysis 2.1).

2.1. Analysis.

2.1

Comparison 2 Silodosin versus tamsulosin, Outcome 1 Urologic symptom scores (short term).

2.2. Quality of life

We included 10 RCTs (Chapple 2011; Kawabe 2006a; Manjunatha 2016a; Miyakita 2010; Pande 2014; Takeshita 2016; Yamanishi 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) with 1707 participants in the analysis (silodosin 859, tamsulosin 848). We used the final value in two studies (Miyakita 2010; Pande 2014) and change from baseline in the remaining studies. Silodosin may result in little to no difference in quality of life (MD ‐0.15, 95% CI ‐0.53 to 0.22; I2 = 93%). We rated the quality of the evidence as low, downgrading for study limitations and inconsistency (Analysis 2.2).

2.2. Analysis.

2.2

Comparison 2 Silodosin versus tamsulosin, Outcome 2 Quality of life (short term).

2.3. Treatment withdrawal for any reason

We included 10 RCTs (Chapple 2011; Manjunatha 2016a; Natarajan 2015; Pande 2014; Takeshita 2016; Watanabe 2011; Yamanishi 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) with 1573 participants in the analysis (silodosin 788, tamsulosin 785). For the cross‐over trial Watanabe 2011, we applied the conservative approach of only using the number of participants who had been initially randomized, due to lack of information about the number of participants in the analysis. We are uncertain about the effect of silodosin on treatment withdrawal for any reason (RR 1.02, 95% CI 0.62 to 1.69; I2 = 60%). We rated the quality of the evidence as very low, downgrading for study limitations, inconsistency, and imprecision (Analysis 2.3).

2.3. Analysis.

2.3

Comparison 2 Silodosin versus tamsulosin, Outcome 3 Treatment withdrawal due to any reason (short term).

2.4. Treament withdrawal due to adverse events

We included nine RCTs (Chapple 2011; Kawabe 2006a; Manjunatha 2016a; Natarajan 2015; Pande 2014; Takeshita 2016; Watanabe 2011; Yokoyama 2011; Yokoyama 2012) with 1572 participants in the analysis (silodosin 779, tamsulosin 793). For the cross‐over trials Watanabe 2011 and Yokoyama 2012, we used only the number of participants who had been initially randomized due to lack of information about the number of participants in the analysis. Silodosin may or may not result in an important increase in treatment withdrawal due to adverse events (RR 1.96, 95% CI 1.12 to 3.44; I2 = 0%). Silodosin would result in 21 more treatment withdrawals for any reason per 1000 men (95% CI 3 more to 52 more). We rated the quality of the evidence as low, downgrading for study limitations and imprecision (Analysis 2.4).

2.4. Analysis.

2.4

Comparison 2 Silodosin versus tamsulosin, Outcome 4 Treatment withdrawal due to adverse events (short term).

2.5. AUR

We included three RCTs (Chapple 2011; Pande 2014; Yokoyama 2011) with 906 participants in the analysis (silodosin 454, tamsulosin 452). We found no event for AUR in either study group.

2.6. Surgical intervention for LUTS/BPH

We included three RCTs (Chapple 2011; Pande 2014; Yokoyama 2011) with 906 participants in the analysis (silodosin 454, tamsulosin 452). We found no event for surgical intervention for LUTS/BPH in either study group.

2.7. Cardiovascular adverse events

We included 11 RCTs (Chapple 2011; Kawabe 2006a; Manjunatha 2016a; Miyakita 2010; Natarajan 2015; Pande 2014; Takeshita 2016; Watanabe 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) with 1955 participants in the analysis (silodosin 967, tamsulosin 988). For cross‐over trials, we took all measurements from silodosin periods and all measurements from tamsulosin periods in Watanabe 2011 and analysed these as if the trial were a parallel‐group trial of silodosin versus tamsulosin. However, we used only the number of participants who had been initially randomized due to lack of information about the number of participants in the analysis (Miyakita 2010; Takeshita 2016; Yokoyama 2012). Silodosin may result in little to no difference in cardiovascular adverse events (RR 0.77, 95% CI 0.53 to 1.12; I2 = 3%). We rated the quality of the evidence as low downgrading for study limitations and imprecision (Analysis 2.5).

2.5. Analysis.

2.5

Comparison 2 Silodosin versus tamsulosin, Outcome 5 Cardiovascular adverse events (short term).

2.8. Sexual adverse events

We included 10 RCTs (Chapple 2011; Jung 2012; Kawabe 2006a; Natarajan 2015; Pande 2014; Takeshita 2016; Watanabe 2011; Yokoyama 2011; Yokoyama 2012; Yu 2011) with 1849 participants in the analysis (silodosin 920, tamsulosin 929). For cross‐over trials, we took all measurements from silodosin periods and all measurements from tamsulosin periods in Watanabe 2011. However, we used only the number of participants who had been initially randomized due to lack of information about the number of participants in the analysis (Yokoyama 2012). Silodosin likely increases sexual adverse events (RR 6.05, 95% CI 3.55 to 10.31; I2 = 27%). Silodosin would result in 141 more sexual adverse events per 1000 men (95% CI 71 more to 261 more). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 2.6).

2.6. Analysis.

2.6

Comparison 2 Silodosin versus tamsulosin, Outcome 6 Sexual adverse events (short term).

Subgroup analysis

We found one cross‐over trial that reported subgroup analyses according to age (70 years or older) and symptom severity (IPSS 20 or more) (Watanabe 2011). We were unable to perform preplanned subgroup analysis due to lack of information in within‐study comparisons.

Sensitivity analysis

We rated only one study (Chapple 2011) as low risk of bias and were unable to perform a sensitivity analysis.

3. Silodosin versus naftopidil

3.1. Urologic symptom scores

We included five RCTs (Masuda 2012; Matsukawa 2016; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) with 652 participants in the analysis (silodosin 325, naftopidil 327). We used final value in one study (Masuda 2012) and change from baseline in the remaining studies. Silodosin may result in little to no difference in urologic symptom scores (MD ‐0.85, 95% CI ‐2.57 to 0.87; I2 = 84%). We rated the quality of the evidence as low, downgrading for study limitations and inconsistency (Analysis 3.1).

3.1. Analysis.

3.1

Comparison 3 Silodosin versus naftopidil, Outcome 1 Urologic symptom scores (short term).

3.2. Quality of life

We included five RCTs (Masuda 2012; Matsukawa 2016; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) with 652 participants in the analysis (silodosin 325, naftopidil 327). We used the final values in one study (Masuda 2012) and change from baseline in the remaining studies. Silodosin may result in little to no difference in quality of life (MD ‐0.17, 95% CI ‐0.60 to 0.27; I2 = 91%). We rated the quality of the evidence as low, downgrading for study limitations and inconsistency (Analysis 3.2).

3.2. Analysis.

3.2

Comparison 3 Silodosin versus naftopidil, Outcome 2 Quality of life (short term).

3.3. Treatment withdrawal for any reason

We included four RCTs (Matsukawa 2016; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) with 659 participants in the analysis (silodosin 334, naftopidil 325). Silodosin may result in little to no difference in treatment withdrawal for any reason (RR 1.25, 95% CI 0.81 to 1.93; I2 = 0%). We rated the quality of the evidence as low, downgrading for study limitations and imprecision (Analysis 3.3).

3.3. Analysis.

3.3

Comparison 3 Silodosin versus naftopidil, Outcome 3 Treatment withdrawal due to any reason (short term).

3.4. Treament withdrawal due to adverse events

We included five RCTs (Masuda 2012; Matsukawa 2016; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) with 738 participants in the analysis (silodosin 372, naftopidil 366). For the cross‐over trial Masuda 2012, we used only the number of participants who had been initially randomized due to lack of information about the number of participants in the analysis. Silodosin may result in little to no difference in treatment withdrawal due to adverse events (RR 1.38, 95% CI 0.66 to 2.89; I2 = 0%). We rated the quality of the evidence as low downgrading for study limitations and imprecision (Analysis 3.4).

3.4. Analysis.

3.4

Comparison 3 Silodosin versus naftopidil, Outcome 4 Treatment withdrawal due to adverse events (short term).

3.5. AUR

We included two RCTs (Yamaguchi 2013; Yokoyama 2011) with 180 participants in the analysis (silodosin 94, naftopidil 86). We found no event for AUR in either study group.

3.6. Surgical intervention for LUTS/BPH

We included two RCTs (Yamaguchi 2013; Yokoyama 2011) with 180 participants in the analysis (silodosin 94, naftopidil 86). We found no event for surgical intervention for LUTS/BPH in either study group.

3.7. Cardiovascular adverse events

We included five RCTs (Masuda 2012; Matsukawa 2016; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) with 808 participants in the analysis (silodosin 411, naftopidil 397). For cross‐over trials, we took all measurements from silodosin periods and all measurements from tamsulosin periods in Masuda 2012. Silodosin may result in little to no difference in cardiovascular adverse events (RR 1.02, 95% CI 0.41 to 2.56; I2 = 0%). We rated the quality of the evidence as low downgrading for study limitations and imprecision (Analysis 3.5).

3.5. Analysis.

3.5

Comparison 3 Silodosin versus naftopidil, Outcome 5 Cardiovascular adverse events (short term).

3.8. Sexual adverse events

We included four RCTs (Masuda 2012; Shirakawa 2013; Yamaguchi 2013; Yokoyama 2011) with 405 participants in the analysis (silodosin 206, naftopidil 199). For cross‐over trials, we took all measurements from silodosin periods and all measurements from tamsulosin periods in Masuda 2012. Silodosin likely increases sexual adverse events (RR 5.93, 95% CI 2.16 to 16.29; I2 = 0%). Silodosin would result in 74 more sexual adverse events per 1000 men (95% CI 17 more to 231 more). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 3.6).

3.6. Analysis.

3.6

Comparison 3 Silodosin versus naftopidil, Outcome 6 Sexual adverse events (short term).

Subgroup analysis

We were unable to perform any subgroup analyses because there were no relevant data in the included studies.

Sensitivity analysis

We rated all of the included studies as high or unclear risk of bias and were unable to perform a sensitivity analysis.

4. Silodosin versus alfuzosin

4.1. Urologic symptom scores

We included one RCT (Manjunatha 2016a) with 60 participants in the analysis (silodosin 30, alfuzosin 30). Silodosin may or may not result in a clinically important increase in urologic symptom scores (MD 3.83, 95% CI 0.12 to 7.54). We rated the quality of the evidence as low, downgrading for study limitations and imprecision (Analysis 4.1).

4.1. Analysis.

4.1

Comparison 4 Silodosin versus alfuzosin, Outcome 1 Urologic symptom scores (short term).

4.2. Quality of life

We included one RCT (Manjunatha 2016a) with 60 participants in the analysis (silodosin 30, alfuzosin 30). Silodosin likely results in little to no difference in quality of life (MD 0.14, 95% CI ‐0.46 to 0.74). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 4.2).

4.2. Analysis.

4.2

Comparison 4 Silodosin versus alfuzosin, Outcome 2 Quality of life (short term).

4.3. Treatment withdrawal for any reason

We included one RCT (Manjunatha 2016a) with 60 participants in the analysis (silodosin 30, alfuzosin 30). We found no event in treatment withdrawal for any reason in either study group.

4.4. Treatment withdrawal due to adverse events

We included one RCT (Manjunatha 2016a) with 60 participants in the analysis (silodosin 30, alfuzosin 30). We found no event in treatment withdrawal due to adverse events in either study group.

4.5. AUR

We did not find any study that reported this outcome.

4.6. Surgical intervention for LUTS/BPH

We did not find any study that reported this outcome.

4.7. Cardiovascular adverse events

We included one RCT (Manjunatha 2016a) with 60 participants in the analysis (silodosin 30, alfuzosin 30). Silodosin may result in little to no difference in cardiovascular adverse events (RR 0.67, 95% CI 0.36 to 1.24). We rated the quality of the evidence as low, downgrading for study limitations and imprecision (Analysis 4.3).

4.3. Analysis.

4.3

Comparison 4 Silodosin versus alfuzosin, Outcome 3 Cardiovascular adverse events (short term).

4.8. Sexual adverse events

We included one RCT (Jung 2012) with 95 participants in the analysis (silodosin 48, alfuzosin 47). Silodosin likely increases sexual adverse events (RR 37.21, 95% CI 5.32 to 260.07). Assuming a baseline risk of 0.6% (Van Kerrebroec 2002), silodosin would result in 217 more sexual adverse events per 1000 men (95% CI 26 more to 1000 more). We rated the quality of the evidence as moderate, downgrading for study limitations (Analysis 4.4).

4.4. Analysis.

4.4

Comparison 4 Silodosin versus alfuzosin, Outcome 4 Sexual adverse events (short term).

Subgroup analysis and sensitivity analysis

We were unable to perform secondary analyses because there were no relevant data or too few data in the included studies.

Discussion

Summary of main results

We included 19 unique studies with 4295 randomized participants across four comparisons for short‐term follow‐up. All studies included men aged over 40 years. The mean age and IPSS of the participants were 66.5 years and 19.1, respectively.

Silodosin versus placebo

Silodosin may reduce urologic symptom scores in an appreciable number of men but not improve quality of life appreciably. Treatment withdrawals for any reason do not appear increased. We are uncertain about the effect of silodosin on cardiovascular adverse events but silodosin likely increases sexual adverse events. There were no clinically important changes in treatment effects in a sensitivity analysis focused on low risk of bias studies.

Silodosin versus tamsulosin

Silodosin may result in little to no differences in urologic symptom scores and quality of life. We are uncertain about rates of treatment withdrawal for any reason. Silodosin may result in little to no difference in cardiovascular adverse events but likely increases sexual adverse events substantially.

Silodosin versus naftopidil

Silodosin may have similar effects in urologic symptom scores, quality of life, rates of treatment withdrawal for any reason and cardiovascular adverse events. Silodosin likely increases sexual adverse events substantially.

Silodosin versus alfuzosin

Silodosin may or may not result in a clinically important increase in urologic symptom scores and likely results in little to no difference in quality of life. We found no event in treatment withdrawal for any reason. Silodosin may not reduce cardiovascular adverse events but likely increases sexual adverse events substantially.

We were unable to perform any of the predefined subgroup analyses.

Overall completeness and applicability of evidence

There were a several limitations that related to overall completeness and applicability of evidence that deserve consideration.

  • Our ability to assess the longer‐term outcomes of silodosin compared to other drugs was limited given that follow‐up duration in almost all studies (18 of 19) was 12 weeks or less. Given the long‐term use of ABs for the treatment of LUTS/BPH in clinical practice, examining the efficacy and safety of silodosin over such a short term appears insufficient to provide assurance of long‐term outcomes.

  • Three of the larger trials included in this review used pre‐randomization run‐in periods (Chapple 2011; Marks 2009; Yu 2011). This may have led to the exclusion of men who were less likely to be compliant or more prone to experiencing adverse events, thereby limiting generalizability.

  • We were unable to determine the effect of silodosin on AUR or surgical interventions due to the lack of events in the included studies. This once again relates to the short follow‐up period. Additional information may have to be drawn from longer‐term observational studies.

  • Rates of adverse events in two studies available in abstract form only were higher than those reported in full‐text studies, raising concern about publication bias (Jung 2012; Yamanishi 2011).

  • Although ABs such as silodosin are commonly used in combination with 5‐ARIs, we did not find any eligible studies.

Quality of the evidence

We consistently downgraded the quality of the evidence by one or two steps to moderate or low. The most common reasons for downgrading were study limitations (issues surrounding allocation concealment and blinding), inconsistency (as reflected by high I2‐values), which we were unable to explain through secondary analyses, and imprecision (wide confidence intervals that crossed the assumed threshold of clinically important difference or due to few events).

Potential biases in the review process

Despite a comprehensive search strategy without any publication or language restrictions, it is possible that we may have missed relevant publications. A majority of studies originated from Japan and India. It is possible that some, in particular negative studies were published in non‐indexed journals or presented at local meetings only and therefore may have escaped our search, resulting in potential publication bias. The number of studies included in this review were insufficient to generate funnel plots; therefore, the risk of publication bias may have been underestimated. A future update of this review should consider the inclusion of regional bibliographic databases to further enhance the search.

We attempted to contact all the study authors on several occasions seeking feedback. While we received additional information from nine study authors, others did not reply. This may represent a source of bias.

For cross‐over trials, we tried to extract the data before cross‐over, as if the study were a parallel‐group trial, in accordance with Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011c). However, two cross‐over trials reported adverse events using all measurements from silodosin periods and all measurements from tamsulosin periods (Masuda 2012; Watanabe 2011). This may have given rise to a unit of analysis error. For other cross‐over trials, we used the number of participants initially randomized as a more conservative approach. In addition, one cross‐over trial used the event rate to report individual adverse events, which may also result in a unit of analysis error (double counting) (Masuda 2012).

Agreements and disagreements with other studies or reviews

To date, no published systematic review exists that focuses on silodosin and has applied the same rigorous methodology as this Cochrane Review, which includes a published protocol, a focus on patient‐important outcomes, an exhaustive literature search and an assessment of the quality of the evidence using GRADE.

In a recently reported systematic review and meta‐analysis, a variety of ABs including silodosin were found to have a similar efficacy compared to placebo but were associated with significantly higher incidences of total adverse events and treatment withdrawal (Novara 2013; Yuan 2015). On the other hand, Ding 2013 reported no significant difference in the incidence of ejaculation disorder between the silodosin and tamsulosin groups, but we are concerned that the numbers used in the meta‐analysis were not accurate. A systematic review of naftopidil reported that silodosin had comparable efficacy with naftopidil but was associated with a higher incidence of sexual adverse events (Castiglione 2014). However, no meta‐analysis was reported and the authors did not rate the quality of the evidence. Collectively, the results of the present review are largely consistent with those of previously published reviews compared to placebo, tamsulosin, and naftopidil.

We did not find any published systematic review comparing silodosin to alfuzosin: this review is the first.

Authors' conclusions

Implications for practice.

Based on our findings, the efficacy of silodosin appears to be similar to other ABs including tamsulosin, naftopidil, and alfuzosin but it is associated with increased sexual adverse effects, specifically abnormal ejaculation, which may be an issue for sexually active men.

Implications for research.

The major shortcoming of the current evidence base for silodosin is the short time‐horizon of most studies, limiting our understanding of the long‐term effectiveness and safety. Given that ABs such as silodosin are used for extended periods of time, typically years, longer‐term trials should be performed (and could be mandated by regulatory agencies). In their absence, observational studies may help inform this issue, however, based on a recent systematic review performed by an Evidence‐Based Practice Center in the United States few informative observational studies exist (Brasure 2016). There is also a consistent need for greater methodological rigor and reporting transparency.

Notes

We have based parts of the Methods section of this protocol on a standard template developed by the Cochrane Metabolic and Endocrine Disorders Group, which has been modified and adapted for use by Cochrane Urology.

Acknowledgements

We acknowledge the support received from the authors of included studies, Christopher R. Chapple, Gyung‐woo Jung, Leonard S. Marks, Satabdi Pande, Hideki Takeshita, Kenya Yamaguchi, Tomonori Yamanishi, and Teruhiko Yokoyama by providing additional data. We are very grateful to Christopher Filson, Herney Andrés García‐Perdomo, Hong Wook Kim, Stavros Gravas, and Valter Silva for having served as peer reviewers. We thank Cochrane Urology and our contact editors Mari Imamura and Muhammad Imran Omar for supporting this review.

Appendices

Appendix 1. Search strategies

Cochrane Library (via Wiley) search strategy
1. "Prostatic Hyperplasia" [Mesh]
2. (hyperplasia NEAR/3 prostat*):ti,ab,kw
3. "hyperplasia of the prostate":ti,ab,kw
4. "prostatic hyperplasia":ab,ti
5. (hypertrophy NEAR/3 prostat*):ti,ab,kw
6. (adenoma* NEAR/3 prostat*):ti,ab,kw
7. "lower urinary tract symptom" [Mesh]
8. "lower urinary tract": ti,ab,kw
9. LUTS: ti,ab,kw
10. "prostatism": ti,ab,kw
11. "prostatism"[Mesh]
12. "Urinary Bladder Neck Obstruction" [Mesh]
13. "bladder outlet obstruction": ti,ab,kw
14. (prostat* NEAR/3 enlarg*): ti,ab,kw
15. 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12 or 13 or 14
16. silodosin: ti,ab,kw
17. "KMD‐3213": ti,ab,kw
18. 16 or 17
MEDLINE (via Pubmed) search strategy
1. "Prostatic Hyperplasia" [Mesh] or prostatic hyperplasia [TIAB]
2. "Lower Urinary Tract Symptoms" [Mesh] or lower urinary tract [TIAB] or LUTS [TIAB]
3. hyperplasia of the prostate [TIAB]
4. "Prostatic hypertrophy" [TIAB]
5. "Prostatic adenoma" [TIAB]
6. "Prostatism" [Mesh] or Prostatism [TIAB]
7. "Urinary Bladder Neck Obstruction" [Mesh] or "Urinary Bladder Neck Obstruction" [TIAB]
8. bladder outlet obstruction [TIAB]
9. 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8
10. silodosin [Supplementary Concept]
11. silodosin [TIAB]
12. KMD‐3213 [TIAB]
13. rapaflo [TIAB]
14. urief [TIAB]
15. urorec [TIAB]
16. 10 or 11 or 12 or 13 or 14 or 15
17. randomized controlled trial [PT]
18. controlled clinical trial [PT]
19. randomized [TIAB]
20. placebo [TIAB]
21. drug therapy [Sh]
22. randomly [TIAB]
23. trial [TIAB]
24. groups [TIAB]
25. 17 or 18 or 19 or 20 or 21 or 22 or 23 or 24
26. 9 AND 16 AND 25
27. (animals [Mesh]) NOT (humans [Mesh] and animals [Mesh])
28. 26 NOT 27
EMBASE (via Elsevier) search strategy
1. 'prostate hypertrophy'/exp
2. (hyper* NEAR/3 prostat*):ab,ti
3. (adenoma* NEAR/3 prostat*):ab,ti
4. 'BPH' or 'BPO' or 'BPE':ab,ti
5. 'lower urinary tract symptom'/exp
6. 'lower urinary tract' or 'LUTS':ab,ti
7. 'prostatism':ab,ti
8. 'prostatism'/exp
9. 'bladder obstruction'/exp
10. ('bladder* NEAR/3 obstruct*)' or 'BOO' :ab,ti
11. (prostat* NEAR/3 (enlarg* or obstruct*)):ab,ti
12. ((urinary or urethra* or urination or LUT*) NEAR/3 (symptom* or complain*)):ab,ti
13. 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12
14. 'silodosin'/exp
15. silodosin or 'KMD‐3213' or kad3213 or rapaflo or silodyx or urief or urorec or Silofast or Thrupas:tw
16. '160970‐54‐7' or '160970‐64‐9' or '169107‐04‐4':rn
17. 14 or 15 or 16
18. random$ or factorial$ or crossover$ or cross over$ or 'cross‐over$' or placebo$ or (doubl$ adj blind$) or (singl$ adj blind$) or assign$ or allocat$ or volunteer$:tw
19. 'crossover procedure'/exp OR 'double blind procedure'/exp OR 'randomized controlled trial'/exp OR 'single blind procedure'/exp
20. 18 or 19
21. 13 AND 17 AND 20
22. ('animals'/exp) NOT ('humans'/exp and 'animals'/exp)
23. 21 NOT 22
Scopus search strategy
1. TITLE‐ABS‐KEY (hyperplasia W/3 prostat*)
2. TITLE‐ABS‐KEY (hypertrophy W/3 prostat*)
3. TITLE‐ABS‐KEY (adenoma* W/3 prostat*)
4. TITLE‐ABS‐KEY (prostat* W/3 (enlarg* or obstruct*))
5. TITLE‐ABS‐KEY ((urinary OR urethra* OR urination OR lut*) W/3 (symptom* OR complain*))
6. TITLE‐ABS‐KEY (prostatism)
7. TITLE‐ABS‐KEY (bladder* W/3 obstruct*)
8. TITLE‐ABS‐KEY ("lower urinary tract")
9. 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8
10. TITLE‐ABS‐KEY (silodosin or 'KMD‐3213' or rapaflo or silodyx or urief or urorec or Silofast or Thrupas)
11. CASREGNUMBER (160970‐54‐7 OR 160970‐64‐9 OR 169107‐04‐4)
12. 10 or 11
13. TITLE‐ABS‐KEY ("Double blind*" OR "Single Blind*" OR "Cross over stud*")
14. 9 AND 12 AND 13
Google Scholar search strategy
allintitle: ("Prostatic Hyperplasia" OR "prostate hypertrophy" OR "hyperplasia of the prostate" OR "lower urinary tract" OR "LUTS" OR prostatism OR "bladder obstruction" OR "bladder outlet obstruction") AND (silodosin OR KMD‐3213 OR rapaflo OR Silodyx OR kad3213 OR urief OR urorec OR Silodal OR Rapilif OR Silofast OR Thrupas)
Web of Science search strategy
1. TS = (hyper* NEAR/3 prostat*) OR (adenoma* NEAR/3 prostat*) OR "BPH" OR "BPO" OR "BPE" OR "lower urinary tract" OR "luts" OR prostatism OR ("bladder NEAR/3 obstruct*") OR "BOO" OR (prostat* NEAR/3 (enlarg* or obstruct*)) OR ((urinary or urethra* or urination or LUT*) NEAR/3 (symptom* or complain*))
2. TS = (silodosin OR KMD‐3213 OR rapaflo OR urief)
3. TS = "clinical trial*" OR "controlled trial*" OR "randomized controlled trial*" OR (single blind*) OR (double blind*)
4. 1 AND 2 AND 3
ClinicalTrials.gov
Basic search = ("Prostatic Hyperplasia" OR "prostate hypertrophy" OR "hyperplasia of the prostate" OR "lower urinary tract" OR "LUTS" OR prostatism OR "bladder obstruction" OR "bladder outlet obstruction") AND (silodosin OR KMD‐3213 OR rapaflo OR urief OR Thrupas)
World Health Organization International Clinical Trials Registry Platform Search Portal
In the title = ("Prostatic Hyperplasia" OR "prostate hypertrophy" OR "hyperplasia of the prostate" OR "lower urinary tract" OR "LUTS" OR prostatism OR "bladder obstruction" OR "bladder outlet obstruction") AND In the intervention= (silodosin OR KMD‐3213 OR rapaflo OR Silodyx OR urief OR urorec OR Silodal OR Thrupas)

Appendix 2. Survey of trial investigators providing information on included trials

  Date trial author contacted (first) Date trial author provided data (latest) Data trial author provided
(short summary)
Chapple 2011 22 Jan 2017 2 Feb 2017 Unadjusted change from baseline in IPSS and QoL, mean and standard deviation of IPSS and QoL at endpoint (12 weeks), the number of participants with acute urinary retention and surgical intervention.
Jung 2012 18 Jan 2017 28 Feb 2017 Author reply: "Full text was not published".
Marks 2009 23 Jan 2017 23 Jan 2017 Author reply: "Pharmaceutics only have a data". We cannot contact study supporter in pharmaceutics due to wrong email address.
Pande 2014 24 Jan 2017 18 May 2017 Mean and standard deviation of IPSS and QoL at baseline and endpoint (12 weeks), the number of participants with acute urinary retention, surgical intervention, cardiovascular adverse events, and sexual adverse events.
Takeshita 2016 24 Oct 2017 3 Nov 2017 Change from baseline to 4 weeks (before cross‐over) IPSS and QoL (mean and standard deviation). Treatment withdrawal and adverse events at 4 weeks (before cross‐over). Method of random sequence generation and allocation concealment.
Yamaguchi 2013 27 Jan 2017 1 Feb 2017 Change from baseline in IPSS and QoL (mean and standard deviation), the number of participants with treatment withdrawal, acute urinary retention, surgical intervention, and cardiovascular adverse events. Method of random sequence generation and blinding.
Yamanishi 2011 27 Jan 2017 31 Jan 2017 Author reply: "Full text was not published".
Yokoyama 2011 30 Jan 2017 13 March 2017 Dates when study was conducted, study duration (intervention), exclusion criteria, conflicts of interest, mean and standard deviation of IPSS and QoL at baseline and endpoint (12 weeks), the number of participants with acute urinary retention and surgical intervention. Method of random sequence generation and blinding.
Yokoyama 2012 24 Oct 2017 29 Oct 2017 Total number of participants who were analyzed and finishing the trial at 3 months (before cross‐over), change from baseline to 3 months (before cross‐over) in IPSS and QoL (mean and standard deviation), Treatment withdrawal at 3 months (before cross‐over). Method of random sequence generation and blinding. Presence of published protocol.

Footnotes

IPSS: International Prostate Symptom Score; QoL: quality of life

Data and analyses

Comparison 1. Silodosin versus placebo.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1 Urologic symptom scores (short term) 3 1743 Mean Difference (IV, Random, 95% CI) ‐2.65 [‐3.23, ‐2.08]
2 Quality of life (short term) 2 820 Mean Difference (IV, Random, 95% CI) ‐0.42 [‐0.71, ‐0.13]
3 Treatment withdrawal due to any reason (short term) 3 1703 Risk Ratio (M‐H, Random, 95% CI) 1.08 [0.70, 1.66]
4 Treatment withdrawal due to adverse events (short term) 4 1967 Risk Ratio (M‐H, Random, 95% CI) 2.29 [1.41, 3.72]
5 Cardiovascular adverse events (short term) 4 1967 Risk Ratio (M‐H, Random, 95% CI) 1.28 [0.67, 2.45]
6 Sexual adverse events (short term) 4 1967 Risk Ratio (M‐H, Random, 95% CI) 26.07 [12.36, 54.97]

Comparison 2. Silodosin versus tamsulosin.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1 Urologic symptom scores (short term) 10 1708 Mean Difference (IV, Random, 95% CI) ‐0.04 [‐1.31, 1.24]
2 Quality of life (short term) 10 1707 Mean Difference (IV, Random, 95% CI) ‐0.15 [‐0.53, 0.22]
3 Treatment withdrawal due to any reason (short term) 10 1573 Risk Ratio (M‐H, Random, 95% CI) 1.02 [0.62, 1.69]
4 Treatment withdrawal due to adverse events (short term) 9 1572 Risk Ratio (M‐H, Random, 95% CI) 1.96 [1.12, 3.44]
5 Cardiovascular adverse events (short term) 11 1955 Risk Ratio (M‐H, Random, 95% CI) 0.77 [0.53, 1.12]
6 Sexual adverse events (short term) 10 1849 Risk Ratio (M‐H, Random, 95% CI) 6.05 [3.55, 10.31]

Comparison 3. Silodosin versus naftopidil.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1 Urologic symptom scores (short term) 5 652 Mean Difference (IV, Random, 95% CI) ‐0.85 [‐2.57, 0.87]
2 Quality of life (short term) 5 652 Mean Difference (IV, Random, 95% CI) ‐0.17 [‐0.60, 0.27]
3 Treatment withdrawal due to any reason (short term) 4 659 Risk Ratio (M‐H, Random, 95% CI) 1.25 [0.81, 1.93]
4 Treatment withdrawal due to adverse events (short term) 5 738 Risk Ratio (M‐H, Random, 95% CI) 1.38 [0.66, 2.89]
5 Cardiovascular adverse events (short term) 5 808 Risk Ratio (M‐H, Random, 95% CI) 1.02 [0.41, 2.56]
6 Sexual adverse events (short term) 4 405 Risk Ratio (M‐H, Random, 95% CI) 5.93 [2.16, 16.29]

Comparison 4. Silodosin versus alfuzosin.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1 Urologic symptom scores (short term) 1 60 Mean Difference (IV, Random, 95% CI) 3.83 [0.12, 7.54]
2 Quality of life (short term) 1 60 Mean Difference (IV, Random, 95% CI) 0.14 [‐0.46, 0.74]
3 Cardiovascular adverse events (short term) 1 60 Risk Ratio (M‐H, Random, 95% CI) 0.67 [0.36, 1.24]
4 Sexual adverse events (short term) 1 95 Risk Ratio (M‐H, Random, 95% CI) 37.21 [5.32, 260.07]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Chapple 2011.

Methods Study design: double‐blind, placebo‐ and active‐controlled, parallel‐group clinical study
Setting/Country: 72 hospital clinics and inpatient units in 11 countries/Europe
Dates when study was conducted: May 2006‐May 2007
Participants Inclusion criteria: men ≥ 50 years with LUTS (defined by a stable IPSS total score ≥ 13 points), bladder outlet obstruction (defined by a Qmax 4 mL/s‐15 mL/s, with a minimum voided volume of ≥ 125 mL), and the evidence of satisfactory compliance with study medication (80%–120% during the placebo run‐in period)
Exclusion criteria: men with improvement in the IPSS total score ≥ 25% in the run‐in period, PVR ≥ 250 mL, intravesical obstruction from any cause other than BPH, history of any procedure considered an intervention for BPH, active urinary tract infection or history of recurrent urinary tract infections, current prostatitis or diagnosis of chronic prostatitis, history of prostate or invasive bladder cancer, significant postural hypotension, use of 5‐ARIs within 6 months, or use of an a‐blocker or phytotherapy within 2 weeks before entry
Total number of participants randomly assigned: 955
Group A (Silodosin)
  • number of all participants randomly assigned: 381

  • age (years): 65.8 ± 7.70

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: 19.1 ± 4.2

  • Qmax (mL/s): 10.78 ± 2.726


Group B (Tamsulosin)
  • number of all participants randomly assigned: 384

  • age (years): 65.9 ± 7.41

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: 18.9 ± 4.3

  • Qmax (mL/s): 10.27 ± 2.726


Group C (Placebo)
  • number of all participants randomly assigned: 190

  • age (years): 66.0 ± 7.37

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: 19.3 ± 4.3

  • Qmax (mL/s): 10.32 ± 2.816

Interventions Run‐in period: 4 weeks, single‐blind, placebo run‐in period
Group A: silodosin 8 mg once daily
Group B: tamsulosin 0.4 mg once daily
Group C: placebo once daily
Duration: 12 weeks
Outcomes Primary outcome
  • change from baseline in the total score (questions 1–7) of the IPSS


How measured: IPSS questionnaire
Time points measured: at screening, at baseline, and after 7, 14, 28, 56, and 84 d of treatment
Time points reported: baseline and end of study
Secondary outcome
  • IPSS storage and voiding symptoms subscores

  • QoL

  • Qmax

  • percentage of treatment responders by IPSS (decrease from baseline ≥ 25%) and by Qmax (increase from baseline ≥ 30%)


How measured: IPSS questionnaire, uroflowmetry
Time points measured: IPSS: see primary outcome/Qmax: at the same time periods as IPSS
Time points reported: baseline and end of study
Safety outcomes
How measured: adverse events, physical examination (BP, heart rate), laboratory tests, and a 12‐lead electrocardiogram
Time points measured: at each visit
Time points reported: at each visit
Subgroup: a post hoc analysis was conducted in the subgroup of participants with nocturia at baseline (defined as at least two voids per night), as assessed by question 7 of the IPSS
Funding sources Recordati Ireland Ltd
Declarations of interest Financial disclosures: I certify that all conflicts of interest, including specific financial interests and relationships and affiliations relevant to the subject matter or materials discussed in the manuscript (eg, employment/affiliation, grants or funding, consultancies, honoraria, stock ownership or options, expert testimony, royalties, or patents filed, received, or pending), are the following: Christopher R. Chapple and Francesco Montorsi are consultants and researchers for Recordati. Teuvo L.J. Tammela, Manfred Wirth, Evert Koldewijn, and Eldiberto Fernandez Fernandez are researchers for Recordati
Notes Language of publication: English
Data (unadjusted IPSS and IPSS‐QoL, AUR, and surgical intervention) were given by contact with study author
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "randomly assigned (in a ratio of 2:2:1, with stratification by center, with blocks of five assigned to each center, produced and managed centrally by an international contract research organization)"
Allocation concealment (selection bias) Low risk Quote: "managed centrally by an international contract research organization"
Blinding of participants and personnel (performance bias) 
 All outcomes Low risk Quote: "A multicenter double‐blind, placebo‐ and active‐controlled parallel group clinical study, all study personnel and participants were blinded to treatment assignment for the entire duration of the study"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Low risk Judgement: 10/381 (2.6%) in silodosin, 8/384 (2.1%) in tamsulosin, and 5/190 (2.6%) in placebo group were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) High risk Judgement: Protocol (NCT00359905) was revised after completion of study (2009) and percentage of treatment responders was added as secondary outcome.
All primary and secondary endpoints were measured at 7, 14, 28, 56, 84 day, but only reported at 12 weeks
Unadjusted data for analysis for IPSS and QoL were not reported
Other bias Unclear risk Judgement: 4 weeks' placebo run‐in period

Jung 2012.

Methods Study design: open‐label, randomized comparative study
Setting/Country: not reported/South Korea
Dates when study was conducted: not reported
Participants Inclusion criteria: sexually active men with BPH
Exclusion criteria: not reported
Total number of participants randomly assigned: 138
Group A (Silodosin)
  • number of all participants randomly assigned: 48

  • age (years): not reported

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported


Group B (Tamsulosin)
  • number of all participants randomly assigned: 43

  • age (years): not reported

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported


Group C (Alfusocin)
  • number of all participants randomly assigned: 47

  • age (years): not reported

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported

Interventions Run‐in period: none
Group A: silodosin 8 mg once daily
Group B: tamsulosin 0.2 mg once daily
Group C: alfuzosin 10 mg once daily
Duration: 4 weeks
Outcomes
  • IPSS

  • Male Sexual Health Questionnaire (MSHQ) evaluating erection, ejaculation and sexual satisfaction

  • the percentage of participants who showed decrease in ejaculate volume or anejaculation


How measured: IPSS, MSHQ
Time points measured: baseline and on the 4th week of medication
Time points reported: baseline and on the 4th week of medication
Subgroup: none
Funding sources None
Declarations of interest Not reported
Notes Language of publication: English
Publication status: abstract (full text has not been published; study author reply)
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Quote: "open‐label, randomized comparative study"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Quote: "open‐label, randomized comparative study"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: the number of participants excluded in analysis was not reported
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: the number of participants excluded in analysis was not reported
Selective reporting (reporting bias) Unclear risk Judgement: protocol was not published and no information related to the review outcomes given
Other bias Unclear risk Judgement: abstract only

Kawabe 2006a.

Methods Study design: randomized, double‐blind, placebo‐controlled study
Setting/Country: 88 centers/outpatient/Japan
Dates when study was conducted: not reported
Participants Inclusion criteria: men ≥ 50 years with total IPSS of ≥ 8, an associated QoL score of ≥ 3, prostate volume (measured by transabdominal or transrectal US) of ≥ 20 mL, a Q max of < 15 mL/s with a voided volume of ≥ 100 mL and a PVR of < 100 mL
Exclusion criteria: men who had antiandrogen preparations for 1 year before the study or had a prostatectomy, intrapelvic radiation therapy or prostatic hyperthermia (transurethral microwave hyperthermia or transurethral needle ablation). Men who had prostate cancer or suspected prostate cancer, neurogenic bladder, bladder neck constriction, urethral stricture, bladder calculus, severe bladder diverticulum, active urinary tract infection requiring medical treatment, renal impairment (serum creatinine ≥ 2.0 mg/dL) and other complications considered likely to affect micturition, severe hepatic disorders, severe cardiovascular disease and a history of orthostatic hypotension
Total number of participants randomly assigned: 457
Group A (Silodosin)
  • number of all participants randomly assigned: 176

  • age (years): 65.4 ± 7.0

  • prostate volume (mL): 36.0 ± 16.9

  • PSA (ng/mL): not reported

  • IPSS: 17.1 ± 5.7

  • Qmax (mL/s): 9.89 ± 2.72


Group B (Tamsulosin)
  • number of all participants randomly assigned: 192

  • age (years): 65.6 ± 7.0

  • prostate volume (mL): 35.7 ± 14.4

  • PSA (ng/mL): not reported

  • IPSS: 17.0 ± 5.7

  • Qmax (mL/s): 9.43 ± 2.79


Group C (Placebo)
  • number of all participants randomly assigned: 89

  • age (years): 65.0 ± 6.9

  • prostate volume (mL): 35.2 ± 16.0

  • PSA (ng/mL): not reported

  • IPSS: 17.1 ± 6.1

  • Qmax (mL/s): 9.96 ± 2.65

Interventions Run‐in period: none
Group A: oral silodosin 4 mg twice daily
Group B: oral tamsulosin 0.2 mg once daily
Group C: oral placebo twice daily
Duration: 12 weeks
Outcomes Primary outcome
  • change in the total IPSS from baseline


How measured: IPSS questionnaire
Time points measured: at the end of wash‐out period and at 1, 2, 4, 8 and 12 weeks during the treatment period
Time points reported: at the end of wash‐out period and at 1, 2, and 12 weeks during the treatment period
Secondary outcome
  • change in Qmax and urodynamics

  • IPSS voiding and storage scores and QoL score


How measured: IPSS questionnaire, uroflowmetry
Time points measured: at the end of wash‐out period and at 1, 2, 4, 8 and 12 weeks during the treatment period
Time points reported: at the end of wash‐out period and 12 weeks
Safety outcomes
How measured: adverse events, physical examinations (BP and heart rate), clinical laboratory tests (hematology, blood chemistry and urine analysis)
Time points measured: at the start of the observation period and at 4 and 12 weeks of treatment (follow‐up visit)
Time points reported: at the start of the observation period and at 12 weeks of treatment
Subgroup: analysis based on LUTS severity, post hoc investigation in Qmax among the 3 treatment groups in the overall subgroup of participants with a change in voided volume of < 50% before and after treatment
Funding sources None
Declarations of interest None
Notes Language of publication: English
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "randomized, double‐blind, placebo controlled study"
Judgement: the randomization method was not defined
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Unclear risk Quote: "randomized, double‐blind, placebo controlled study"
Judgement: placebo‐controlled study, but double‐blind was not clear who was blinded
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Quote: "randomized, double‐blind, placebo controlled study"
Judgement: double‐blind was not clear who was blinded
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Low risk Judgement: 1/176 (0.5%) in silodosin group, 0/192 (0%) in tamsulosin, and 0/89 (0%) were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: 1/176 (0.5%) in silodosin group, 0/192 (0%) in tamsulosin, and 0/89 (0%) were not included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: 1/176 (0.5%) in silodosin group, 0/192 (0%) in tamsulosin, and 0/89 (0%) were not included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: 1/176 (0.5%) in silodosin group, 0/192 (0%) in tamsulosin, and 0/89 (0%) were not included in analysis
Selective reporting (reporting bias) High risk Judgement: protocol was not published and additional post hoc subgroup investigation was not defined in method section
Other bias High risk Judgement: baseline imbalance in QoL; primary endpoint was adjusted analysis by baseline QoL. Drug administration times were different between groups

Manjunatha 2016a.

Methods Study design: prospective, randomized, comparative, open‐label study
Setting/Country: tertiary care hospital/India
Dates when study was conducted: June 2013‐June 2014
Participants Inclusion criteria: men ≥ 45 years with symptomatic BPH with IPSS of ≥ 8, QoL of ≥ 3, and Qmax of < 15 mL/s, but > 4 mL/s with a voided volume of > 100 mL
Exclusion criteria: severe hepatic or renal insufficiency, urinary tract infections, urethral stricture, neurogenic bladder, PSA ≥ 5 ng/mL, history of urethral or prostatic surgery, hypotension or severe untreated hypertension, history of esophageal or intestinal obstruction, history of multiple drug abuse, significant psychiatric problems, serious disease or malignancy, increased risk of QTc (corrected QT Interval in the heart's electrical cycle) prolongation (e.g. hypokalemia, concomitant use of Class Ia and III antiarrhythmics, antipsychotics, tricyclic antidepressants, etc.), concomitant use of 5‐ARIs and strong cytochrome P450 3A4 inhibitors, likelihood of requiring catheterization within next 3 months
Total number of participants randomly assigned: 90
Group A (Silodosin)
  • number of all participants randomly assigned: 30

  • age (years): 64.00 ± 11.14

  • prostate volume (mL): 40.57 ± 16.45

  • PSA (ng/mL): 1.89 ± 1.06

  • IPSS: 15.93 ± 6.03

  • Qmax (mL/s): 10.03 ± 3.35


Group B (Tamsulosin)
  • number of all participants randomly assigned: 30

  • age (years): 63.60 ± 9.05

  • prostate volume (mL): 40.33 ± 21.55

  • PSA (ng/mL): 2.21 ± 1.33

  • IPSS: 21.63 ± 7.63

  • Qmax (mL/s): 9.58 ± 3.62


Group C (Alfuzosin)
  • number of all participants randomly assigned: 30

  • age (years): 63.43 ± 8.91

  • prostate volume (mL): 44.43 ± 27.72

  • PSA (ng/mL): 2.29 ± 1.27

  • IPSS: 19.2 ± 9.6

  • Qmax (mL/s): 10.18 ± 3.45

Interventions Run‐in period: none
Group A: silodosin capsule 8 mg once daily in the morning (before breakfast)
Group B: tamsulosin tablet/capsule 0.4 mg (before meals) once daily at bedtime
Group C: alfuzosin sustained release tablet 10 mg (immediately after meals) once daily
Duration: 12 weeks
Outcomes Primary outcome
  • change in total IPSS from baseline


How measured: IPSS questionnaire
Time points measured: baseline, after 2 weeks (visit 1), 4 weeks (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4)
Time points reported: baseline, after 2 weeks (visit 1), 4 weeks (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4)
Secondary outcome
  • change in Qmax

  • subjective symptom scores of IPSS (voiding and storage scores)

  • QoL


How measured: uroflowmetry, IPSS questionnaire
Time points measured: baseline, after 2 weeks (visit 1), 4 weeks (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4)
Time points reported: baseline, after 2 weeks (visit 1), 4 weeks (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4)
Safety outcomes
How measured: side effects, cardiovascular parameters, pulse rate, and BP (supine and standing)
Time points measured: baseline, after 2 weeks (visit 1), 4 weeks (visit 2), 8 weeks (visit 3), and 12 weeks (visit 4)
Time points reported: cumulative results from follow‐up visits
Subgroup: none
Funding sources None
Declarations of interest None
Notes Language of publication: English
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "prospective, randomized".
Judgement: random number table in protocol
Allocation concealment (selection bias) Low risk Judgement: no information in article, but "Sequentially numbered, sealed, opaque envelopes" in protocol
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Judgement: open‐label randomized study
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Judgement: open‐label randomized study
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Low risk Quote: "there were no drop‐outs or protocol violations during the study and all the ninety subjects were included in the analysis"
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Quote: "there were no drop‐outs or protocol violations during the study and all the ninety subjects were included in the analysis"
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Quote: "there were no drop‐outs or protocol violations during the study and all the ninety subjects were included in the analysis"
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Quote: "there were no drop‐outs or protocol violations during the study and all the ninety subjects were included in the analysis"
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: the number of participants with sexual adverse events in tamsulosin and alfuzosin group were not reported
Selective reporting (reporting bias) Low risk Judgement: protocol (Clinical Trials Registry‐India (CTRI/2013/07/003805)) was published and the prespecified outcomes were well described.
Other bias High risk Judgement: probably, baseline imbalance in IPSS score, PSA, PVR (silodosin group has markedly lower) and prostate volume (alfuzosin group has markedly higher prostate volume).

Marks 2009.

Methods Study design: parallel‐group, double‐blind, randomized study
Setting/Country: multicenter/USA
Dates when study was conducted: May 2005‐August 2006
Participants Inclusion criteria: men aged ≥ 50 years with an IPSS of ≥ 13, a Qmax of 4 mL/s‐15 mL /s and a PVR < 250 mL
Exclusion criteria: men with intravesical obstruction unrelated to BPH; bladder calculi; history of or current condition affecting bladder function; prior surgical intervention to relieve BPH or bladder neck obstruction; active urinary tract infection or history of recurrent urinary tract infection within the past 2 years; prostatitis within the past 3 months; BPH‐unrelated urinary retention within the past 3 months; and a history of recurring prostatitis (> 3 times within the past year), prior or current prostate cancer or PSA > 10 ng/mL; prior invasive bladder cancer; bladder catheterization or bladder or prostate instrumentation within the past 30 days and history of or current significant postural hypotension, including changes in systolic (> 30 mm Hg) or diastolic (> 20 mm Hg) BP or heart rate (more than 20 beats per minute), and lightheadedness, fainting, blurred vision, profound weakness or syncope upon change in position
Total number of participants randomly assigned: 923
Group A (Silodosin)
  • number of all participants randomly assigned: 466

  • age (years): 64.6 ± 8.1

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: 21.3 ± 5.1

  • Qmax (mL/s): 8.7 ± 2.6


Group B (Placebo)
  • number of all participants randomly assigned: 457

  • age (years): 64.7 ± 8.1

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: 21.3 ± 4.9

  • Qmax (mL/s): 8.9 ± 2.8

Interventions Run‐in period: single‐blind, placebo run in for 4 weeks
Group A: silodosin 8 mg once daily with breakfast
Group B: placebo once daily with breakfast
Duration: 12 weeks
Outcomes Primary outcome
  • mean change from baseline to week 12 in total IPSS


How measured: IPSS questionnaire
Time points measured: at weeks 0 (baseline), 0.5, 1, 2, 4 and 12
Time points reported: at weeks 0 (baseline), 0.5, 1, 2, 4 and 12
Secondary outcome
  • mean change in Qmax from baseline to week 12


How measured: uroflow assessed by study investigator and a blinded central reader
Time points measured: at baseline, 2‐6 h after the first dose, and at weeks 1, 2, 4 and 12
Time points reported: at baseline, 2‐6 h after the first dose, and at weeks 1, 2, 4 and 12
Safety outcomes
How measured: adverse event/additional safety assessments included 12‐lead electrocardiograms, clinical laboratory tests and vital sign measurements including postural hypotension tests and physical examinations
Time points measured: at every visit except at post‐randomization week 0.5
Time points reported: cumulative results from follow‐up visits
Subgroup: none
Funding sources Allergan, American Medical Systems, Astellas, Bayer, Beckman Coulter, Diagnostic Ultrasound, GTX, GlaxoSmithKline, Gen‐Probe, Indevus, Light Sciences Oncology, Lilly/ICOS, Merck, Novartis, Onconome, Pfizer, Sanofi, Solvay, Watson, National Institutes of Health, CapCURE, Pardee Foundation and Seder Foundation
Declarations of interest Allergan, American Medical Systems, Astellas, Bayer, Beckman Coulter, Diagnostic Ultrasound, GTX, GlaxoSmithKline, Gen‐Probe, Indevus, Light Sciences Oncology, Lilly/ICOS, Merck, Novartis, Onconome, Pfizer, Sanofi, Solvay, Watson, National Institutes of Health, CapCURE, Pardee Foundation and Seder Foundation
Notes Language of publication: English
Study author reply: "Pharmaceutics only have the data". We cannot contact study supporter in pharmaceutics due to wrong email address
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "randomly assigned (1:1) to double‐blind treatment, randomization schedule using PROC PLAN in SAS."
Allocation concealment (selection bias) Low risk Judgement: no information in article, but "Sequentially numbered, sealed, opaque envelopes" in protocol
Blinding of participants and personnel (performance bias) 
 All outcomes Low risk Quote: "Blinding was maintained throughout the study by the use of identical medication packaging with placebo matching silodosin in size and external appearance. Emergency information labels that indicated the patient’s assigned treatment were available to the investigator should knowledge of treatment assignment be needed to ensure the patient’s well‐being. If unblinding of the investigator, site personnel or the patient was required in a particular case that patient was to be discontinued from the study."
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Low risk Quote: "Blinding was maintained throughout the study by the use of identical medication packaging with placebo matching silodosin in size and external appearance. Emergency information labels that indicated the patient’s assigned treatment were available to the investigator should knowledge of treatment assignment be needed to ensure the patient’s well‐being. If unblinding of the investigator, site personnel or the patient was required in a particular case that patient was to be discontinued from the study."
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: discontinued/lost to follow‐up were 53/466 (11.4%) and 38/457 (8.3%), Loss of follow‐up 6/466 (1.3%) and 3/457 (0.3%), but discrepancies in the reason of discontinuation (47/466, 35/457) between intervention and placebo group. Last observations were carried forward to impute values missing for week 12
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) Low risk Judgement: protocols (NCT00224107, NCT00224120) were published and the prespecified outcomes were well described
Other bias High risk Judgement: single‐blind placebo run‐in for 4 weeks; participants with ≥ 30% decrease in IPSS or an increase in Qmax of ≥ 3 mL/s during the run‐in period were excluded from randomization

Masuda 2012.

Methods Study design: randomized, cross‐over study
Setting/Country: multicenter/Japan
Dates when study was conducted: November 2009‐March 2011
Participants Inclusion criteria: men ≥ 50 years with prostate estimated volume of > 20 cm3, IPSS of ≥ 8, a QoL score of ≥ 3 points
Exclusion criteria: men with organic diseases other than BPH (prostate cancer, bladder tumor, prostatitis, urethral stricture, etc.), transurethral resection of prostate or minimally invasive treatment, under catheterization and under self‐donating urination, active urinary tract infections, combined neuropathic bladder and nervous system disorder, received hormone‐based prostatic hyperplasia treatment medicine 6 months before the start of the study, received androgen‐receptor blocking agent 6 weeks before the study, and those judged unsuitable by the attending physician
Total number of participants randomly assigned: 92
Group A (Silodosin)
  • number of all participants randomly assigned: 44

  • age (years): 66.5 ± 5.6

  • prostate volume (mL): 38.8 ± 13.1

  • PSA (ng/mL): not reported

  • IPSS: 18.6 ± 5.5

  • Qmax (mL/s): 8.0 ± 3.7


Group B (Naftopidil)
  • number of all participants randomly assigned: 48

  • age (years): 68.5 ± 5.7

  • prostate volume (mL): 45.7 ± 17.8

  • PSA (ng/mL): not reported

  • IPSS: 17.6 ± 5.0

  • Qmax (mL/s): 9.3 ± 4.9

Interventions Run‐in period: none
Group A: silodosin 2 mg‐4 mg twice a day for 2 weeks in the morning and evening, and 4 mg twice daily in the morning and evening for 4 more weeks, if there were no problems such as side effects
Group B: naftopidil 50 mg‐75 mg once in the morning for 2 weeks, and if there were no problems such as side effects 75 mg was administered once in the morning for 4 more weeks
Duration: 6 weeks (before cross‐over)/total 12 weeks
Outcomes Primary outcome
  • total score of IPSS


How measured: IPSS questionnaire
Time points measured: before and after treatment (6 weeks and 12 weeks)
Time points reported: before and after treatment (6 weeks and 12 weeks)
Secondary outcome
  • IPSS subscore

  • QoL score

  • OABSS

  • Qmax

  • PVR

  • participant preference for the drug


How measured: IPSS: IPSS questionnaire/OABSS: OABSS questionnaire/PVR: abdominal US/patient preference: questionnaire survey
Time points measured: before administration of the drug and 4 weeks, 8 weeks
Time points reported: before administration of the drug and 4 weeks, 8 weeks
Safety outcomes
How measured: adverse events
Time points measured: not reported
Time points reported: at the end of study (12 weeks)
Subgroup: none
Funding sources Not reported
Declarations of interest Not reported
Notes Language of publication: Japanese
No wash‐out period between cross‐over
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) High risk Quote: "all patients who got consent were assigned in turn to 'Naf‐Silo group' and 'Silo‐Naf group' alternately"
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL High risk Judgement: 14/44 (31.9%) and 14/48 (29.2%) participants in the silodosin and naftopidil group were not included in the analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: not available (cross‐over trial)
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: 5/88 (5.7%) and 17/96 (17.8%) participants in the silodosin and tamsulosin group were not included in the analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: 5/88 (5.7%) and 17/96 (17.8%) participants in the silodosin and tamsulosin group were not included in the analysis
Selective reporting (reporting bias) Unclear risk Judgement: prespecified study outcomes were well described but protocol was not published
Other bias Unclear risk Judgement: no wash‐out period

Matsukawa 2016.

Methods Study design: prospective, open‐label, randomized study
Setting/Country: 52 urologists participated at a total of 44 investigational sites/outpatients/Japan
Dates when study was conducted: May 2012‐September 2013
Participants Inclusion criteria: men with total IPSS 8 or greater, IPSS QoL score 3 or greater, total OABSS 3 or greater, 1 or more urinary urgency episodes per week, prostate volume 20 ml or greater on transabdominal US, Qmax less than 15 ml per second at a voided volume of 100 ml or greater and PVR less than 150 ml, and age 50 years or greater
Exclusion criteria: men received oral treatment with a1‐blockers, anticholinergic agents, 5a‐reductase inhibitors, antidepressants, anti‐anxiety agents or sex hormonal agents, they had neurogenic bladder dysfunction, bladder calculi or active urinary tract infection, severe cardiac disease, renal dysfunction (serum creatinine levels 2 mg/dl or more) or hepatic dysfunction (aspartate and alanine aminotransferase concentrations more than twice normal values)
Total number of participants randomly assigned: 350
Group A (Silodosin)
  • number of all participants randomly assigned: 175

  • age (years): 70.6 ± 7.8

  • prostate volume (mL): 39.6 ± 16.7

  • PSA (ng/mL): 3.0 ± 3.1

  • IPSS: 18.8 ± 6.2

  • Qmax (mL/s): 8.2 ± 3.6


Group B (Naftopidil)
  • number of all participants randomly assigned: 175

  • age (years): 70.3 ± 7.8

  • prostate volume (mL): 38.6 ± 14.8

  • PSA (ng/mL): 3.0 ± 3.1

  • IPSS: 18.9 ± 6.1

  • Qmax (mL/s): 8.4 ± 3.0

Interventions Run‐in period: none
Group A: silodosin 4 mg per day for 4 weeks, followed by 8 mg per day for 8 weeks
Group B: naftopidil 50 mg per day for 4 weeks, followed by 75 mg per day for 8 weeks
Duration: 12 weeks
Outcomes Primary outcome
  • changes in IPSS

  • changes in IPSS QoL

  • changes in OABSS


How measured: IPSS and OABSS questionnaire
Time points measured: baseline 4 and 12 weeks after the start of treatment
Time points reported: baseline 4 and 12 weeks after the start of treatment
Secondary outcome
  • changes in Qmax

  • changes in PVR


How measured: uroflowmetry
Time points measured: baseline 4 and 12 weeks after the start of treatment
Time points reported: baseline 4 and 12 weeks after the start of treatment
Safety outcomes
How measured: not reported
Time points measured: not reported
Time points reported: not reported
Subgroup: none
Funding sources Not reported
Declarations of interest Not reported
Notes Language of publication: English
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Simple randomization using a random number table"
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Quote: "prospective, open label, randomized"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Quote: "prospective, open label, randomized"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 18/175 (10.2%) and 18/175 (10.2%) participants in silodosin and naftopidil group were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all subjects who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all subjects who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: information was partly reported (the number of withdrawal due to cardiovascular adverse events were only reported)
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: no information given
Selective reporting (reporting bias) Unclear risk Judgement: prespecified outcomes were well described, but protocol was not published
Other bias Low risk Judgement: not detected

Miyakita 2010.

Methods Study design: randomized, cross‐over study
Setting/Country: multicenter/Japan
Dates when study was conducted: May 2006‐July 2007
Participants Inclusion criteria: men with IPSS ≥ 8 points; QoL score ≥ 3 points; prostate volume measured by US ≥ 20 mL; void volume ≥ 100 mL; and Qmax < 15 mL/s
Exclusion criteria: already used any a1‐blocker for the treatment of hypertension; taking vardenafil hydrochloride hydrate; and otherwise judged by an attending physician to be inappropriate
Total number of participants randomly assigned: 97
Group A (Silodosin)
  • number of all participants randomly assigned: 46

  • age (years): 68.2 ± 8.6

  • prostate volume (mL): 41.3 ± 25.3

  • PSA (ng/mL): not reported

  • IPSS: 16.6 ± 5.2

  • Qmax (mL/s): 9.4 ± 3.5


Group B (Tamsulosin)
  • number of all participants randomly assigned: 51

  • age (years): 70.1 ± 8.9

  • prostate volume (mL): 37.8 ± 16.3

  • PSA (ng/mL): not reported

  • IPSS: 18.2 ± 5.8

  • Qmax (mL/s): 9.7 ± 4.4

Interventions Run‐in period: none
Group A: silodosin 8 mg/d (4 mg/dose, twice daily)
Group B: tamsulosin 0.2 mg/d (0.2 mg/dose, once daily)
Duration: 4 weeks (before cross‐over)/total 8 weeks
Outcomes Primary outcome
  • change in total IPSS from baseline


How measured: IPSS questionnaire
Time points measured: before administration of the drug and 1, 2, 4, 6 and 8 weeks after initiation of administration
Time points reported: before administration of the drug, and 4 weeks and 8 weeks
Secondary outcome
  • Qmax

  • PVR

  • BP

  • heart rate

  • participant preference for the drug


How measured: not reported
Time points measured: before administration of the drug, and 4 weeks and 8 weeks
Time points reported: before administration of the drug, and 4 weeks and 8 weeks
Safety outcomes
How measured: adverse events
Time points measured: throughout the study period
Time points reported: throughout the study period
Subgroup: none
Funding sources Not reported
Declarations of interest Not reported
Notes Language of publication: English
No wash‐out period between cross‐over
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL High risk Judgement: 12/46 (26.1%) and 20/51 (39.3%) participants in the silodosin and tamsulosin group were not included in the analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: not available (cross‐over trial)
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) Unclear risk Judgement: prespecified outcomes were well described, but protocol was not published
Other bias Unclear risk Judgement: no wash‐out period

Natarajan 2015.

Methods Study design: parallel, randomized
Setting/Country: tertiary hospital/Tamil Nadu, India
Dates when study was conducted: August 2013‐April 2015
Participants Inclusion criteria: men > 50 years with bothersome LUTS from BPH and IPSS > 7
Exclusion criteria: history of LUTS but not BPH, AUR in past 6 months, raised PSA level at baseline, serious co‐morbidity of vital organs, use of concomitant medication having anticholinergic, androgenic or estrogenic influence, on other α‐adrenergic antagonists or diuretics or with a history of prostatic or per urethral surgery or substance abuse
Total number of participants randomly assigned: 57
Group A (Silodosin)
  • number of all participants randomly assigned: 28

  • age (years): 61‐62

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported


Group B (Tamsulosin)
  • number of all participants randomly assigned: 29

  • age (years): 61‐62

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported

Interventions Run‐in period: none
Group A: silodosin 8 mg capsule once daily after dinner
Group B: tamsulosin 0.4 mg controlled‐release capsule once daily after dinner
Duration: 12 weeks
Outcomes Primary outcome
  • change in the total IPSS and QoL

  • change in the total QoL


How measured: IPSS questionnaire
Time points measured: baseline and 12 weeks
Time points reported: baseline and 12 weeks
Secondary outcome
  • proportion of participants who became completely or relatively symptom free (IPSS < 8) after 12 weeks of treatment

  • change in prostate size

  • change in Qmax


How measured: IPSS, US by blinded radiologist, uroflowmetry by blinded operator
Time points measured: baseline and 12 weeks
Time points reported: symptom free and change in Qmax not reported; change in prostate size, baseline and 12 weeks
Safety outcomes
How measured: adverse affects
Time points measured: at follow‐up visits (4, 8, 12 weeks)
Time points reported: cumulative results from follow‐up visits
Subgroup: none
Funding sources Not reported
Declarations of interest Not reported
Notes Language of publication: English
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: no information given
Selective reporting (reporting bias) High risk Quote: "Silodosin showed a significant decrease in IPSS versus tamsulosin at 2 weeks"
Judgement: protocol was not published and there was no information of 2 weeks' follow‐up. Secondary endpoints (proportion of participants who became completely or relatively symptom free (IPSS < 8), changes in Qmax) were not reported
Other bias Unclear risk Judgement: no data on baseline characteristics

NCT00793819.

Methods Study design: parallel, randomized, double‐blinded study
Setting/Country: NR
Dates when study was conducted: January 2009‐October 2009
Participants Inclusion criteria: men in good general health and ≥ 50 years, with symptoms of moderate‐severe BPH and nocturia (≥ 2 episodes per night)
Exclusion criteria: men with 1) medical conditions that would confound the efficacy evaluation 2) medical conditions in which it would be unsafe to use an alpha‐blocker 3) the use of concomitant drugs that would confound the efficacy evaluation 4) the use of concomitant drugs that would be unsafe with this alpha‐blocker
Total number of participants randomly assigned: 209
Group A (Silodosin)
  • number of all participants randomly assigned: 111

  • age (years): 64.6 ± 8.03

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported


Group B (Placebo)
  • number of all participants randomly assigned: 98

  • age (years): 64.2 ± 8.92

  • prostate volume (mL): not reported

  • PSA (ng/mL): not reported

  • IPSS: not reported

  • Qmax (mL/s): not reported

Interventions Run‐in period: not reported
Group A: silodosin 8 mg daily
Group B: 1 placebo capsule daily
Duration: 12 weeks
Outcomes Primary outcome
  • change in nocturia episodes


How measured: not reported
Time points measured: before and 12 weeks after treatment
Time points reported: before and 12 weeks after treatment
Secondary outcomes
  • change in QoL questionnaire responses


How measured: not reported
Time points measured: before and 12 weeks after treatment
Time points reported: before and 12 weeks after treatment
Subgroup: not reported
Funding sources Watson Pharmaceuticals
Declarations of interest Watson Pharmaceuticals
Notes Language of publication: English
Publication status: NCT00793819 (results in clinicaltrials.gov)
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Low risk Quote: "parallel randomized double blinded (Participant, Investigator)"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Low risk Quote: "parallel randomized double blinded (Participant, Investigator)"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) Low risk Judgement: protocol (NCT00793819) was published and the prespecified outcomes were well described
Other bias Unclear risk Judgement: protocol only

Pande 2014.

Methods Study design: single‐blind, parallel‐group, randomized, controlled trial
Setting/Country: outpatient/tertiary care hospital/Kolkata, India
Dates when study was conducted: July 2012‐June 2013
Participants Inclusion criteria: ambulatory, treatment naïve, men > 50 years with bothersome LUTS from BPH and IPSS > 7
Exclusion criteria: history of LUTS but not BPH, AUR in past 6 months, raised PSA level at baseline, serious co‐morbidity of vital organs, use of concomitant medication having anticholinergic, androgenic or estrogenic influence, on other α‐adrenergic antagonists or diuretics or with a history of prostatic or per urethral surgery or substance abuse
Total number of participants randomly assigned: 61
Group A (Silodosin)
  • number of all participants randomly assigned: 32

  • age (years): 61.4 ± 7.88

  • prostate volume (mL): 42.0 ± 19.96

  • PSA (ng/mL): not reported

  • IPSS: 18.4 ± 3.32

  • Qmax (mL/s): 15.5


Group B (Tamsulosin)
  • number of all participants randomly assigned: 29

  • age (years): 62.6 ± 7.55

  • prostate volume (mL): 35.6 ± 9.56

  • PSA (ng/mL): not reported

  • IPSS: 18.4 ± 3.94

  • Qmax (mL/s): 15.9

Interventions Run‐in period: none
Group A: silodosin 8 mg capsule once daily
Group B: tamsulosin 0.4 mg controlled‐release capsule once daily
Duration: 12 weeks
Outcomes Primary outcome
  • total IPSS score in addition to the QoL


How measured: IPSS questionnaire
Time points measured: at 4, 8 and 12 weeks
Time points reported: at 4, 8 and 12 weeks
Secondary outcome
  • proportion of participants who became completely or relatively symptom free (IPSS < 8) after 12 weeks of treatment

  • change in prostate size, in terms of volume, as assessed at US by a radiologist unaware of treatment allocation

  • changes in Qmax and allied parameters assessed at uroflowmetry by a blinded operator


How measured: IPSS questionnaire, US by blinded radiologist, uroflowmetry by blinded investigator
Time points measured: baseline and 12 weeks
Time points reported: baseline and 12 weeks
Safety outcomes
How measured: vital signs, treatment‐emergent adverse events, postural hypotension and retrograde ejaculation, laboratory investigations (complete blood count, fasting plasma glucose, routine liver function tests and creatinine level)
Time points measured: lab ‐ at baseline and study end/others ‐ each visit
Time points reported: lab ‐ at baseline and study end/others ‐ cumulative results from follow up visits
Subgroup: none
Funding sources M/s Cipla Limited, Mumbai, donated both study drugs and reimbursed part of the cost of laboratory investigations on request
Declarations of interest None
Notes Language of publication: English
Data (IPSS, QoL, AUR, surgical intervention, cardiovascular, sexual adverse events) were given by contact with study author
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "randomized in 1:1 ratio to one of the two study groups in fixed blocks of 8, using computer generated random number list"
Allocation concealment (selection bias) Low risk Quote: "Allocation concealment was achieved using the serially numbered, opaque, sealed envelope technique"
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Quote: "single blind, parallel group, randomized, controlled trial", "The capsules were removed from their commercial blister strip packaging and repackaged in air‐tight, screw cap containers and suitably labelled and coded as trial medication. Repackaging was done with the help of residents not otherwise involved in the study. Capsule identity was not revealed to the patients who received the total medication in three installments"
Judgement: participants were blinded, but investigator and observer might be not blinded
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Quote: "single blind, parallel group, randomized, controlled trial", "The capsules were removed from their commercial blister strip packaging and repackaged in air‐tight, screw cap containers and suitably labelled and coded as trial medication. Repackaging was done with the help of residents not otherwise involved in the study. Capsule identity was not revealed to the patients who received the total medication in three installments"
Judgement: participants were blinded, but investigator and observer might be not blinded
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 6/32 (18.7%) in silodosin, 2/29 (6.9%) in tamsulosin lost to follow‐up: missing data were imputed by last observation carried forward strategy
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Low risk Judgement: all participants who were randomized in each group were included in analysis. We received the data after contacting the study author.
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) Low risk Judgement: protocol (Clinical Trials Registry‐India (CTRI/2014/01/004366)) was published and the prespecified outcomes were well described. Data (AUR, surgical intervention, cardiovascular, sexual adverse events) were given by contact with study author
Other bias Low risk Judgement: not detected

Shirakawa 2013.

Methods Study design: randomized, open‐label, controlled, multicenter study
Setting/Country: Kobe University School or other collaborating institutions/Japan
Dates when study was conducted: July 2007‐March 2011
Participants Inclusion criteria: men with: BPH/ LUTS with a total IPSS ≥ 8 points; QoL index ≥ 3 points; Qmax < 15 mL/s; prostate volume ≥ 20 mL; and either without a history of using any a1‐receptor blocker (hereafter, drug‐naive group) or who had continued to use tamsulosin (0.2 mg once daily) for ≥ 3 months and wanted to switch the medication to another oral drug (hereafter, drug‐switching group)
Exclusion criteria: other diseases such as prostate cancer, bladder tumor, cystolithiasis, prostatitis, urethral stricture, or active urinary tract infection; complication of neurogenic bladder or disease suspected of neurogenic bladder; and whose participation in the study was deemed inappropriate by their primary physician(s)
Total number of participants randomly assigned: 121
Group A (Silodosin)
  • number of all participants randomly assigned: 61

  • age (years): 70.98 ± 6.69

  • prostate volume (mL): 38.24 ± 12.94

  • PSA (ng/mL): not reported

  • IPSS: 17.53 ± 5.4

  • Qmax (mL/s): 9.87 ± 4.50


Group B (Naftopidil)
  • number of all participants randomly assigned: 60

  • age (years): 70.50 ± 6.58

  • prostate volume (mL): 39.39 ± 25.96

  • PSA (ng/mL): not reported

  • IPSS: 17.56 ± 6.7

  • Qmax (mL/s): 11.13 ± 6.53

Interventions Run‐in period: none
Group A: silodosin 4 mg twice daily
Group B: naftopidil 50 mg once daily
Duration: 8 consecutive weeks
Outcomes
  • total IPSS

  • subtotal IPSS of storage symptoms

  • subtotal IPSS of voiding symptoms

  • post‐micturition symptoms

  • QoL index


How measured: IPSS questionnaire
Time points measured: at baseline, 4 and 8 weeks
Time points reported: at baseline, 4 and 8 weeks
Safety outcomes
How measured: adverse events
Time points measured: not reported
Time points reported: cumulative results from follow‐up visits
Subgroup: drug‐naïve/drug‐switching
Funding sources Not reported
Declarations of interest None
Notes Language of publication: English
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "randomly assigned (using a random number table)"
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Quote: "randomized, open‐label, controlled multicenter study"
Judgement: no one was blinded (protocol)
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Quote: "randomized, open‐label, controlled multicenter study"
Judgement: no one was blinded (protocol)
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Low risk Judgement: 5/61 (8.2%) and 4/60 (6.7%) participants in silodosin and naftopidil were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk 2/61 (3.3%) and 3/60 (5.0%) participants in silodosin and naftopidil were not included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: 2/61 (3.3%) and 3/60 (5.0%) participants in silodosin and naftopidil were not included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: 2/61 (3.3%) and 3/60 (5.0%) participants in silodosin and naftopidil were not included in analysis
Selective reporting (reporting bias) Low risk Judgement: protocol (UMIN000008331) was published. While the results were shown separately in participants with drug naïve and switching group, review outcomes were well described
Other bias Low risk Not detected

Takeshita 2016.

Methods Study design: randomized, cross‐over study
Setting/Country: 4 community‐based hospitals/Japan
Dates when study was conducted: February 2011‐July 2014
Participants Inclusion criteria: men aged ≥ 50 years with LUTS/BPH, an IPSS of ≥ 8, QoL score of ≥ 3, and US‐estimated prostatic volume of ≥ 20 mL
Exclusion criteria: those that had taken any alpha‐blocker within the previous 28 days; those currently taking phosphodiesterase type 5 inhibitors, 5‐ARIs, or antiandrogens; those with severe renal dysfunction (estimated glomerular filtration rate < 30 mL/min per 1.73 m2); and those judged to be inappropriate by the attending physicians
Total number of participants randomly assigned: 34
Group A (Silodosin)
  • number of all participants randomly assigned: 18

  • age (years): 69.6 ± 5.4

  • prostate volume (mL): 38.7 ± 11.6

  • PSA (ng/mL): 5.9 ± 5.9

  • IPSS: 17.1 ± 7.3

  • Qmax (mL/s): 9.5 ± 4.9


Group B (Tamsulosin)
  • number of all participants randomly assigned: 16

  • age (years): 69.4 ± 7.0

  • prostate volume (mL): 47.3 ± 30.4

  • PSA (ng/mL): 5.4 ± 4.4

  • IPSS: 15.2 ± 7.0

  • Qmax (mL/s): 10.2 ± 3.8

Interventions Run‐in period: none
Group A: silodosin 4 mg once daily in the morning
Group B: tamsulosin 0.2 mg once daily in the morning
Duration: 4 weeks (before cross‐over)/total 8 weeks
Outcomes Primary outcome
  • changes in IPSS and QoL score


How measured: IPSS questionnaire
Time points measured: baseline to weeks 4 and 8
Time points reported: baseline to weeks 4 and 8
Secondary outcome
  • uroflowmetry

  • BP

  • changes in subjective parameters including IPSS subscores and OABSS

  • participant preference for the drug


How measured: participant preference for the drug by self‐administered questionnaire
Time points measured: before administration of the drug, and 4 weeks and 8 weeks
Time points reported: before administration of the drug, and 4 weeks and 8 weeks
Safety outcomes
How measured: adverse events
Time points measured: baseline to weeks 4 and 8
Time points reported: baseline to weeks 4 and 8
Subgroup: none
Funding sources Not reported
Declarations of interest None
Notes Language of publication: English
No wash‐out period between cross‐over
Data (IPSS, QoL, treatment withdrawal, cardiovascular, sexual adverse events) were given by contact with study author
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Judgement: author reply "random number table"
Allocation concealment (selection bias) Low risk Judgement: author reply "sealed opaque envelope"
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Quote: "open‐label, randomized crossover study"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Quote: "open‐label, randomized crossover study"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 2/18 (11.2%) and 2/16 (12.5%) participants in the silodosin and tamsulosin group were not included in the analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: not available (cross‐over trial)
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) Low risk Judgement: protocol (JPRN‐UMIN000004918) was published and prespecified, study outcomes were analyzed as planned. Data (IPSS, QoL, treatment withdrawal, cardiovascular, sexual adverse events) were provided after contact with study author
Other bias Unclear risk Judgement: no wash‐out period

Watanabe 2011.

Methods Study design: randomized, cross‐over study
Setting/Country: 3 institutions/Japan
Dates when study was conducted: February 2008‐September 2009
Participants Inclusion criteria: men with LUTS associated with BPH and had an IPSS ≥ 8 and an IPSS‐QoL score ≥ 2
Exclusion criteria: not reported
Total number of participants randomly assigned: 102
Group A (Silodosin)
  • number of all participants randomly assigned: 51

  • age (years): 69.3 ± 8.3

  • prostate volume (mL): 36.6 ± 18.3

  • PSA (ng/mL): not reported

  • IPSS: 16.4 ± 5.0

  • Qmax (mL/s): 9.1 ± 5.7


Group B (Tamsulosin)
  • number of all participants randomly assigned: 51

  • age (years): 69.9 ± 8.4

  • prostate volume (mL): 35.1 ± 13.0

  • PSA (ng/mL): not reported

  • IPSS: 18.1 ± 6.2

  • Qmax (mL/s): 8.8 ± 5.2

Interventions Run‐in period: none
Group A: silodosin 4 mg, orally, twice daily
Group B: tamsulosin 0.2 mg, orally, once daily
Duration: 4 weeks (before cross‐over)/total 8 weeks
Outcomes Primary outcome
  • participant preference for the drug and reason of preference


How measured: questionnaire and open question
Time points measured: 8 weeks
Time points reported: 8 weeks
Secondary outcome
  • IPSS

  • IPSS‐QoL

  • Qmax

  • mean urinary flow rate

  • PVR


How measured: not reported
Time points measured: baseline and at 4 and 8 weeks
Time points reported: baseline and at 4 and 8 weeks
Safety outcomes
How measured: adverse events
Time points measured: not reported
Time points reported: during the study
Subgroup: age ≥ 70/IPSS ≥ 20
Funding sources Not reported
Declarations of interest None
Notes Language of publication: English
No wash‐out period between cross‐over
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Quote: "open‐label study"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Quote: "open‐label study"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 9/51 (17.7%) and 9/51 (17.7%) participants in the silodosin and tamsulosin group were not included in the analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: not available (cross‐over trial)
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: 14/102 (13.8%) and 11/102 (10.8%) participants in the silodosin and tamsulosin group were not included in the analysis (study author reported the results after cross‐over)
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: 14/102 (13.8%) and 11/102 (10.8%) participants in the silodosin and tamsulosin group were not included in the analysis (study author reported the results after cross‐over)
Selective reporting (reporting bias) Unclear risk Judgement: no protocol available
Other bias Unclear risk Judgement: no wash‐out period

Yamaguchi 2013.

Methods Study design: parallel, randomized trial
Setting/Country: Nihon University School of Medicine/Japan
Dates when study was conducted: December 2007‐November 2010
Participants Inclusion criteria: men with BPH 50 years with significant LUTS and deteriorated QoL, with IPSS of ≥ 8 and its QoL score of ≥ 3
Exclusion criteria: established prostate cancer, neurogenic bladder and any other complications that affect micturitional status; underwent prostate surgery, intervention or radiation therapy
Total number of participants randomly assigned: 109
Group A (Silodosin)
  • number of all participants randomly assigned: 58

  • age (years): 69.3 ± 7.8

  • prostate volume (mL): 33.2 ± 21.2

  • PSA (ng/mL): 2.8 ± 3.3

  • IPSS: 16.9 ± 5.5

  • Qmax (mL/s): 10.4 ± 5.0


Group B (Naftopidil)
  • number of all participants randomly assigned: 51

  • age (years): 70.0 ± 7.0

  • prostate volume (mL): 39.5 ± 18.0

  • PSA (ng/mL): 3.9 ± 3.5

  • IPSS: 18.9 ± 7.0

  • Qmax (mL/s): 9.9 ± 5.3

Interventions Run‐in period: none
Group A: silodosin 8 mg/d
Group B: naftopidil 75 mg/d
Duration: 12 weeks
Outcomes
  • IPSS

  • QoL

  • IIEF‐5

  • Qmax

  • PVR


How measured: questionnaire, uroflowmetry
Time points measured: before and 4, 8, and 12 weeks after treatment
Time points reported: IPSS, QoL, IIEF‐5: before and 4, 8, and 12 weeks after treatment/Qmax, PVR: before and 12 weeks after treatment
Subgroup: none
Funding sources Not reported
Declarations of interest None
Notes Language of publication: English
Data (IPSS, QoL, treatment withdrawal, AUR, surgical intervention, cardiovascular adverse events) were given by contact with study author
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Judgement: author reply "random number table envelope method"
Allocation concealment (selection bias) Unclear risk Judgement: author reply "random number table envelope method"
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Judgement: author reply "not blinded"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Judgement: author reply "not blinded"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL High risk Judgement: 17/58 (29.3%) and 13/51 (25.5) participants in silodosin and naftopidil group were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis. We received the data after contacting the study author
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis. We received the data after contacting the study author
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Low risk Judgement: all participants who were randomized in each group were included in analysis. We received the data after contacting the study author
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis. We received the data after contacting the study author
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all sexually active subjects (silodosin: 23/53 (44%), naftopidil: 21/44 (47%)) who were randomized in each group were included in analysis
Selective reporting (reporting bias) Unclear risk Judgement: data (treatment withdrawal, AUR, and surgical intervention) were given by contact with study author, but protocol was not published
Other bias Low risk Judgement: not detected

Yamanishi 2011.

Methods Study design: parallel, randomized trial
Setting/Country: not reported
Dates when study was conducted: not reported
Participants Inclusion criteria: men with IPSS total score ≥ 8, Qmax < 15 mL/s, total prostate volume measured by US > 20 mL
Exclusion criteria: prostate cancer, urethral stricture, apparent neurogenic bladder and those on medication that might affect voiding function such as alpha‐blockers, anticholinergics and/or antiandrogen drugs
Total number of participants randomly assigned: 149
Group A (Silodosin)
  • number of all participants randomly assigned: 75

  • age (years): 71.3 ± 8.2

  • prostate volume (mL): 42.0 ± 23.7

  • PSA (ng/mL): 3.2 ± 3.6

  • IPSS: 18.8 ± 7.3

  • Qmax (mL/s): 7.7 ± 2.8


Group B (Tamsulosin)
  • number of all participants randomly assigned: 74

  • age (years): 72.2 ± 7.6

  • prostate volume (mL): 41.2 ± 23.0

  • PSA (ng/mL): 3.7 ± 3.7

  • IPSS: 17.8 ± 6.4

  • Qmax (mL/s): 8.4 ± 3.3

Interventions Run‐in period: none
Group A: silodosin 4 mg twice daily
Group B: tamsulosin 0.2 mg‐0.4 mg daily
Duration: 12 months
Outcomes
  • IPSS

  • QoL

  • average flow rate

  • Qmax

  • PVR


How measured: questionnaire, uroflowmetry
Time points measured: before, and at 1, 3, 6 and 12 months after the therapy
Time points reported: before, and at 1, 3, 6 and 12 months after the therapy
Subgroup: none
Funding sources Not reported
Declarations of interest Not reported
Notes Language of publication: English
Publication status: abstract (full text has not been published; study author reply)
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL High risk Judgement: 19/75 (25.3%) and 30/74 (40.5%) participants in silodosin and tamsulosin were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: no information given
Selective reporting (reporting bias) Unclear risk Judgement: protocol was not published and no information related to the review outcomes given
Other bias Unclear risk Judgement: abstract only

Yokoyama 2011.

Methods Study design: parallel, randomized trial
Setting/Country: Kawasaki Medical School/Japan
Dates when study was conducted: June 2007‐December 2008
Participants Inclusion criteria: men with LUTS aged 50–80 years and with IPSS ≥ 8
Exclusion criteria: received oral treatment with 5‐ARIs, anticholinergic agents, antidepressants, or sex hormonal agents, had neurogenic bladder dysfunction, bladder calculi, or active urinary tract infection, or had severe cardiac disease, renal dysfunction (serum creatinine > 2 mg/dL), or hepatic dysfunction
Total number of participants randomly assigned: 136
Group A (Silodosin)
  • number of all participants randomly assigned: 45

  • age (years): 70.2 ± 0.9

  • prostate volume (mL): 33.3 ± 2.3

  • PSA (ng/mL): not reported

  • IPSS: 18.7 ± 0.7

  • Qmax (mL/s): 9.03 ± 0.6


Group B (Tamsulosin)
  • number of all participants randomly assigned: 45

  • age (years): 71.5 ± 1.1

  • prostate volume (mL): 32.5 ± 2.0

  • PSA (ng/mL): not reported

  • IPSS: 18.0 ± 1.1

  • Qmax (mL/s): 8.56 ± 0.5


Group C (Naftopidil)
  • number of all participants randomly assigned: 46

  • age (years): 69.1 ± 1.2

  • prostate volume (mL): 35.0 ± 3.1

  • PSA (ng/mL): not reported

  • IPSS: 17.4 ± 0.8

  • Qmax (mL/s): 8.63 ± 0.5

Interventions Run‐in period: none
Group A: silodosin 4 mg twice a day
Group B: tamsulosin 0.2 mg once a day
Group C: naftopidil 50 mg once a day
Duration: 12 weeks
Outcomes
  • IPSS

  • QoL

  • IIEF

  • Qmax

  • PVR


How measured: questionnaire (IPSS, QoL, IIEF), uroflowmetry, US (PVR)
Time points measured: before, and 1 and 3 months
Time points reported: before, and 1 and 3 months
Subgroup: none
Funding sources Not reported
Declarations of interest None (study author reply)
Notes Language of publication: English
Data (IPSS, QoL, AUR, and surgical intervention) from study author
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Judgement: study author reply "Computer generated central randomization"
Allocation concealment (selection bias) Low risk Judgement: study author reply "Computer generated central randomization"
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Judgement: study author reply "participants were not blinded"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Unclear risk Judgement: no information given
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 4/45 (8.9%), 6/45 (13.3%), and 4/46 (8.7) participants in silodosin, tamsulosin, and naftopidil group were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Unclear risk Judgement: 4/45 (8.9%), 6/45 (13.3%), and 4/46 (8.7) participants in silodosin, tamsulosin, and naftopidil group were not included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: 4/45 (8.9%), 6/45 (13.3%), and 4/46 (8.7) participants in silodosin, tamsulosin, and naftopidil group were not included in analysis. We received the data after contacting the study author
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: 4/45 (8.9%), 6/45 (13.3%), and 4/46 (8.7) participants in silodosin, tamsulosin, and naftopidil group were not included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all sexually active subjects (silodosin: 11/45 (24.4%), tamsulosin 12/45 (26.6%), naftopidil: 15/46 (31.9%)) were included in analysis
Selective reporting (reporting bias) Unclear risk Judgement: prespecified outcomes were well described and data were given by study author, but protocol was not published
Other bias High risk Judgement: drug administration times were different among groups. Baseline imbalance in PVR, but the fact that silodosin group had much higher PVR may underestimate the effect size

Yokoyama 2012.

Methods Study design: randomized, cross‐over study
Setting/Country: single center/Japan
Dates when study was conducted: June 2008‐March 2010
Participants Inclusion criteria: men aged 50 years who had a total IPSS ≥ 8 and a QoL index ≥ 3
Exclusion criteria: prostate cancer, neurogenic bladder, urethral stricture, active urinary tract infection and other complications considered likely to affect micturition
Total number of participants randomly assigned: 46
Group A (Silodosin)
  • number of all participants randomly assigned: 23

  • age (years): 68.9 ± 5.6

  • prostate volume (mL): 35.0 ± 18.4

  • PSA (ng/mL): 2.5 ± 3.4

  • IPSS: 19.3 ± 4.9

  • Qmax (mL/s): 7.2 ± 2.9


Group B (Tamsulosin)
  • number of all participants randomly assigned: 23

  • age (years): 70.0 ± 6.8

  • prostate volume (mL): 36.1 ± 15.5

  • PSA (ng/mL): 3.5 ± 3.7

  • IPSS: 21.1 ± 6.8

  • Qmax (mL/s): 7.6 ± 3.0

Interventions Run‐in period: none
Group A: silodosin 4 mg twice daily
Group B: tamsulosin 0.2 mg once daily
Duration: 3 months (before cross‐over)/1 month wash‐out/3 months (after cross‐over)/total 7 months
Outcomes
  • IPSS

  • QoL index

  • Qmax

  • PVR


How measured: IPSS questionnaire and transabdominal US (PVR)
Time points measured: before and after treatment (not reported in method)
Time points reported: before and after treatment (1 months, 3 months, 4 months, and 7 months)
Safety outcomes
How measured: adverse events
Time points measured: not reported
Time points reported: not reported
Subgroup: none
Funding sources None
Declarations of interest None
Notes Language of publication: English
One month wash‐out period between cross‐over
Data (IPSS, QoL, treatment withdrawal for any reason, method of random sequence generation, allocation concealment, and blinding) were given by contact with study author
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Judgement: study author reply "random number table"
Allocation concealment (selection bias) Unclear risk Judgement: study author reply "sealed envelope"
Blinding of participants and personnel (performance bias) 
 All outcomes High risk Judgement: study author reply "participants were blinded"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes High risk Judgement: study author reply "participants were blinded"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 5/46 (10.9%) participants were not included in the analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: not available (cross‐over trial)
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Low risk Judgement: all participants who were randomized in each group were included in analysis
Selective reporting (reporting bias) Unclear risk Judgement: prespecified outcomes are well described and data were given by study author, but protocol was not published (study author confirmed via email)
Other bias Low risk Judgement: not detected

Yu 2011.

Methods Study design: parallel, randomized, double‐blinded
Setting/Country: 9 medical centers/Taiwan
Dates when study was conducted: July 2007‐September 2008
Participants Inclusion criteria: men aged ≥ 40 years with an IPSS of ≥ 13, a health‐related QoL score of ≥ 3, a prostate volume of ≥ 20 mL, and a Qmax of < 15 mL/s with a voided volume of ≥ 100 mL
Exclusion criteria: men with a history of previous prostate surgery, prostate cancer, neurogenic bladder, bladder neck constriction, urethral stricture, bladder calculus, active urinary tract infection, a PVR of > 250 mL, exposure to sex hormone within 3 months prior to the wash‐out period, renal dysfunction (serum creatinine of > 2.0 mg/dL), a history of severe liver impairment, severe cardiovascular diseases, severe hypotension, and known hypersensitivity or history of active substance abuse (including alcohol) within the past 2 years
Total number of participants randomly assigned: 209
Group A (Silodosin)
  • number of all participants randomly assigned: 105

  • age (years): 67.5 ± 9.3

  • prostate volume (mL): 44.8 ± 24.2

  • PSA (ng/mL): not reported

  • IPSS: 19.3 ± 4.5

  • Qmax (mL/s): 10.3 ± 2.8


Group B (Tamsulosin)
  • number of all participants randomly assigned: 104

  • age (years): 65.0 ± 8.8

  • prostate volume (mL): 38.2 ± 16.7

  • PSA (ng/mL): not reported

  • IPSS: 19.8 ± 4.5

  • Qmax (mL/s): 10.6 ± 2.8

Interventions Run‐in period: 7‐day wash‐out and 7‐day observation periods
Group A: silodosin 4 mg twice daily
Group B: tamsulosin 0.2 mg in the morning and one placebo capsule in the evening
Duration: 12 weeks
Outcomes Primary outcome
  • change in the IPSS from baseline

  • participants who achieved 25% reduction in the IPSS from baseline


How measured: IPSS questionnaire
Time points measured: 0 (initiation of treatment), 2, 4, 8, and 12 weeks
Time points reported: 0 (initiation of treatment), 2, 4, 8, and 12 weeks in figure
Secondary outcome
  • Qmax

  • health‐related QoL from baseline

  • changes in IPSS subscores (voiding and storage symptom scores)


How measured: uroflowmetry, IPSS questionnaire
Time points measured: 0, 4 and 12 weeks/0 (initiation of treatment), 2, 4, 8, and 12 weeks
Time points reported: 0 and 12 weeks
Safety outcomes
How measured: all adverse events/BP/pulse rate/laboratory tests
Time points measured: 0 (initiation of treatment), 2, 4, 8, and 12 weeks/0, 4 and 12 weeks/follow‐up visits
Time points reported: 0 and 12 weeks/cumulative results from follow‐up visits (adverse events)
Subgroup: none
Funding sources Synmosa pharmaceutical company
Declarations of interest All authors were study investigators for Synmosa
Notes Language of publication: English
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Judgement: no information given
Allocation concealment (selection bias) Unclear risk Judgement: no information given
Blinding of participants and personnel (performance bias) 
 All outcomes Low risk Quote: "both investigators and patients were ‘blinded’ to treatment"
Blinding of outcome assessment (detection bias) 
 Subjective outcomes Low risk Quote: "both investigators and patients were ‘blinded’ to treatment"
Blinding of outcome assessment (detection bias) 
 Objective outcomes Low risk Judgement: objective outcomes are not likely affected by lack of blinding
Incomplete outcome data (attrition bias) 
 Urologic symptom scores/ QoL Unclear risk Judgement: 18/105 (17.1%), 21/104 (20.2%) participants who were randomized in silodosin, tamsulosin group were not included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal for any reason Low risk Judgement: all participants who were randomized in each group were included in analysis
Incomplete outcome data (attrition bias) 
 Treatment withdrawal due to adverse events Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 AUR/ surgical intervention Unclear risk Judgement: no information given
Incomplete outcome data (attrition bias) 
 Cardiovascular adverse events Unclear risk Judgement: 18/105 (17.1%), 21/104 (20.2%) participants who were randomized in silodosin, tamsulosin group were not included in analysis
Incomplete outcome data (attrition bias) 
 Sexual adverse events Unclear risk Judgement: 18/105 (17.1%), 21/104 (20.2%) participants who were randomized in silodosin, tamsulosin group were not included in analysis
Selective reporting (reporting bias) Unclear risk Judgement: protocol was published, but protocol was not found due to the absence of protocol number
Other bias High risk Judgement: serious baseline imbalance in age, IPSS, prostate volume, health‐related QoL, Qmax in as treated analysis. Two weeks' observation run‐in period

5‐ARIs: 5‐alpha reductase inhibitors; AUR: acute urinary retention; BP: blood pressure; BPH: benign prostatic hyperplasia; IIEF: International Index of Erectile Function; IPSS: International Prostate Symptom Score; LUTS: lower urinary tract symptoms; OABSS: Overactive Bladder Symptom Score; PSA: prostate‐specific antigen; PVR: postvoid residual; Qmax; maximum flow rate; QoL: quality of life; US: ultrasound

Characteristics of excluded studies [ordered by study ID]

Study Reason for exclusion
Abramowicz 2009 Review
Alcántara Montero 2016 Review
Araki 2013 Wrong study design (non randomized trial)
Boeri 2016 Wrong study design (study design not described, silodosin versus silodosin plus Serenoa repens)
Cakiroglu 2016 Wrong comparator (silodosin versus transurethral incision of prostate)
Chapple 2009 Wrong study design (integrated safety analysis)
Curran 2011 Review
JPRN‐UMIN000007917 Wrong comparator (silodosin 4 mg once daily versus silodosin 4 mg twice daily)
Kawabe 2006b Wrong study design (non randomized study)
Kobayashi 2008 Wrong outcome (semen parameters)
Kobayashi 2009 Wrong outcome (semen parameters)
Manjunatha 2016b Wrong outcome (cost effectiveness)
Matsukawa 2012 Wrong comparator (silodosin versus silodosin and propiverine)
Matsukawa 2017 Wrong comparator (silodosin and dutasteride versus dutasteride)
Michel 2011 Wrong study population (participants with ≥ 2 voids per night )
Montorsi 2010 Review
Montorsi 2013 Review
Prescrire Int 2011 Review (medical article)
Prescrire Int 2012 Review (medical article)
Roehrborn 2009 Wrong study population (participants with retrograde ejaculation as adverse event)
Yoshida 2017 Wrong comparator (silodosin versus phosphodiesterase 5 inhibitor)
Yoshihisa 2012 Wrong comparator (silodosin versus silodosin and propiverine)
Zhou 2011 Wrong outcome (pharmacokinetics and adverse events (not available))

Characteristics of studies awaiting assessment [ordered by study ID]

CTRI/2010/091/000526.

Methods Randomized, parallel‐group, active, controlled trial
Participants Inclusion criteria
  • Ages: > 45 years and < 80 years

  • Genders eligible for study: male only

  • Men in good general health and ≥ 50 years, with symptoms of moderate‐severe BPH

  • A mean symptoms score of ≥ 8 (symptoms of BPH assessed as per American Urological Association Symptom Index, which assesses the occurrence of seven symptoms characteristic of BPH during the preceding week, each scored on a scale from 0 (absent) to 5 (severe)).

  • Mean Qmax of ≤ 15 mL/s and ≤ 4 mL/s, with a minimal voided volume of 125 mL, and a mean PVR of < 300 mL

  • PSA values of > 4 ng/mL

  • Renal function defined as a serum creatinine level of < 2.0 mg/dL and creatinine clearance of > 20 mL/min by Cockroft and Gault formula

Interventions Group A: silodosin capsule 8 mg
Group B: tamsulosin ER capsules 0.4 mg
Duration: 12 weeks
Outcomes
  • change in baseline score on the American Urological Association Symptom Index

  • change in baseline urine flow rate

  • change in PSA

  • safety and tolerability

Notes Funding source: MSN Laboratories Ltd
Publication status: CTRI/2010/091/000526 (final publication status has not been clarified)

Devana 2014.

Methods Open‐label, parallel study
Participants Inclusion criteria: men with symptomatic BPH aged 50‐80 years, with IPSS > 8 or QoL score > 2
Exclusion criteria: not reported
Interventions Group A: silodosin 8 mg/d
Group B: tamsulosin 0.4 mg/d
Group C: alfuzosin 10 mg/d
Duration: 1 month
Outcomes
  • IPSS

  • QoL scores

  • Qmax

  • PVR

  • side effects

Notes Publication status: abstract (final publication status has not been clarified)

Jha 2015.

Methods Parallel, randomized study
Participants Inclusion criteria: not reported
Exclusion criteria: not reported
Interventions Group A: silodosin 8 mg daily at night
Group B: tamsulosin 0.4 mg daily at night
Duration: 6 months
Outcomes
  • IPSS

  • QoL

  • digital rectal exam

  • PSA

  • Qmax

  • PVR

  • side effects

Notes Publication status: abstract (final publication status has not been clarified)

JPRN‐UMIN000003125.

Methods Open‐label, parallel, randomized study
Participants Inclusion criteria: male patients with newly diagnosed LUTS according to the treatment algorithm of the Clinical Practice Guidelines for male LUTS
Exclusion criteria: men who meet any of the following criteria are excluded:
  • taking anticholinergic agents, antidepressants, or antianxiety agents

  • a history of urinary retention, urinary tract infections, macroscopic hematuria, lower urinary tract surgery, radiotherapy for the pelvic organs, or neurologic disease

  • pyuria, renal impairment, calculus bladder, or urine cytology positive

  • elevated PSA levels (≥ 4.0 ng/mL)

  • 9th CLSS score ≥ 2

  • not willing to receive drug treatment

  • cannot answer the questionnaire by themselves

  • classified as ineligible by the investigator

Interventions Group A: silodosin 8 mg daily
Group B: tamsulosin 0.2 mg daily
Duration: not reported
Outcomes
  • IPSS

  • Overactive Bladder Symptom Score

  • uroflowmetry

  • PVR

  • prostate volume

Notes Funding source: none
Publication status: JPRN‐UMIN000003125 (final publication status has not been clarified)

JPRN‐UMIN000005151.

Methods Parallel, randomized study
Participants Inclusion criteria
Men aged ≥ 50 years with previously‐untreated BPH with total IPSS ≥ 8 and QoL score ≥ 2
Interventions Group A: silodosin 4 mg twice daily
Group B: tamsulosin hydrochloride 0.2 mg daily
Duration: not reported
Outcomes
  • IPSS and QoL score

  • uroflowmetry

  • PVR

  • prostate volume

  • Japanese‐Gastrointestinal symptom Rating Scale

  • Bristol Stool Form

  • safety (adverse events)

  • rate of discontinuation

Notes Funding source: none
Publication status: JPRN‐UMIN000005151 (final publication status has not been clarified)

JPRN‐UMIN000008538.

Methods Parallel, randomized study
Participants Inclusion criteria
Men aged 50‐90 years
1) a total IPSS of ≥ 8
2) Qmax of ≤ 15 mL/s as evaluated by uroflowmetry
3) prostate volume of ≥ 15 mL as measured by prostatic US
Interventions Group A: silodosin
Group B: tamsulosin
Duration: not reported
Outcomes
  • changes in IPSS

  • uroflowmetry

  • bladder diary

Notes Funding source: Dokkyo Medical University
Publication status: JPRN‐UMIN000008538 (final publication status has not been clarified)

JPRN‐UMIN000011556.

Methods Parallel, randomized study
Participants Inclusion criteria
Men aged ≥ 50 years
  • Participant must have urinary symptoms associated with BPH

  • Participant must give written informed consent

  • Participant must satisfy the following conditions during screening conducted at the start of the treatment period (Day 0): the total IPSS voiding symptom score is ≥ 8, the QoL score is ≥ 2, prostate volume is ≥ 20 mL, Qmax is ≤ 15 mL/s, and PVR is ≤ 100 mL

  • Participant must not use prohibited concomitant drugs except at least 2 weeks before the start of the treatment period (Day 0), or the subject must be able to undergo a drug wash‐out for at least 2 weeks before the start of the treatment period

  • Participant must be an outpatient who can fill in the questionnaire

  • Participant must be ≥ 65 years at the time of signing the informed consent

Interventions Group A: silodosin 4 mg, once daily after breakfast
Group B: tamsulosin 0.2 mg, once daily after breakfast
Duration: not reported
Outcomes
  • IPSS total score and subscores

  • QoL score

  • Overactive bladder symptom score

  • urodynamic parameters

  • blood pressure

  • adverse events and adverse drug reactions

Notes Funding source: Kissei Pharmaceutical Co., Ltd
Publication status: JPRN‐UMIN000011556 (final publication status has not been clarified)

Mandal 2013.

Methods Open‐label, parallel study
Participants Inclusion criteria: men with symptomatic BPH aged 50‐80 years with IPSS > 8 or QoL score > 2
Exclusion criteria: other causes of bladder outlet obstruction, prostate and bladder malignancy, renal failure, hepatic failure and prior history of prostatectomy
Interventions Group A: silodosin 8 mg/d
Group B: tamsulosin 0.4 mg/d
Group C: alfuzosin 10 mg/d
Duration: 1 month
Outcomes
  • IPSS

  • QoL

  • Qmax

  • average urinary flow rate

  • PVR

  • side effects

Notes Publication status: abstract (final publication status has not been clarified)

Manohar 2014.

Methods Parallel, randomized study
Participants Inclusion criteria: patients of LUTS in BPH
Exclusion criteria: not reported
Interventions Group A: silodosin 8 mg once/d
Group B: tamsulosin 0.4 mg once/d
Group C: alfuzosin 10 mg once/d
Duration: 12 weeks
Outcomes
  • IPSS

  • QoL

  • Qmax

  • PVR

  • safety profile

Notes Publication status: abstract (final publication status has not been clarified)

Miyamae 2011.

Methods Parallel, randomized study
Participants Inclusion criteria: men with untreated BPH
  • gave their informed consent to participate in this study

  • outpatients aged ≥ 50 years

  • total IPSS of ≥ 8 and QoL score of ≥ 3

  • never been treated with an alpha‐1‐blocker or had not received an alpha‐1‐blocker for at least 4 weeks

  • judged by attending physicians to be eligible for participation in the study


Exclusion criteria: not reported
Interventions Group A: silodosin 4 mg twice daily
Group B: tamsulosin 0.2 mg once daily
Duration: 1 week
Outcomes
  • IPSS

  • Overactive Bladder Symptom Score

  • QoL rating scale

  • side effects

Notes Publication status: abstract (final publication status has not been clarified)

NCT01222650.

Methods Parallel, randomized, double‐blind, placebo‐controlled study
Participants Inclusion criteria: men with BPH and LUTS
Exclusion criteria: history of prostatectomy, intrapelvic radiation therapy, thermotherapy of prostate or prostatic hyperthermia, prostate cancer or suspected prostate cancer, any clinically relevant cardiovascular, hepatic or renal disorder
Interventions Group A: not reported
Group B: not reported
Duration: 12 weeks
Outcomes
  • change in IPSS total score from baseline

  • change in IPSS subscore

  • QoL score

  • Qmax from baseline

Notes Funding source: Kissei Pharmaceutical Co., Ltd
Publication status: NCT01222650 (final publication status has not been clarified)

Pawar 2015.

Methods Parallel, randomized study
Participants Inclusion criteria: men > 45 years with symptomatic BPH (increased daytime frequency, urgency and nocturia and or voiding symptoms (hesitancy, incomplete voiding, impaired stream or interruption of stream), increased daytime micturition, nocturia > 2, maximum flow rate < 15 mL/sec with a voided volume of ≥ 150 mL, PVR < 100 mL by abdominal US, IPSS > 13 points, international prostatic symptom bother score > 3 points
Exclusion criteria: not reported
Interventions Group A: silodosin 8 mg once daily
Group B: tamsulosin 0.2 mg once daily
Duration: 12 weeks
Outcomes
  • clinical determination of IPSS, Qmax index

  • Qmax (mL/s)

  • time to maximum flow rate

  • average flow rate

  • average flow time

  • PVR

  • prostate size by transabdominal ultrasonographically

  • adverse events

Notes Publication status: abstract (final publication status has not been clarified)

BPH: benign prostatic hyperplasia; IPSS: International Prostate Symptom Score; LUTS: lower urinary tract symptoms; PSA: prostate specific antigen; PVR: postvoid residual; Qmax; maximum flow rate; QoL: quality of life; US: ultrasound

Characteristics of ongoing studies [ordered by study ID]

CTRI/2013/10/004112.

Trial name or title A study to find out the medication which offers maximum health benefits with minimum spending by the patient among the three commonly used medications in the treatment of benign prostatic hyperplasia, a disease in aging men which results in bothersome urinary problems
Methods Randomized, parallel group trial
Participants Inclusion criteria
  • Men ≥ 45 years with BPH and associated LUTS

  • IPSS ≥ 8

  • QoL score ≥ 3

  • Qmax < 15 mL/s but > 4 mL/s with a voided volume of >100 mL

  • Willingness to give written informed consent and to comply with the study procedure, and available for regular follow‐up

Interventions Group A: silodosin oral, 8 mg once daily
Group B: tamsulosin oral, 0.4 mg once daily
Group C: alfuzosin oral, 10 mg once daily
Duration: 12 weeks
Outcomes Treatment success is defined as ≥ 30% improvement in IPSS from baseline and its maintenance over the study period
Starting date September 2013
Contact information manjunatha5ramaiah@gmail.com
Notes Funding source: none

JPRN‐UMIN000003609.

Trial name or title The effects of selective alpha‐1‐adrenergic receptor antagonists in patients with lower urinary tract symptoms caused by benign prostatic hyperplasia who failed to obtain sufficient efficacy by previous alpha‐1‐blockades. A comparison of naftopidil or silodosin
Methods Parallel, randomized
Participants Inclusion criteria
Men with LUTS caused by BPH who failed to obtain sufficient efficacy by naftopidil 50 mg and with IPSS of ≥ 9, a QoL score of ≥ 2, a prostate volume of ≥ 20 mL are eligible for enrolment
Interventions Group A: silodosin 8 mg
Group B: naftopidil 75 mg
Duration: not reported
Outcomes
  • IPSS

  • QoL score

  • blood pressure

  • uroflowmetry

Starting date May 2010
Contact information Email address was not given
Notes Funding source: none

BPH: benign prostatic hyperplasia; IPSS: International Prostate Symptom Score; LUTS: lower urinary tract symptoms; Qmax; maximum flow rate; QoL: quality of life

Differences between protocol and review

This review was based on a published protocol with differences as described here.

  • Types of studies: we included parallel, randomized trials as well as cross‐over designs. We revised the 'Types of studies' and 'Unit of analysis issues' section accordingly.

  • Types of participants: we redefined the type of participants as adult men aged 40 years and over. Given that alpha‐blockers are used to treat patients with all degrees of LUTS, we deleted the IPSS criterion under ‘Types of participants’.

  • Types of outcome measures: we renamed primary and secondary outcomes and added details in 'method and timing of outcome measurement' for all outcomes.

  • Assessment of risk of bias in included studies: we added details with regards to assessing risk of bias. We considered all outcomes susceptible to performance bias and assessed them in one group. We redefined subjective and objective outcomes for detection bias.

  • Subgroup and sensitivity analysis: we planned to perform subgroup and sensitivity analyses limited to the primary outcomes.

  • Appendix: we have summarized the search strategy for each database in Appendix 1.

Contributions of authors

JH Jung (JHJ): conception and study design, drafting the protocol, searching for trials, study selection, extracting data, assessing risk of bias, performing data analysis, interpretation of data, and drafting the review.

J Kim (JK): drafting the protocol, searching for trials, study selection, extracting data, assessing risk of bias, performing data analysis, and drafting the review.

R MacDonald (RM): study design, drafting the protocol, providing clinical and methodological advices on the review, and final approval.

B Reddy (BR): providing clinical and methodological advice on the review, and drafting the review.

MH Kim (MHK): creating search strategies, drafting the protocol, and searching for trials.

P Dahm (PD): conception and study design, providing clinical and methodological advice on the review, and final approval.

Sources of support

Internal sources

  • Yonsei University Wonju College of Medicine, Korea, South.

  • Minneapolis VA Medical Center, Minneapolis, Minnesota, USA.

  • University of Minnesota, Minneapolis, Minnesota, USA.

External sources

  • No sources of support provided, Other.

Declarations of interest

JHJ: none known

JK: none known

RM: none known

BR: none known

MHK: none known

PD: PD serves as Co‐ordinating Editor of Cochrane Urology. However, he was not involved in the editorial processing or decision‐making for this review. Other editors of Cochrane Urology managed the editorial process, including final sign‐off for this review.

New

References

References to studies included in this review

Chapple 2011 {published data only}

  1. Chapple CR, Montorsi F, Desgrandchamps F, Silodosin European Study Group. Efficacy of silodosin as compared with tamsulosin and placebo for the treatment of the signs and symptoms of benign prostatic hyperplasia. a multicentre, randomised, double‐blind, controlled trial. European Urology Supplements 2010;9(2):313. [DOI: ] [Google Scholar]
  2. Chapple CR, Montorsi F, Tammela TL, Wirth M, Koldewijn E, Fernandez Fernandez E, et al. 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. European Urology 2011;59(3):342‐52. [DOI: 10.1016/j.eururo.2010.10.046; PUBMED: 21109344] [DOI] [PubMed] [Google Scholar]
  3. Chapple CR, Montorsi F, Tammela Tl, Wirth M, Koldewijn E, Fernandez Fernandez E. 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. Urologia 2012;5:38‐42, 44‐5. [PUBMED: 23342615] [PubMed] [Google Scholar]
  4. EUCTR2005‐005665‐11‐GB. Evaluation of the efficacy and safety of silodosin vs. Tamsulosin and placebo in the treatment of the signs and symptoms of benign prostatic hyperplasia. Multicentre, randomised, double‐blind, controlled trial with an optional long‐term open‐label extension phase. apps.who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2005‐005665‐11‐GB (date first received 10 October 2017).
  5. NCT00359905. Evaluation of the Efficacy and Safety of Silodosin in the Treatment of the Signs and Symptoms of BPH. https://clinicaltrials.gov/ct2/show/NCT00359905?term=NCT00359905&rank=1 (date first received 10 October 2017).

Jung 2012 {published data only}

  1. Jung GW, Park SJ, Seo J. Effect of short‐term treatment with silodosin on ejaculatory function in sexually active men with benign prostate hyperplasia. Journal of Urology 2012; Vol. 187, issue 4 Supplement:e511. [DOI: ]
  2. Park SJ, Jung GW, Seo JH. Effect of short‐term treatment with silodosin on ejaculatory function in sexually active men with benign prostate hyperplasia. European Urology Supplements 2012;11(1):e744. [DOI: ] [Google Scholar]

Kawabe 2006a {published data only}

  1. Kawabe K, Yoshida M, Homma Y, Silodosin Clinical Study Group. Silodosin, a new alpha1A‐adrenoceptor‐selective antagonist for treating benign prostatic hyperplasia: results of a phase III randomized, placebo‐controlled, double‐blind study in Japanese men. BJU International 2006;98(5):1019‐24. [DOI: 10.1111/j.1464-410X.2006.06448.x; PUBMED: 16945121] [DOI] [PubMed] [Google Scholar]
  2. Yoshida M, Kawabe K, Homma Y. Silodosin, a new effective alpha 1A‐adrenoceptor selective antagonist for the treatment of benign prostatic hyperplasia: results of a phase 3 randomized, placebo‐controlled, double‐blind study. Journal of Urology 2005;173(4 Supplement):445. [Google Scholar]

Manjunatha 2016a {published data only}

  1. CTRI/2013/07/003805. A clinical study of the effects of the three licensed drugs alfuzosin, tamsulosin and silodosin in patients with benign prostatic hyperplasia, a disease predominantly causing urinary problems in aging men. apps.who.int/trialsearch/Trial2.aspx?TrialID=CTRI/2013/07/003805 (date first received 01 September 2017).
  2. Manjunatha R, Pundarikaksha HP, Madhusudhana HR, Amarkumar J, Hanumantharaju BK. A randomized, comparative, open‐label study of efficacy and tolerability of alfuzosin, tamsulosin and silodosin in benign prostatic hyperplasia. Indian Journal of Pharmacology 2016; Vol. 48, issue 2:134‐40. [DOI: 10.4103/0253-7613.178825; PUBMED: 27127315] [DOI] [PMC free article] [PubMed]

Marks 2009 {published data only}

  1. Gittelman MC, Marks LS, Hill LA, Volinn W, Hoel G. Efficacy and safety of silodosin in different age groups of men with symptoms of benign prostatic hyperplasia. Journal of the American Geriatrics Society 2009;57(Supplement s1):S122. [DOI: 10.1111/j.1532-5415.2009.02272.x] [DOI] [Google Scholar]
  2. Marks LS, Gittelman MC, Hill LA, Volinn W, Hoel G. Rapid efficacy of the highly selective α1A‐adrenoceptor antagonist silodosin in men with signs and symptoms of benign prostatic hyperplasia: pooled results of 2 phase 3 studies. Journal of Urology 2009;181(6):2634‐40. [DOI: 10.1016/j.juro.2009.02.034; PUBMED: 19371887] [DOI] [PubMed] [Google Scholar]
  3. Marks LS, Gittelman MC, Hill LA, Volinn W, Hoel G. Rapid efficacy of the highly selective α1A‐adrenoceptor antagonist silodosin in men with signs and symptoms of benign prostatic hyperplasia: pooled results of 2 phase 3 studies. Journal of Urology 2013;189(1 Suppl):S122‐8. [DOI: http://dx.doi.org/10.1016/j.juro.2012.11.020] [DOI] [PubMed] [Google Scholar]
  4. Marks LS, Gittelman MC, Hill LA, Volinn W, Hoel G. Rapid response of irritative and obstructive urinary symptoms of benign prostatic hyperplasia to treatment with silodosin in 2 randomized studies. Journal of Urology 2009;181(4):694. [DOI: ] [Google Scholar]
  5. NCT00224107. A New Drug for Benign Prostatic Hyperplasia (BPH) Compared With Placebo. clinicaltrials.gov/ct2/show/NCT00224107?term=NCT00224107&rank=1 (date first received 10 October 2017).
  6. NCT00224120. A new drug for benign prostatic hyperplasia (BPH) compared with placebo. clinicaltrials.gov/ct2/show/NCT00224120?term=NCT00224120&rank=1 (date first received 10 October 2017).
  7. NCT00224133. The evaluation of the safety of a new drug for benign prostatic hyperplasia used for 9 months. clinicaltrials.gov/ct2/show/NCT00224133?term=NCT00224133&rank=1 (date first received 10 October 2017).

Masuda 2012 {published data only}

  1. Masuda M, Jinza S, Masuko H, Asakura T, Hashiba T. Comparison of naftopidil and silodosin in the treatment of male lower urinary tract symptoms associated with benign prostatic hyperplasia: a randomized, crossover study. Acta Urologica Japonica 2012;58(12):671‐8. [PUBMED: 23328162] [PubMed] [Google Scholar]

Matsukawa 2016 {published data only}

  1. Matsukawa Y, Funahashi Y, Takai S, Majima T, Ogawa T, Narita H, et al. Comparison of silodosin and naftopidil for efficacy in the treatment of benign prostatic enlargement complicated by overactive bladder: a randomized, prospective study (SNIPER study). Journal of Urology 2017;197(2):452‐8. [DOI: 10.1016/j.juro.2016.08.111; PUBMED: 27615436] [DOI] [PubMed] [Google Scholar]

Miyakita 2010 {published data only}

  1. Miyakita H, Yokoyama E, Onodera Y, Utsunomiya T, Tokunaga M, Tojo T, et al. Short‐term effects of crossover treatment with silodosin and tamsulosin hydrochloride for lower urinary tract symptoms associated with benign prostatic hyperplasia. International Journal of Urology 2010;17(10):869‐75. [DOI: ] [DOI] [PubMed] [Google Scholar]

Natarajan 2015 {published data only}

  1. Natarajan B, Kalra Y. Silodosin versus tamsulosin in symptomatic benign prostatic hyperplasia‐Our experience. Journal Of Pharmacy 2015;5(6):8‐10. [DOI: 10.9790/XXXXXXXXX] [DOI] [Google Scholar]

NCT00793819 {unpublished data only}

  1. NCT00793819. A double‐blind, placebo‐controlled phase 2 study of silodosin 8 mg daily for the treatment of nocturia in men with benign prostatic hyperplasia. clinicaltrials.gov/ct2/show/NCT00793819?term=NCT00793819&rank=1 (date first received 01 September 2017).

Pande 2014 {published data only}

  1. CTRI/2014/01/004366. Evaluation of the effectiveness and safety of silodosin in comparison to tamsulosin in benign prostatic hyperplasia. ctri.nic.in/Clinicaltrials/pdf_generate.php?trialid=7666&EncHid=&modid=&compid=%27,%277666det%27 (date first received 10 October 2017).
  2. Pande S, Hazra A, Kundu AK. Evaluation of silodosin in comparison to tamsulosin in benign prostatic hyperplasia: a randomized controlled trial. Indian Journal of Pharmacology 2014;46(6):601‐7. [DOI: 10.4103/0253-7613.144912] [DOI] [PMC free article] [PubMed] [Google Scholar]

Shirakawa 2013 {published data only}

  1. JPRN‐UMIN000008331. A comparative study on the clinical effects of silodosin and naftopidil in Japanese patients with lower urinary tract symptoms associated with benign prostatic hyperplasia. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000008331 (date first received 10 October 2017).
  2. Shirakawa T, Haraguchi T, Matsumoto M, Morishita S, Minayoshi K, Miyazaki J, et al. A comparative study on the clinical effects of silodosin and naftopidil in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia. Urology 2010;76(3 Supplement):S106. [Google Scholar]
  3. Shirakawa T, Haraguchi T, Matsumoto Y, Takeda M, Morishita S, Minayoshi K, et al. A comparative study on the clinical effects of silodosin and naftopidil in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia. International Urogynecology Journal and Pelvic Floor Dysfunction 2011;22:S1209. [Google Scholar]
  4. Shirakawa T, Haraguchi T, Matsumoto Y, Takeda M, Morishita S, Minayoshi K, et al. A comparative study on the clinical effects of silodosin and naftopidil in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia (abstract number 887). Joint Meeting of the International Continence Society (ICS) and the International Urogynecological Association. 2010:23‐27.
  5. Shirakawa T, Haraguchi T, Shigemura K, Morishita S, Minayoshi K, Miyazaki J, et al. Silodosin versus naftopidil in Japanese patients with lower urinary tract symptoms associated with benign prostatic hyperplasia: a randomized multicenter study. International Journal of Urology 2013; Vol. 20, issue 9:903‐10. [DOI: 10.1111/iju.12055; PUBMED: 23252453] [DOI] [PubMed]
  6. Shirakawa T, Soga H, Haraguchi T, Takeda M, Morishita S, Sakai Y, et al. A comparative study on the clinical effects of silodosin and naftopidil in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia (abstract number 608). 38th Annual Meeting of the International Continence Society (ICS). 2008:20‐24.

Takeshita 2016 {published data only}

  1. JPRN‐UMIN000004918. Silodosin 4mg versus tamsulosin 0.2mg once daily; randomized crossover study. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000004918 (date first received 25 October 2017).
  2. Takeshita H, Moriyama S, Arai Y, Washino S, Saito K, Chiba K, et al. Randomized crossover comparison of the short‐term efficacy and safety of single half‐dose silodosin and tamsulosin hydrochloride in men with lower urinary tract symptoms secondary to benign prostatic hyperplasia. LUTS: Lower Urinary Tract Symptoms 2016;8(1):38‐43. [DOI: 10.1111/luts.12106] [DOI] [PubMed] [Google Scholar]

Watanabe 2011 {published data only}

  1. Watanabe T, Ozono S, Kageyama S. A randomized crossover study comparing patient preference for tamsulosin and silodosin in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia. Journal of International Medical Research 2011;39(1):129‐42. [PUBMED: 21672315 ] [PubMed] [Google Scholar]
  2. Watanabe T, Ozono S, Kageyama S. Erratum to A randomized crossover study comparing patient preference for tamsulosin and silodosin in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia (J Int Med Res 2011;39:129‐42). Journal of International Medical Research 2011;39(3):1122. [PubMed] [Google Scholar]

Yamaguchi 2013 {published data only}

  1. Takahashi S, Yamaguchi K. Treatment of benign prostatic hyperplasia and aging: impacts of alpha‐1 blockers on sexual function. Journal of Men's Health 2011;8(Supplement 1):S25‐8. [Google Scholar]
  2. Yamaguchi K, Aoki Y, Yoshikawa T, Hachiya T, Saito T, Takahashi S. Silodosin versus naftopidil for the treatment of benign prostatic hyperplasia: a multicenter randomized trial. International Journal of Urology 2013;20(12):1234‐8. [DOI: 10.1111/iju.12160; PUBMED: 23731168] [DOI] [PubMed] [Google Scholar]

Yamanishi 2011 {published data only}

  1. Yamanishi T, Sakakibara R, Uchiyama T. Comparison of the effects of silodosin and tamsulosin for the treatment of benign prostatic hyperplasia (Abstract number 290). 41st Annual Meeting of the International Continence Society. 2011.

Yokoyama 2011 {published data only}

  1. Yokoyama T, Hara R, Fukumoto K, Fujii T, Jo Y, Miyaji Y, et al. Effects of three types of alpha‐1 adrenoceptor blocker on lower urinary tract symptoms and sexual function in males with benign prostatic hyperplasia. International Journal of Urology 2011;18(3):225‐30. [DOI: 10.1111/j.1442-2042.2010.02708.x; PUBMED: 21272091] [DOI] [PubMed] [Google Scholar]

Yokoyama 2012 {published data only}

  1. Yokoyama T, Fukumoto K, Hara R, Fujii T, Jo Y, Miyaji Y, et al. Comparison of different a1‐adrenoceptor antagonists, tamsulosin hydrochloride and silodosin for treatment of male lower urinary tract symptoms: a prospective randomized crossover study. International Journal of Urology 2011;18(5):404. [DOI: 10.1111/j.1442-2042.2011.02758.x] [DOI] [Google Scholar]
  2. Yokoyama T, Hara R, Fujii T, Jo Y, Miyaji Y, Nagai A. Comparison of two different α1‐adrenoceptor antagonists, tamsulosin and silodosin, in the treatment of male lower urinary tract symptoms suggestive of benign prostatic hyperplasia: a prospective randomized crossover study. LUTS: Lower Urinary Tract Symptoms 2012;4(1):14‐8. [DOI: 10.1111/j.1757-5672.2011.00099.x; PUBMED: 26676453] [DOI] [PubMed] [Google Scholar]

Yu 2011 {published data only}

  1. Yu HJ, Lin AT, Yang SS, Tsui KH, Wu HC, Cheng CL, et al. Non‐inferiority of silodosin to tamsulosin in treating patients with lower urinary tract symptoms (LUTS) associated with benign prostatic hyperplasia (BPH). BJU International 2011;108(11):1843‐8. [DOI: 10.1111/j.1464-410X.2011.10233.x; PUBMED: 21592295 ] [DOI] [PubMed] [Google Scholar]

References to studies excluded from this review

Abramowicz 2009 {published data only}

  1. Abramowicz M, Zuccotti G, Pflomm JM, Daron SM, Houst BM, Zanone CE, et al. Silodosin (Rapaflo) for benign prostatic hyperplasia. Medical Letter on Drugs and Therapeutics 2009;51(1303):3‐4. [PubMed] [Google Scholar]

Alcántara Montero 2016 {published data only}

  1. Alcántara Montero A. Silodosin in the treatment of lower urinary tract symptoms due to benign prostatic hyperplasia: The standard approach and «real life» use [Silodosina en el tratamiento de los síntomas del tracto urinario inferior en el varón por hiperplasia benigna de próstata: enfoque estándar y uso en la «vida real»]. Revista Mexicana de Urología 2016;76(6):352‐9. [DOI: 10.1016/j.uromx.2016.09.005] [DOI] [Google Scholar]
  2. Alcántara Montero A. The role of alpha blockers in the treatment of lower urinary tract symptoms/benign prostatic hyperplasia: are all the same? Silodosin in "real life". Semergen 2016;42 Suppl 2:1‐9. [DOI: 10.1016/S1138-3593(16)30245-3; PUBMED: 27889014 ] [DOI] [PubMed] [Google Scholar]

Araki 2013 {published data only}

  1. Araki T, Monden K, Araki M. Comparison of 7 α1‐adrenoceptor antagonists in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia: a short‐term crossover study. Acta Medica Okayama 2013;67(4):245‐51. [PUBMED: 23970323] [DOI] [PubMed] [Google Scholar]

Boeri 2016 {published data only}

  1. Boeri L, Capogrosso P, Ventimiglia E, Croce G, Moretti D, Scano R, et al. Clinically meaningful improvements in LUTS/BPH severity in men treated with silodosin plus Serenoa repens or silodosin alone. Journal of Urology 2016;195(4):e464. [DOI: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Cakiroglu 2016 {published data only}

  1. Cakiroglu B, Hazar İ, Sinanoglu O, Arda E, Ekici S. Comparison of transurethral incision and silodosin in LUTS patients in terms of retrograde ejaculation. European Urology Supplements 2016;5(15):e1236. [DOI: 10.1016/S1569-9056(16)15063-8] [DOI] [Google Scholar]

Chapple 2009 {published data only}

  1. Chapple CR, Silodosin European Study Group. Silodosin: a new α‐adrenoceptor antagonist highly selective for the lower urinary tract. European Urology Supplements 2009;8(4):238. [DOI: ] [Google Scholar]

Curran 2011 {published data only}

  1. Curran MP. Silodosin: treatment of the signs and symptoms of benign prostatic hyperplasia. Drugs 2011;71(7):897‐907. [DOI: 10.2165/11204780-000000000-00000] [DOI] [PubMed] [Google Scholar]

JPRN‐UMIN000007917 {unpublished data only}

  1. JPRN‐UMIN000007917. Efficacy of silodosin in benign prostatic hyperplasia with overactive bladder. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000007917 (date first received 01 September 2017).

Kawabe 2006b {published data only}

  1. Kawabe K, Yoshida M, Arakawa S, Takeuchi H. Long‐term evaluation of silodosin, a new α1A‐adrenoceptor selective antagonist for the treatment of benign prostatic hyperplasia: phase III long‐term study. Japanese Journal of Urological Surgery 2006;19:153‐64. [Google Scholar]

Kobayashi 2008 {published data only}

  1. Kobayashi K, Masumori N, Hisasue SI, Kato R, Hashimoto K, Itoh N, et al. Inhibition of seminal emission Is the main cause of anejaculation induced by a new highly selective α1A‐Blocker in normal volunteers. Journal of Sexual Medicine 2008;5(9):2185‐90. [DOI: 10.1111/j.1743-6109.2008.00779.x] [DOI] [PubMed] [Google Scholar]

Kobayashi 2009 {published data only}

  1. Kobayashi K, Masumori N, Kato R, Hisasue S, Furuya R, Tsukamoto T. Orgasm is preserved regardless of ejaculatory dysfunction with selective alpha 1A‐blocker administration. International Journal of Impotence Research 2009;21(5):306‐10. [DOI: 10.1038/ijir.2009.27] [DOI] [PMC free article] [PubMed] [Google Scholar]

Manjunatha 2016b {published data only}

  1. Manjunatha R, Pundarikaksha HP, Madhusudhana HR. A prospective analysis of the cost‐effectiveness of alfuzosin, tamsulosin and silodosin for 12 weeks in benign prostatic hyperplasia. International Journal of Basic & Clinical Pharmacology 2016;5(6):2481‐7. [DOI: 10.18203/2319-2003.ijbcp20164109] [DOI] [Google Scholar]

Matsukawa 2012 {published data only}

  1. Matsukawa Y, Hattori R, Mazima T, Yamamoto T, Yoshino Y, Gotoh M. Is combination therapy with an anticholinergic agent and an alpha1‐adrenoceptor antagonist useful as first‐line treatment in patients with benign prostatic hyperplasia complicated by overactive bladder? A randomized, prospective, comparative study using a urodynamic study. European Urology Supplements 2012;11(1):e753. [DOI: ] [Google Scholar]

Matsukawa 2017 {published data only}

  1. Matsukawa Y, Takai S, Funahashi Y, Majima T, Kato M, Yamamoto T, et al. Effects of withdrawing alpha‐1 blocker from the combination therapy with alpha‐1 blocker and 5‐alpha‐reductase inhibitor in patients with lower urinary tract symptoms suggestive of benign prostatic hyperplasia: a prospective and comparative trial using urodynamics. Journal of Urology 2017 [Epub ahead of print]; Vol. May. [DOI: 10.1016/j.juro.2017.05.031; PUBMED: 28499730 ] [DOI] [PubMed]

Michel 2011 {published data only}

  1. Michel MC, Casi M, Antonellini A. Silodosin consistently improves nocturia in men with LUTS suggestive of BPH. Analysis of three phase III placebo‐controlled studies. European Urology Supplements 2011;10(2):121‐2. [DOI: ] [Google Scholar]

Montorsi 2010 {published data only}

  1. Montorsi F. Profile of silodosin. European Urology Supplements 2010;9(4):491‐5. [DOI: 10.1016/j.eursup.2010.04.001] [DOI] [PubMed] [Google Scholar]

Montorsi 2013 {published data only}

  1. Montorsi F. Profile of silodosin. Urologiia 2013;2:112‐4, 116‐7. [PUBMED: 23789376 ] [PubMed] [Google Scholar]

Prescrire Int 2011 {published data only}

  1. Prescribe International. Silodosin. Prescrire International 2011;20(118):180. [PubMed] [Google Scholar]

Prescrire Int 2012 {published data only}

  1. Prescribe International. Silodosin: erectile and ejaculatory disorders. Prescrire International 2012; Vol. 21, issue 127:129. [PubMed]

Roehrborn 2009 {published data only}

  1. Roehrborn CG, Lepor H, Kaplan SA. Retrograde ejaculation induced by silodosin is the result of relaxation of smooth musculature in the male uro‐genital tracts and is associated with greater urodynamic and symptomatic improvements in men LUTS secondary to BPH. Journal of Urology 2009;181(4):694‐5. [DOI: ] [Google Scholar]

Yoshida 2017 {published data only}

  1. Yoshida M, Origasa H, Seki N. Comparison of silodosin versus tadalafil in patients with lower urinary tract symptoms associated with benign prostatic hyperplasia. Lower Urinary Tract Symptoms 2017 [Epub ahead of print]; Vol. April. [DOI: 10.1111/luts.12177; 28439987] [DOI] [PubMed]

Yoshihisa 2012 {published data only}

  1. Yoshihisa M, Ryohei H, Shun T, Tokunori Y, Momokazu G. The effect of combination therapy with an anticholinergic agent and an + 1‐adrenoceptor antagonist in patients with benign prostatic hyperplasia complicated by an overactive bladder: a randomized, prospective, comparative, urodynamic study. International Journal of Urology 2012;19:180. [DOI: 10.1111/j.1442-2042.2012.03167.x] [DOI] [Google Scholar]

Zhou 2011 {published data only}

  1. Zhou Y, Sun PH, Liu YW, Zhao X, Meng L, Cui YM. Safety and pharmacokinetic studies of silodosin, a new alpha(1A)‐adrenoceptor selective antagonist, in healthy Chinese male subjects. Biological and Pharmaceutical Bulletin 2011;34(8):1240‐5. [PUBMED: 21804212 ] [DOI] [PubMed] [Google Scholar]

References to studies awaiting assessment

CTRI/2010/091/000526 {unpublished data only}

  1. CTRI/2010/091/000526. A clinical trial to study the effects of silodosin in patients with benign prostatic hyperplasia. apps.who.int/trialsearch/Trial2.aspx?TrialID=CTRI/2010/091/000526 (date first received 01 September 2017).

Devana 2014 {published data only}

  1. Devana SK, Singh SK, Ravi Mohan SM, Mandal AK. A prospective randomized study on the short term efficacy of three alpha 1 adrenergic blockers in patients with symptomatic benign prostatic hyperplasia. Indian Journal of Urology 2014; Vol. 30:S96.

Jha 2015 {published data only}

  1. Jha SK, Kumar A, Vasudeva P, Kumar R, Singh H, Kumar G. Prospective randomised comparative study of safety and efficacy of silodosin versus tamsulosin in the medical management of benign prostatic enlargement causing lower urinary tract symptoms. Indian Journal of Urology 2015; Vol. 31:S69.

JPRN‐UMIN000003125 {unpublished data only}

  1. JPRN‐UMIN000003125. CLSS‐based assessment of alfa‐adrenoceptor blockers for male LUTS (CLAM‐STUDY). apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000003125 (date first received 01 September 2017).

JPRN‐UMIN000005151 {unpublished data only}

  1. JPRN‐UMIN000005151. The clinical study for evaluating the early efficacy and safety of alpha‐1‐adrenoreceptor antagonists in patient with benign prostatic hyperplasia. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000005151 (date first received 01 September 2017).

JPRN‐UMIN000008538 {unpublished data only}

  1. JPRN‐UMIN000008538. Comparison of the long‐term efficacy of silodosin and tamsulosin for the treatment of benign prostatic hyperplasia. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000008538 (date first received 01 September 2017).

JPRN‐UMIN000011556 {unpublished data only}

  1. JPRN‐UMIN000011556. Comparative study of early efficacy of once‐daily dosing between silodosin 4mg and tamsulosin 0.2mg for voiding symptoms associated with benign prostatic hyperplasia. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000011556 (date first received 01 September 2017).

Mandal 2013 {published data only}

  1. Mandal A, Devana S, Singh S, Mavuduru R. Evaluation of short term efficacy of different alfa adrenergic blockers on LUTS in patients with symptomatic BPH. Journal of Endourology 2013; Vol. 27:A75‐76.

Manohar 2014 {published data only}

  1. Manohar CS, Kamath AJ. Prospective randomize study of clinical out come of management of luts in BPH using silodosin/tamsulosin/alfuzosin. Indian Journal of Urology 2014; Vol. 30:S94‐5.

Miyamae 2011 {published data only}

  1. Miyamae K, Kitani K, Miyamoto K, Nakakuma K, Yamamoto T, Maehara A, et al. Investigation of the early effects of 1‐blockers in a comparative study focusing on patient evaluation. International Urogynecology Journal and Pelvic Floor Dysfunction 2011; Vol. 22:S253‐4.

NCT01222650 {unpublished data only}

  1. NCT01222650. A randomized, double‐blind, placebo‐controlled, multicentre study of KSO‐0400 in BPH patients with LUTS. clinicaltrials.gov/ct2/show/NCT01222650?term=NCT01222650&rank=1 (date first received 01 September 2017).

Pawar 2015 {published data only}

  1. Pawar DS, Kumar A, Singh R, Singh SK. Comparative evaluation of silodosin and tamsulosin in treatment of patients with lower urinary tract symptoms with benign prostatic hyperplasia. Indian Journal of Urology 2015; Vol. 31:S137.

References to ongoing studies

CTRI/2013/10/004112 {unpublished data only}

  1. CTRI/2013/10/004112. A study to find out the medication which offers maximum health benefits with minimum spending by the patient among the three commonly used medications in the treatment of benign prostatic hyperplasia, a disease in aging men which results in bothersome urinary problems. apps.who.int/trialsearch/Trial2.aspx?TrialID=CTRI/2013/10/004112 (date first received 01 September 2017).

JPRN‐UMIN000003609 {unpublished data only}

  1. JPRN‐UMIN000003609. The effects of selective alpha‐1‐adrenergic receptor antagonists in patients with lower urinary tract symptoms caused by benign prostatic hyperplasia who failed to obtain sufficient efficacy by previous alpha‐1‐blockades. A comparison of naftopidil or silodosin. apps.who.int/trialsearch/Trial2.aspx?TrialID=JPRN‐UMIN000003609 (date first received 01 September 2017).

Additional references

AUA Practice Guidelines Committee 2003

  1. AUA Practice Guidelines Committee. AUA guideline on management of benign prostatic hyperplasia (2003). Chapter 1: Diagnosis and treatment recommendations. Journal of Urology 2003;170(2 Pt 1):530‐47. [DOI] [PubMed] [Google Scholar]

Barry 1992

  1. Barry MJ, Fowler FJ Jr, O'Leary MP, Bruskewitz RC, Holtgrewe HL, Mebust WK, et al. The American Urological Association symptom index for benign prostatic hyperplasia. The Measurement Committee of the American Urological Association. Journal of Urology 1992;148(5):1549‐57; discussion 1564. [DOI] [PubMed] [Google Scholar]

Barry 1995

  1. Barry MJ, Williford WO, Chang Y, Machi M, Jones KM, Walker‐Corkery E, et al. Benign prostatic hyperplasia specific health status measures in clinical research: how much change in the American Urological Association symptom index and the benign prostatic hyperplasia impact index is perceptible to patients?. Journal of Urology 1995;154(5):1770‐4. [DOI] [PubMed] [Google Scholar]

Barry 1997

  1. Barry MJ, Fowler FJ Jr, Bin L, Pitts JC 3rd, Harris CJ, Mulley AG Jr. The natural history of patients with benign prostatic hyperplasia as diagnosed by North American urologists. Journal of Urology 1997;157(1):10‐4; discussion 14‐5. [PubMed] [Google Scholar]

Brasure 2016

  1. Brasure M, MacDonald R, Dahm P, Olson CM, Nelson VA, Fink HA, et al. AHRQ comparative effectiveness reviews. Newer Medications for Lower Urinary Tract Symptoms Attributed to Benign Prostatic Hyperplasia: a Review. Rockville (MD): Agency for Healthcare Research and Quality (US), 2016. [PubMed] [Google Scholar]

Castiglione 2014

  1. Castiglione F, Benigni F, Briganti A, Salonia A, Villa L, Nini A, et al. Naftopidil for the treatment of benign prostate hyperplasia: a systematic review. Current Medical Research and Opinion 2014;30(4):719‐32. [DOI] [PubMed] [Google Scholar]

Cornu 2010

  1. Cornu JN, Cussenot O, Haab F, Lukacs B. A widespread population study of actual medical management of lower urinary tract symptoms related to benign prostatic hyperplasia across Europe and beyond official clinical guidelines. European Urology 2010;58(3):450‐6. [DOI] [PubMed] [Google Scholar]

Covidence [Computer program]

  1. Veritas Health Innovation. Covidence. Version accessed 17 November 2017. Melbourne, Australia: Veritas Health Innovation, 2013.

Crawford 2006

  1. Crawford ED, Wilson SS, McConnell JD, Slawin KM, Lieber MC, Smith JA, et al. Baseline factors as predictors of clinical progression of benign prostatic hyperplasia in men treated with placebo. Journal of Urology 2006;175(4):1422‐6; discussion 1426‐7. [DOI] [PubMed] [Google Scholar]

Cui 2012

  1. Cui Y, Zong H, Zhang Y. The efficacy and safety of silodosin in treating BPH: a systematic review and meta‐analysis. International Urology and Nephrology 2012;44(6):1601‐9. [DOI] [PubMed] [Google Scholar]

Dahm 2016

  1. Dahm P, Brasure M, MacDonald R, Olson CM, Nelson VA, Fink HA, et al. Comparative effectiveness of newer medications for lower urinary tract symptoms attributed to benign prostatic hyperplasia: a systematic review and meta‐analysis. European Urology 2016 Oct 4 [Epub ahead of print]. [DOI: 10.1016/j.eururo.2016.09.032] [DOI] [PMC free article] [PubMed]

Deeks 2011

  1. Deeks JJ, Higgins JP, Altman DG, editor(s). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Ding 2013

  1. Ding H, Du W, Hou ZZ, Wang HZ, Wang ZP. Silodosin is effective for treatment of LUTS in men with BPH: a systematic review. Asian Journal of Andrology 2013;15(1):121‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]

Djavan 1999

  1. Djavan B, Marberger M. A meta‐analysis on the efficacy and tolerability of alpha1‐adrenoceptor antagonists in patients with lower urinary tract symptoms suggestive of benign prostatic obstruction. European Urology 1999;36(1):1‐13. [DOI] [PubMed] [Google Scholar]

EAU 2017

  1. European Association of Urology. Treatment of non‐neurogenic male LUTS. uroweb.org/guideline/treatment‐of‐non‐neurogenic‐male‐luts/ (date first received 17 August 2017).

Egan 2016

  1. Egan KB. The epidemiology of benign prostatic hyperplasia associated with lower urinary tract symptoms: prevalence and incident rates. Urologic Clinics of North America 2016;43(3):289‐97. [DOI] [PubMed] [Google Scholar]

EndNote [Computer program]

  1. Clarivate Analytics. EndNote. Version 7.5. Clarivate Analytics, 2016.

Fusco 2016

  1. Fusco F, Palmieri A, Ficarra V, Giannarini G, Novara G, Longo N, et al. Alpha1‐blockers improve benign prostatic obstruction in men with lower urinary tract symptoms: a systematic review and meta‐analysis of urodynamic studies. European Urology 2016;69(6):1091‐101. [DOI] [PubMed] [Google Scholar]

Gacci 2014

  1. Gacci M, Ficarra V, Sebastianelli A, Corona G, Serni S, Shariat SF, et al. Impact of medical treatments for male lower urinary tract symptoms due to benign prostatic hyperplasia on ejaculatory function: a systematic review and meta‐analysis. Journal of Sexual Medicine 2014;11(6):1554‐66. [DOI] [PubMed] [Google Scholar]

GRADEpro GDT 2015 [Computer program]

  1. McMaster University (developed by Evidence Prime). GRADEpro GDT 2015. Version accessed 17 November 2017. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.

Guyatt 2008

  1. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck‐Ytter Y, Schünemann HJ, GRADE Working Group. GRADE: what is "quality of evidence" and why is it important to clinicians?. BMJ (Clinical Research Ed.) 2008;336(7651):995‐8. [DOI: 10.1136/bmj.39490.551019.BE] [DOI] [PMC free article] [PubMed] [Google Scholar]

Guyatt 2011a

  1. Guyatt GH, Oxman AD, Kunz R, Brozek J, Alonso‐Coello P, Rind D, et al. GRADE guidelines 6. Rating the quality of evidence‐‐imprecision. Journal of Clinical Epidemiology 2011;64(12):1283‐93. [DOI] [PubMed] [Google Scholar]

Guyatt 2011b

  1. Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction‐GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383‐94. [DOI: 10.1016/j.jclinepi.2010.04.026] [DOI] [PubMed] [Google Scholar]

Higgins 2002

  1. Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta‐analysis. Statistics in Medicine 2002;21(11):1539‐58. [DOI: 10.1002/sim.1186] [DOI] [PubMed] [Google Scholar]

Higgins 2003

  1. Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ (Clinical Research Ed.) 2003;327(7414):557‐60. [DOI: 10.1136/bmj.327.7414.557] [DOI] [PMC free article] [PubMed] [Google Scholar]

Higgins 2011a

  1. Higgins JPT, Deeks JJ (editors). Chapter 7: Selecting studies and collecting data. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011b

  1. Higgins JP, Altman DG, Sterne JAC, editor(s). Chapter 8: Assessing risk of bias in included studies. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011c

  1. Higgins JP, Deeks JJ, Altman DG, editor(s). Chapter 16: Special topics in statistics. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Jaeschke 1989

  1. Jaeschke R, Singer J, Guyatt GH. Measurement of health status. Ascertaining the minimal clinically important difference. Controlled Clinical Trials 1989;10(4):407‐15. [DOI] [PubMed] [Google Scholar]

Johnston 2010

  1. Johnston BC, Thorlund K, Schunemann HJ, Xie F, Murad MH, Montori VM, et al. Improving the interpretation of quality of life evidence in meta‐analyses: the application of minimal important difference units. Health and Quality of Life Outcomes 2010;8:116. [DOI] [PMC free article] [PubMed] [Google Scholar]

Johnston 2013

  1. Johnston BC, Patrick DL, Busse JW, Schünemann HJ, Agarwal A, Guyatt GH. Patient‐reported outcomes in meta‐analyses‐‐Part 1: assessing risk of bias and combining outcomes. Health and Quality of Life Outcomes 2013;11:109. [DOI] [PMC free article] [PubMed] [Google Scholar]

Juliao 2012

  1. Juliao AA, Plata M, Kazzazi A, Bostanci Y, Djavan B. American Urological Association and European Association of Urology guidelines in the management of benign prostatic hypertrophy: revisited. Current Opinion in Urology 2012;22(1):34‐9. [DOI] [PubMed] [Google Scholar]

Keehn 2016

  1. Keehn A, Taylor J, Lowe FC. Phytotherapy for benign prostatic hyperplasia. Curr Urology Reports 2016;17(7):53. [DOI] [PubMed] [Google Scholar]

Kozminski 2015

  1. Kozminski MA, Wei JT, Nelson J, Kent DM. Baseline characteristics predict risk of progression and response to combined medical therapy for benign prostatic hyperplasia (BPH). BJU International 2015;115(2):308‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]

Lepor 2007

  1. Lepor H. Alpha blockers for the treatment of benign prostatic hyperplasia. Reviews in Urology 2007;9(4):181‐90. [PMC free article] [PubMed] [Google Scholar]

Liberati 2009

  1. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta‐analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Medicine 2009;6(7):e1000100. [DOI: 10.1371/journal.pmed.1000100] [DOI] [PMC free article] [PubMed] [Google Scholar]

MacDonald 2005

  1. MacDonald R, Wilt TJ. Alfuzosin for treatment of lower urinary tract symptoms compatible with benign prostatic hyperplasia: a systematic review of efficacy and adverse effects. Urology 2005;66(4):780‐8. [DOI] [PubMed] [Google Scholar]

Martin 2014

  1. Martin S, Lange K, Haren MT, Taylor AW, Wittert G. Risk factors for progression or improvement of lower urinary tract symptoms in a prospective cohort of men. Journal of Urology 2014;191(1):130‐7. [DOI] [PubMed] [Google Scholar]

McConnell 2003

  1. McConnell JD, Roehrborn CG, Bautista OM, Andriole GL Jr, Dixon CM, Kusek JW, et al. Medical Therapy of Prostatic Symptoms (MTOPS) Research Group. The long‐term effect of doxazosin, finasteride, and combination therapy on the clinical progression of benign prostatic hyperplasia. New England Journal of Medicine 2003;349(25):2387‐98. [DOI] [PubMed] [Google Scholar]

McVary 2011

  1. McVary KT, Roehrborn CG, Avins AL, Barry MJ, Bruskewitz RC, Donnell RF, et al. Update on AUA guideline on the management of benign prostatic hyperplasia. Journal of Urology 2011;185(5):1793‐803. [DOI] [PubMed] [Google Scholar]

Milani 2005

  1. Milani S, Djavan B. Lower urinary tract symptoms suggestive of benign prostatic hyperplasia: latest update on alpha‐adrenoceptor antagonists. BJU International 2005;95 Suppl 4:29‐36. [DOI] [PubMed] [Google Scholar]

Minneman 1994

  1. Minneman KP, Esbenshade TA. Alpha 1‐adrenergic receptor subtypes. Annual Review of Pharmacology and Toxicology 1994;34:117‐33. [DOI] [PubMed] [Google Scholar]

Nair 2016

  1. Nair SM, Pimentel MA, Gilling PJ. A review of laser treatment for symptomatic BPH (benign prostatic hyperplasia). Current Urology Reports 2016;17(6):45. [DOI] [PubMed] [Google Scholar]

Netto 1999

  1. Netto NR Jr, Lima ML, Netto MR, D'Ancona CA. Evaluation of patients with bladder outlet obstruction and mild international prostate symptom score followed up by watchful waiting. Urology 1999;53(2):314‐6. [DOI] [PubMed] [Google Scholar]

Novara 2013

  1. Novara G, Tubaro A, Sanseverino R, Spatafora S, Artibani W, Zattoni F, et al. Systematic review and meta‐analysis of randomized controlled trials evaluating silodosin in the treatment of non‐neurogenic male lower urinary tract symptoms suggestive of benign prostatic enlargement. World Journal of Urology 2013;31(4):997‐1008. [DOI] [PubMed] [Google Scholar]

Novara 2015

  1. Novara G, Chapple CR, Montorsi F. 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. BJU International 2015;115(5):802‐14. [DOI] [PubMed] [Google Scholar]

Oelke 2012

  1. Oelke M, Giuliano F, Mirone V, Xu L, Cox D, Viktrup L. Monotherapy with tadalafil or tamsulosin similarly improved lower urinary tract symptoms suggestive of benign prostatic hyperplasia in an international, randomised, parallel, placebo‐controlled clinical trial. European Urology 2012;61(5):917‐25. [DOI] [PubMed] [Google Scholar]

Osman 2012

  1. Osman NI, Chapple CR, Cruz F, Desgrandchamps F, Llorente C, Montorsi F. Silodosin: a new subtype selective alpha‐1 antagonist for the treatment of lower urinary tract symptoms in patients with benign prostatic hyperplasia. Expert Opinion on Pharmacotherapy 2012;13(14):2085‐96. [DOI] [PubMed] [Google Scholar]

Preston 2000

  1. Preston RA, Materson BJ, Reda DJ, Williams DW. Placebo‐associated blood pressure response and adverse effects in the treatment of hypertension: observations from a Department of Veterans Affairs Cooperative Study. Archives of Internal Medicine 2000;160(10):1449‐54. [DOI] [PubMed] [Google Scholar]

RevMan 2014 [Computer program]

  1. Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Roehrborn 2008

  1. Roehrborn CG. Pathology of benign prostatic hyperplasia. International Journal of Impotence Research 2008;20 Suppl 3:S11‐8. [DOI] [PubMed] [Google Scholar]

Rosenzweig 1993

  1. Rosenzweig P, Brohier S, Zipfel A. The placebo effect in healthy volunteers: influence of experimental conditions on the adverse events profile during phase I studies. Clinical Pharmacology and Therapeutics 1993;54(5):578‐83. [DOI] [PubMed] [Google Scholar]

Schilit 2009

  1. Schilit S, Benzeroual KE. Silodosin: a selective alpha1A‐adrenergic receptor antagonist for the treatment of benign prostatic hyperplasia. Clinical Therapeutics 2009;31(11):2489‐502. [DOI] [PubMed] [Google Scholar]

Schulman 2003

  1. Schulman CC. Lower urinary tract symptoms/benign prostatic hyperplasia: minimizing morbidity caused by treatment. Urology 2003;62(3 Suppl 1):24‐33. [DOI] [PubMed] [Google Scholar]

Schünemann 2011

  1. Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and ‘Summary of findings' tables. In: Higgins JP, Green S, editor(s), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Seki 2015

  1. Seki N, Takahashi R, Yamaguchi A, Ito K, Takayama K, Nanri K, et al. Non‐inferiority of silodosin 4 mg once daily to twice daily for storage symptoms score evaluated by the International Prostate Symptom Score in Japanese patients with benign prostatic hyperplasia: a multicenter, randomized, parallel‐group study. International Journal of Urology 2015;22(3):311‐6. [DOI] [PubMed] [Google Scholar]

Sterne 2011

  1. Sterne JAC, Egger M, Moher D (editors). Chapter 10: Addressing reporting biases. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Intervention. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Van Kerrebroec 2002

  1. Kerrebroec P, Jardin A, Cangh P, Laval KU, ALFORTI Study Group. Long‐term safety and efficacy of a once‐daily formulation of alfuzosin 10 mg in patients with symptomatic benign prostatic hyperplasia: open‐label extension study. European Urology 2002;41(1)(1):54‐60; discussion 60‐1. [DOI] [PubMed] [Google Scholar]

Wilt 2006

  1. Wilt TJ, MacDonald R. Doxazosin in the treatment of benign prostatic hypertrophy: an update. Clinical Interventions in Aging 2006;1(4):389‐401. [DOI] [PMC free article] [PubMed] [Google Scholar]

Yap 2009

  1. Yap TL, Brown C, Cromwell DA, Meulen J, Emberton M. The impact of self‐management of lower urinary tract symptoms on frequency‐volume chart measures. BJU International 2009;104(8):1104‐8. [DOI] [PubMed] [Google Scholar]

Yoo 2012

  1. Yoo TK, Cho HJ. Benign prostatic hyperplasia: from bench to clinic. Korean Journal of Urology 2012;53(3):139‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]

Yoshida 2007

  1. Yoshida M, Homma Y, Kawabe K. Silodosin, a novel selective alpha 1A‐adrenoceptor selective antagonist for the treatment of benign prostatic hyperplasia. Expert Opinion on Investigational Drugs 2007;16(12):1955‐65. [DOI] [PubMed] [Google Scholar]

Yuan 2015

  1. Yuan JQ, Mao C, Wong SYS, Yang ZY, Fu XH, Dai XY, et al. Comparative effectiveness and safety of monodrug therapies for lower urinary tract symptoms associated with benign prostatic hyperplasia a network meta‐analysis. Medicine 2015;94(27):7. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Cochrane Database of Systematic Reviews are provided here courtesy of Wiley

RESOURCES