Next Article in Journal
Multiple Adverse Drug Reactions to Calcineurin Inhibitors in a Renal Transplant Patient
Next Article in Special Issue
Rethinking the Role of Saw Palmetto Extract for Men with Lower Urinary Tract Symptoms in North America
Previous Article in Journal / Special Issue
Serenoa Repens (Saw Palmetto) for Lower Urinary Tract Symptoms (LUTS): The Evidence for Efficacy and Safety of Lipidosterolic Extracts. Part II
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Serenoa Repens (Saw Palmetto) for Lower Urinary Tract Symptoms (LUTS): The Evidence for Efficacy and Safety of Lipidosterolic Extracts. Part III

Community Practice of Hematology, Oncology and Internal Medicine, Focus on Prostate Cancer and Prostate Diseases, Nevada City, CA 95959, USA
Uro 2021, 1(3), 155-179; https://doi.org/10.3390/uro1030017
Submission received: 4 July 2021 / Revised: 28 July 2021 / Accepted: 3 August 2021 / Published: 6 August 2021

Abstract

:
Parts I and II of this three-part series indicated how a global review of both English-language and non-English language papers, plus a focus on a lipidosterolic extract of Serenoa repens (LSESr) having a standardized fatty acid profile, have together engendered new insights about the biological activity of LSESr vs. LUTS. In this last part, data from the world literature is presented that confirms that LSESr efficacy is the predominant finding in clinical trials. Despite two placebo-controlled clinical trials performed in the U.S. that failed to confirm a benefit of LSESr vs. placebo in LUTS, the global body of the peer-reviewed literature attests not only to efficacy but also to safety. Results will be presented of important trials that compare LSESr to alpha-blockers such as tamsulosin (Flomax®) as well as to 5α-reductase inhibitors such as finasteride (Proscar®) that demonstrate consistent findings of near equivalency between LSESr and these pharmacologic agents. Studies relating data indicative of an additive effect or synergy between LSESr and tamsulosin will also be presented. The heightened effectiveness of LSESr in men with severe LUTS vs. moderate LUTS expands the importance of our scrutiny of the global literature concerning LSESr. Of great consequence are the contributions of non-English language peer-reviewed publications that have consistently provided evidence of LSESr efficacy in treating LUTS/BPH. These peer-reviewed articles have shown that the effect of LSESr is not that of a placebo. Finally, a comparison of the LSESr extraction products used in the treatment of LUTS, and a discussion of the milieu factors that affect the natural history of LUTS and influence the outcome of clinical trials, complete this detailed analysis of LSESr vs. LUTS.

1. LSESr and the Placebo Effect: Is There a Resolution?

How Can We Address the Negative Clinical Trials of LSESr vs. Placebo in Male LUTS?

At least 48 systematic reviews and meta-analyses of Serenoa repens have been analyzed as part of due diligence in scrutinizing 190 studies involved in this report. The Willetts study from Australia and the STEP and CAMUS studies from the U.S. are the three major reports presenting negative findings on LSESr efficacy vs. LUTS. The S. repens Treatment for Enlarged Prostates (STEP) [1] and Complementary and Alternative Medicine for Urological Symptoms (CAMUS) trials [2] were randomized, placebo-controlled trials. The findings of STEP and CAMUS contributed to the negative assessment of the efficacy of Serenoa repens, later reported in the Cochrane 2012 meta-analysis [3]. The STEP study randomized a total of 206 men with moderate-to-severe BPH to treatment with placebo (104 patients) or to a supercritical carbon dioxide extract of Serenoa repens (sCESr) (102 patients) using a dose of 160 mg bid [1]. The clinical endpoints of the STEP (Bent) and CAMUS (Barry) trials along with additional information are shown in Table 1.
In STEP, patients were stratified into two groups using the AUA-SI, with moderate LUTS being a score of 8–19 vs. severe LUTS with a score of 20–35. Both the treatment and placebo groups showed a decrease in the AUA-SI of approximately −1.5 during the one-month run-in phase. At 12-months post-randomization there were no significant differences in AUA-SI or urinary flow rate. The AUA-SI decreased −0.68 points from a baseline of 15.7 in patients receiving sCESr vs. a decrease of 0.72 points from a baseline of 15.0 in the placebo group. Similar non-significant changes for sCESr and placebo in the BPHII (BPH Impact Index) were seen from baseline to study end (−0.33) vs. (−0.09), and for Qmax (+0.42 mL/s) vs. (−0.01 mL/s), respectively (Table 1). The sCESr product used in the STEP trial had never been used in a prior study nor assessed in a subsequent study. Perhaps STEP is valid, or perhaps the product used has a lower quality profile than that of other LSESr products that have demonstrated efficacy in the 60 peer-reviewed studies of LSESr vs. LUTS/BPH.
The CAMUS study, published in 2011, was not an investigation of the efficacy of complementary medicine, as per the title, but instead of a particular ethanolic extract of Serenoa repens (EESr) having the brand name Prosta Urgenin® Uno (Rottapharm/Madaus). This was identical to the EESr used 14 years earlier by Derakhshani et al. [4], but surprisingly not discussed by the CAMUS authors [2]. CAMUS randomized 306 men with LUTS to placebo (n = 155) vs. Prosta Urgenin Uno (n = 151) over a study duration of 72 weeks (1.5 years). Eligibility criteria included an AUA-SI of between 8 to 24, and a Qmax ≥ 4 mL/s [2]. The daily dose of the EESr was escalated every 24 weeks, from 320 mg to 640 mg and to 960 mg/day. Results showed a decrease in AUA-SI from 14.69 to 11.70 (20% improvement) for placebo vs. 14.42 to 12.22 (15% improvement) for the EESr product (not statistically significant) [2,5] (Table 1). In the Derakhshani 1997 study, there were 1461 patients from 357 practices in Germany that were assessed for IPSS at the end of three months. Therefore, almost ten times the number of patients were assessed in Derakhshani 1997 vs. CAMUS 2011, but with strikingly different results. In Derakhshani 1997, the mean decrease in IPSS after three months was −7.4 points, representing a 40.4% improvement. The QoL improved by 45.9% (n = 1461) and the Qmax by +3.7 mL/s (30.8%) (n = 1277) (Table 2). The change in IPSS of −7.4 in the Derakhshani study far exceeds the threshold of −3 points cited by many authors as defining a significant therapeutic response [6,7,8,9]. There is no obvious explanation to reconcile these significant differences in outcomes. In the course of this global analysis of LSESr vs. LUTS, we found a total of 58 peer-reviewed articles that met our criteria for evaluability; these included the CAMUS and the Derakhshani studies. The results of the mean changes in IPSS, QoL and Qmax for the 55 positives of the 58 total reports indicate significant improvements in all parameters. This will be discussed in detail in the sections that follow.
The bottom line is that STEP and CAMUS lessened enthusiasm for the use of Serenoa repens in the United States. Scrutiny of all publications related to both trials does not disclose any obvious shortcomings to account for the lack of efficacy of LSESr in the study participants.

2. Therapeutic Comparator Studies of LSESr vs. LUTS

2.1. HESr and EESr Are Not Inferior to Tamsulosin or to Finasteride

In contrast to STEP and CAMUS finding no evidence of efficacy of LSESr vs. LUTS, a significant benefit has been observed in controlled trials of both HESr and EESr vs. therapeutic comparators. Four trials have compared the therapeutic activity of HESr with that of an alpha-blocker such as tamsulosin, or to a 5α-reductase inhibitor such as finasteride (Table 3).
All HESr studies have used Permixon® (Pierre Fabre Medicament S.A.). In fact, all HESr published studies cited in our review have used Permixon, and only the 1987 study of Ollé Carreras did not specifically identify the HESr product used. An investigation in LUTS therapy involving Permixon vs. the alpha-blocker tamsulosin (Flomax®) was the PERMAL 12-month study reported by Debruyne in 2002 [10]. This large study of over 700 men, with an IPSS eligibility criterion of >10, disclosed a decrease in the IPSS of −4.4 points in both the Permixon and tamsulosin groups, with almost identical percentage improvements of 28% and 29%, respectively (Table 3). The changes in the Qmax for Permixon vs. tamsulosin were +1.8 vs. +1.9 mL/s, respectively. After three months of treatment, 34% of the patients on Permixon had an improvement in Qmax of at least 3 mL/s that persisted at 12 months. Similarly, at three months, 35% of the tamsulosin group improved the Qmax to at least 3 mL/s, and at 12 months 37% of the tamsulosin group had this response. The results of Permixon vs. tamsulosin are almost identical.
Another comparative study was done by Latil et al. in 2015. They performed a randomized, double-blind study comparing HESr at 320 mg/day in 83 patients vs. tamsulosin 0.4 mg/day in 86 patients over three months [11]. The average IPSS score with Permixon decreased from 17.7 at baseline to 13.2 (−4.5) on day 90. In comparison, the respective IPSS decrease for tamsulosin was 16.8 to 10.3 (−6.5) (Table 3).
In 2020, Alcaraz et al. [12] reported their follow-up to the QUALIPROST study of 2016 [16] that compared HESr (Permixon) vs. tamsulosin vs. a combination of the two agents. The combination arm results were statistically superior related to IPSS, QoL, BII at p values of 0.002, 0.001, and 0.007, respectively (supplementary document at https://www.mdpi.com/2077-0383/9/9/2909/s1, accessed on 30 July 2021). The IPSS, QoL, and Qmax results are shown in Table 3. The quality of life as assessed from the bother question of the IPSS is not shown but had similar findings of −2.8 (35.9%), −2.6 (32.9%), and −3.4 (41%) for tamsulosin, Permixon, and the combination, respectively.
A comparison of HESr with the 5-α reductase inhibitor finasteride was done in 1996 by Carraro et al. [13]. This trial evaluated 951 men over 26 weeks to ascertain the efficacy of Permixon (n = 467) vs. finasteride (n = 484) using the IPSS as the primary endpoint. The baseline IPSS was 15.7 ± 5.8 and 15.7 ± 5.7, for Permixon and finasteride, respectively. At 26-weeks, the IPSS decreased by −5.8 and −6.1, representing improvements of 37% vs. 39%, respectively. The onset of action for both Permixon and finasteride was as early as six weeks after initiating treatment and was associated with a similar degree of improvement in the IPSS, with both treatment approaches improving by 22% (p < 0.001). However, by 26 weeks there was further improvement, which reached nearly 40% for both treatments. This change in IPSS over time in response to LSESr is important when looking at the results of studies of short duration, (i.e., 4 to 6 weeks), and in hindsight validates our evaluability criterion of at least two or more months study duration to see the full impact of LSESr vs. LUTS on the IPSS. Regarding QoL, 70% of patients reported an improvement at 26-weeks with the QoL measurement (question 8 of the IPSS) dropping from −3.63 to −2.25 with Permixon, and from −3.66 to −2.15 with finasteride. These represent improvements of 38% and 41%, respectively. Qmax at baseline was 10.6 mL/s for Permixon and 10.8 mL/s for finasteride, and at 26-weeks was +2.7 mL/s and +3.2 mL/s, respectively, reflecting 25% improvement for Permixon vs. 30% improvement for finasteride (Table 3). Comparing side effects, patients receiving finasteride experienced a statistically significant deterioration in sexual function vs. those receiving Permixon. This difference was noted from the first follow-up at six weeks and continued to be significant at 26 weeks [13].
Five years after the PERMAL study, Hizli et al. [14] reported the results of a 6 months investigation of EESr vs. tamsulosin. They used Prostagood®, which is licensed to Abdi İbrahim Pharmaceuticals in Turkey and identical to Prostagutt® (Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany) and also known as WS 1473. This study involved three arms: EESr vs. tamsulosin vs. the combination of both agents (combo), with 20 patients in each cohort. In this open-label study, EESr was given as 320 mg/day and tamsulosin 0.4 mg/day, with eligibility criteria of an IPSS ≥ 10, Qmax of <15 mL/s, a gland volume of ≥25 cc, and a PSA of ≤4. At 6-months, the IPSS changes were −6.1, −4.6, and −4.9, for HESr, tamsulosin and the combination, respectively. The percentage improvements were 34%, 28% and 31%, respectively. Qmax changes were +3.2, +3.7 and +4.2 for percentage improvements of 34%, 35% and 42%, respectively. QoL results showed improvements of −2.6, −2.1 and −2.2, with respective percentage improvements of 62%, 60% and 63% (Table 3).
Argirovic et al. published their 6-month study in 2013 involving a total of 265 patients and compared the EESr Prostamol® Uno (Berlin-Chemie) vs. tamsulosin monotherapy vs. the combination of Prostamol plus tamsulosin. This study found virtually identical results to that of Hizli et al., insofar as both the absolute values and percentage changes from baseline to the end of study [14]. Results comparing baseline with the study-end were statistically significant, but this was not the case when comparing the three regimen’s study-end results for IPSS, QoL and Qmax, with p values of 0.1, 0.1, and 0.3, respectively.
These six studies of HESr or EESr vs. a prescription drug comparator show consistent findings relating to the efficacy of LSESr vs. LUTS. Together, they involve a total of 2752 patients and have ample data on the clinical endpoints of IPSS, QoL and Qmax. This represents a comparable degree of improvement in the absolute values and percentage changes between HESr, the alpha-blocker (tamsulosin), and a 5-α reductase inhibitor (finasteride), and a similar degree of efficacy between EESr and the alpha-blocker tamsulosin.
While it is true that these are not placebo-controlled studies, we would have to conclude that LSESr is either an active agent vs. LUTS or that tamsulosin and finasteride are no better than a placebo. After the exhaustive in-depth review of all peer-reviewed studies on LSESr vs. LUTS, the “duck principle” appears applicable (Figure 1).

2.2. LSESr Efficacy Is Greater in Severe vs. Moderate LUTS

The greatest improvement in IPSS using a hexanic extract of Serenoa repens (HESr) was observed in those patients with severe lower urinary tract symptoms (LUTS), defined as an IPSS of 20 to 35. In a follow-up to the 2002 PERMAL study [17], a subset of patients with a baseline IPSS of >19 had a decrease of −7.8 points in the Permixon group (65 patients) vs. a −5.8 decrease in the tamsulosin group (49 patients). The corresponding mean percentage decrease in the total IPSS was 35.2% vs. 25.0%, respectively [17]. Further analysis showed that patients with the higher baseline IPSS (>21) had still greater improvement with Permixon and tamsulosin with average IPSS of −9.3 and −6.0 compared to −6.9 and −5.5 found in patients with a baseline IPSS of 20–21, respectively. Flow symptoms (aka obstructive, voiding) symptoms improved more than storage symptoms (aka irritative, filling) symptoms for groups having an IPSS of 20–21 as well as an IPSS >21 with a similar pattern of response for Permixon and tamsulosin. These findings have been extracted from the text and graphs in the Debruyne 2002 and 2004 papers and are shown in Table 4.
Brown and Emberton endorsed the findings of the Debruyne 2004 paper with the following editorial comments. “There is no reason not to take this study seriously. It was part of a European multi-center large-scale study that was well designed and thought out”. “Overall, it appears that phytotherapy is as valid a pharmacotherapy as α-blockers and 5α-reductase inhibitors in the management of men with BPH/LUTS. Indeed, it may have less adverse effects, be better tolerated, and cheaper”. Emberton noted that in the UK phytotherapy cannot be prescribed and that urologists should be aware and informed about phytotherapy as it will inevitably become part of the standard medical therapy for men with BPH/LUTS. “The previous lack of standardization of herbal remedies that once prevented doctors from recommending these products is now much improved” [17].
The Breza study in 2005 from Slovakia evaluated 596 patients who received Prostamol® Uno, an EESr, at a dose of 320 mg/day [18]. In the total population, the change in IPSS over one year was −5.89 (35.9%), with 84% of patients experiencing more than a 3-point drop. In a subgroup of 150 patients with a mean baseline IPSS of 23.3 (range 20–33), the mean IPSS decreased to a post-treatment value of 15.5 or −7.8 points reflecting a 33.5% improvement. However, as Barry et al. pointed out, the change in absolute values in the IPSS may not be as valid as the percentage change in measurements of the IPSS [19]. This appears to be a logical criticism, and if true the percentage changes seen in the Breza study are possibly not of statistical significance. In the Debruyne 2004 study, percentage changes could not be calculated for the IPSS subgroups 20–21 and >21, while they could for the IPSS subgroups >10 vs. >19. Therefore, for Debruyne 2004, only in these subgroups (>10 vs. >19) can it be concluded that the greatest changes in IPSS percentage and absolute values occur in those patients with the more severe baseline values.
The 2020 follow-up to the QUALIPROST study was mentioned earlier. An analysis of response to Permixon vs. tamsulosin vs. the combination of the two in men presenting with IPSS baseline values of >12 indicated a significant improvement in the combination arm over either monotherapy arms in men with an IPSS of >19. In the per protocol (PP) cohort of these severe LUTS patients, the absolute change in IPPS was −7.8 for HESr, −8.2 for tamsulosin, and −10.5 for the combo. The percentage improvements could not be calculated because data for the baseline values in the severe group (IPSS > 19) were not provided. For all PP patients (an IPSS > 12), the PP results were −5.6, −5.9, and −7.3, with percentage improvements of 29.9%, 31.5%, and 37.2% for HESr, tamsulosin and the combo, respectively. In this study and others, it would be important to compare the changes in IPPS in the cohort of patients with severe LUTS (20–35 IPSS) vs. those with moderate LUTS (8–19 IPSS), instead of comparing the severe cohort with all patients. Using the mean IPSS of all pa-tients diminishes the differential results in contrast to the results found when comparing the IPSS changes between the moderate and severe cohorts.
The study performed by Eickenberg et al. in 1997 performed a comparison between men with a baseline IPSS of ≤18 vs. ≥19. In that study, 6967 patients were treated with an EESr (Sita®) at a dose of 320 mg/day for 6 months. At the study end, the IPSS was −8.0. A subgroup analysis based on the IPSS categories ≤18 vs. ≥19 revealed the same mean percentage improvement of 41% at study end [20]. This issue of degree of efficacy of LSESr based on the severity of the baseline IPSS warrants further scrutiny and, perhaps in a follow-up study, the authors of QUALIPROST will clarify this issue by comparing IPSS changes in the severe with the moderate LUTS cohorts. Notwithstanding, the 2020 QUALIPROST publication [12] and its supplement (URL provided earlier) are valuable contributions and raise the possibility of a synergistic effect of LSESr with the alpha-blocker tamsulosin.

3. Peer-Reviewed Evaluable Studies of LSESr vs. LUTS

3.1. Fifty-five out of Fifty-Eight Evaluable Studies Indicate Efficacy

31 English-language and 27 non-English-language peer-reviewed publications were identified and were evaluable. Considering only peer-reviewed evaluable English-language papers on LSESr vs. the endpoints of IPSS, QoL, and Qmax, data was extracted (SBS) from 31 publications for analysis. Not all studies reported all three endpoints. Of the 31 studies, Bent 2006 (STEP) [1], Barry 2011 (CAMUS) [2], and Willetts 2003 [21] represent the three publications reporting no effect of LSESr vs. LUTS. The remaining 28 studies (90%) show results that for the most part are not consistent with a placebo effect (Table 5).
An English-language trial conducted in Egypt by El-Demiry and published in 2004 as an abstract in the British Journal of Urology International was identified late in our exhaustive search of the Serenoa literature [22]. El-Demiry evaluated 200 patients over 6 months using Permixon 160 mg bid after a 2-week washout period. IPSS, QoL, Qmax, residual volume, prostate volume, and PSA assessments were made after 1, 3, and 6 months of treatment. A total of 190 patients completed the study. Significant improvement was seen in the IPSS (−6.6, 30%) at 1 month, and further improvement at 6 months (−11.4, 51%, p < 0.0001). QoL improved by a mean value of 73% at 6 months, and Qmax increased significantly, by +2.8 mL/s at 1 month, up to +3.7 mL/s at 3 months, and further improvement by +4.4 mL/s (45.4%) at 6 months (p < 0.0001; Table 5) [22].
If non-English-language peer-reviewed papers are considered, 27 evaluable publications can be identified (Table 6). All but two studies indicated the efficacy of LSESr vs. LUTS using an IPSS threshold of ≥−3. One additional study by Tosto did not meet the threshold of clinical significance when using the criteria of an IPSS percentage improvement of less than 35% [23]. The majority of these non-English-language studies were not cited by authors of the major English-language literature on LSESr. The publication dates of these non-English-language publications range from 1983 to 2013. It would seem improbable for the beneficial effects of LSESr to be the result of a placebo effect given the (a) consistency regarding efficacy across so many studies published in different countries over close to 40 years (1983 to 2021), and (b) the average improvement of the IPSS of −6.7, and −5.3 for hexane, and ethanol extraction methods, respectively, given the cited threshold of changes in IPSS of more than −3 or −4 as being able to differentiate placebo from active agents [6,7,8,9].
In addition, head-to-head studies, placebo-controlled and non-placebo-controlled studies of LSESr, showed similar, if not superior, efficacy when compared with the 5α-reductase inhibitor finasteride or with the α-blocker tamsulosin as shown earlier in Table 3. If LSESr is a placebo, then so too are finasteride and tamsulosin. This is what Frater-Schröder concluded in the 2009 editorial entitled “when a = b and a = c, then b = c” [48]. This editorial was directed at the Cochrane 2012 meta-analysis [3] and its criticism of the Carraro 2006 [13] and Debruyne 2002 [10] studies. In Frater-Schröder’s opinion, “Quasi-scientifically-based reports like this Cochrane review weaken the importance and value of phytotherapy in the awareness of experts and the general public” [48]. At the time this editorial was published, Frater-Schröder was the co-secretary and an active member of the scientific committee of ESCOP, an organization that described the results of some Serenoa studies, but whose committee report never came to conclusions about the efficacy of LSESr [49].

3.2. Previous Key Assessments of the Literature (Novara and Vela-Navarrete)

Two key meta-analyses reviewed the efficacy and safety of the HESr (Permixon) in the treatment of LUTS [50,51]. The Novara 2016 meta-analysis identified seven randomized and controlled clinical trials each conducted with Permixon and concluded that Permixon improved peak urinary flow rate (Qmax) and decreased nocturia compared with placebo. However, 6 of 7 studies cited by Novara were considered (SBS) non-evaluable for the following reasons. Three studies had less than 20 patients at study end [52,53,54], two studies had a duration of only four weeks [55,56], and one study presented unclear data [579]. Two additional studies reviewed by Novara concluded that Permixon relieved LUTS comparably to tamsulosin [10,11]. These latter two studies did meet the requirements for evaluability (Debruyne 2002 and Latil 2015, Table 3). Finally, in two other studies reviewed by Novara, Permixon and tamsulosin were used as therapy in combination, but Permixon was not evaluated as monotherapy, so efficacy could not be concluded [58,59]. In contrast to tamsulosin and finasteride, Permixon had little impact on sexual function and the safety profile of HESr was comparable to placebo [50].
Similar to the Novara meta-analysis, Vela-Navarrete reviewed 27 studies using HESr as monotherapy in patients with LUTS at the standard dose of 320 mg/day [51]. This 2018 meta-analysis included 15 randomized and controlled studies and 12 observational studies conducted under conditions of routine clinical practice. The authors concluded that the standardized HESr was well-tolerated and effective for the long-term treatment of LUTS/BPH and that HESr reduced nocturia and improved peak urinary flow rate vs. placebo. Moreover, patients receiving the standardized HESr had a statistically significant mean improvement in the IPSS, decreasing from baseline by −5.73 points (p < 0.0001), and well above the minimum 3-point improvement cited by Barry 1995 as a threshold for clinical significance [6]. The mean IPSS per Vela-Navarrete above is very close in value to the mean IPSS of −5.1 and −6.0 shown in the 31 English-language and the 27 evaluable non-English studies shown in Table 5 and Table 6, respectively. Vela-Navarrete’s review included 27 studies, vs. the 58 studies employing all extraction methods in this review. The Vela-Navarrete 2018 review included the 1997 open-label study by Foroutan et al. [34] conducted in Austria, with 592 patients evaluated over 3 months. This study showed an improvement in IPSS (−6.48; 38%), in QoL (−1.49; 45%), and in Qmax (+5.85 mL/s; 66%). This study was initially missed in this author’s (SBS) search of the Serenoa literature; it is an important evaluable study (see Table 6). Of note with these results, and results presented in Parts I–III, is that Roehrborn et al. found an improvement in symptom score of ≥35% to be “clinically significant” [23].
An additional non-English-language study not easily discoverable with standard search approaches is the open, multicenter study by Medeiros et al., published in 2000 in Portuguese [35]. They evaluated 130 patients from 17 urology centers over 3 months. The IPSS was significantly improved (−6.54, 37.5%, p < 0.0001), as was QoL (−1.37, 38.6%, p < 0.0001) and Qmax (+1.95 mL/s, 22%, p < 0.0001; Table 6). Medeiros et al. used a different scale for QoL assessment, scoring 6 (worst) to 1 (best), rather than the established scoring of 6 (worst) and 0 (best).

4. In the Final Analysis, the Effect of LSESr vs. LUTS Is Not a Placebo Effect

There is no question that the lack of efficacy of LSESr per the STEP and CAMUS double-blind placebo-controlled trials resulted in diminishing physician acceptance of LSESr in the US. This negative impact has been and continues to be compounded by the absence of tight regulations concerning quality requirements and the commercial prevalence of non-standardized Serenoa products in the United States. In other words, the marketplace in the US is flooded with saw palmetto products that range from good to inferior quality. In contrast, LSESr products from hexane, ethanol, or carbon dioxide extraction processes that meet a standardized profile are widely used in Europe, and accordingly physician perception of LSESr vs. LUTS is of a significantly higher degree in Europe, Asia, and South America than in the US.
Adding to the complexity of the above issue concerning the quality of Serenoa repens products is that the carbon dioxide LSESr product used in STEP was never evaluated in another trial, which would have confirmed or refuted the conclusions reached in STEP. Did the results of STEP represent an outlier study, in contrast with the results of many other published studies? With the evaluability criteria detailed in Part I, a total of 58 evaluable peer-reviewed studies of LSESr were found (SBS) using the three different extraction methodologies. Thirteen of these 58 used a lipidosterolic product from carbon dioxide extraction as did STEP. The mean clinical endpoints of IPSS, QoL, and Qmax in these 13 studies were −4.6, −1.2, and +3.5, respectively. It is important to note that these results include both negative studies using carbon dioxide extraction (STEP [1], and Willetts [21]). If these two negative studies are eliminated from analysis, the mean results are −8.4 for IPSS, −1.5 for QoL, and +4.2 mL/s for Qmax, with percentage improvements of 53%, 42%, and 34%, respectively (see Table 7). Once again, per Roehrborn et al.: “A clinically significant improvement in lower urinary tract symptoms was prospectively defined as a 35% or better improvement in the AUA Symptom Index (AUASI) compared to baseline.”
Adding to the complexity of the above issue concerning the quality of Serenoa repens products is that the carbon dioxide LSESr product used in STEP was never evaluated in another trial, which would have confirmed or refuted the conclusions reached in STEP. Did the results of STEP represent an outlier study, in contrast with the results of many other published studies? With the evaluability criteria detailed in Part I, a total of 58 evaluable peer-reviewed studies of LSESr were found (SBS) using the three different extraction methodologies. Thirteen of these 58 used a lipidosterolic product from carbon dioxide extraction as did STEP. The mean clinical endpoints of IPSS, QoL, and Qmax in these 13 studies were −4.6, −1.2, and +3.5, respectively. It is important to note that these results include both negative studies using carbon dioxide extraction (STEP [1], and Willetts [21]). If these two negative studies are eliminated from analysis, the mean results are −8.4 for IPSS, −1.5 for QoL, and +4.2 mL/s for Qmax, with percentage improvements of 53%, 42%, and 34%, respectively (see Table 7). Once again, per Roehrborn et al.: “A clinically significant improvement in lower urinary tract symptoms was prospectively defined as a 35% or better improvement in the AUA Symptom Index (AUASI) compared to baseline.”
The CAMUS 2011 study publication used Prosta Urgenin Uno, an EESr. An earlier paper by Derakhshani in 1997 involving 1047 men evaluated the identical product over a treatment period of three months. Findings showed a mean change in IPSS of −7.4, QoL improvement by 46%, and Qmax increase of 3.7 mL/s [4]. Of the 58 evaluable studies reviewed by this author (SBS), 21 (36%) used an ethanol extract product and all showed positive results except for the CAMUS study. The mean clinical endpoints of IPSS, QoL, and Qmax, including the negative CAMUS study, were −5.1, −1.8, and +2.8, respectively. The percentages of mean improvement were 34%, 45% and 25%, respectively. If the CAMUS data for IPSS are removed, the IPSS mean results further improve from −5.1 to −5.3 (see Table 7). After examining all extraction processes, including the outcome from negative studies, and including all evaluable peer-reviewed articles published in every language, it would appear that the benefit of LSESr vs. LUTS is undeniable.
Cochrane 2012 presented results from studies comparing Serenoa repens vs. placebo [3]. Some of these references were considered non-evaluable (SBS) because the patient number at the end of the study was less than 20, and/or the study duration was less than 2 months. Contrary to the position taken by the authors of Cochrane 2012, those non-evaluable studies (SBS) did indicate efficacy of Serenoa repens vs. LUTS. Data from these studies considered negative by Cochrane 2012 included a Qmax +3.4 mL/s, a decrease in nocturia with 55% improvement, and a decrease in urgency of 65%. In contrast to the Cochrane 2012 assessment, the 2018 Vela-Navarrete meta-analysis confirmed the efficacy of the HESr product Permixon vs. placebo (or comparator) in randomized clinical trials. Of the 15 trials considered by Vela-Navarrete 2018, seven are placebo-controlled [51]. Vela-Navarrete 2018 used a random effect model and considered publication bias. The outcome of their review determined that HESr compared to placebo was associated with 0.64 fewer voids/night (95% confidence interval (CI) −0.98 to 0.31, p < 0.001) and an increase in Qmax of +2.75 mL/s (95% CI 0.57 to 4.93; p = 0.01). Figure 2 in the Vela-Navarrete 2018 paper [51] showed a forest plot for Qmax from four studies (Boccafoschi 1983 [52], Emili 1983 [53], Tasca 1985 [54], and Descotes 1995 [55]) comparing HESr (n = 122) to placebo (n = 133). The studies that supported efficacy for a decrease in nocturia and improvement in Qmax were not impacted by study heterogeneity, and no publication bias could be found in the 2018 review and meta-analysis by Vela-Navarrete et al. [51].
A table of the 17 English and non-English-language placebo-controlled studies for Serenoa repens vs. LUTS/BPH is presented in Table 8. For the HESr studies, Vela-Navarrete 2018 did not cite the Mandressi 1983 study [56], which also included a separate Pygeum intervention arm. Boccafoschi 1983, Emili 1983, Mandressi 1983, Champault 1984 [60], Tasca 1985, and Descotes 1995 were all considered by Cochrane 2012 [61]. Including clinical studies of LSESr using hexane, ethanol, and CO2 extraction processes, a robust set of literature for placebo-controlled trials for Serenoa repens exists and confirms that the body of evidence for the efficacy of Serenoa repens does not represent a placebo effect.
The Boyle 2004 meta-analysis analyzed eight randomized clinical trials and presented findings consistent with the data presented above in (Table 8). Boyle et al. found that HESr vs. LUTS was associated with a 5-point reduction in IPSS and significant improvements in Qmax and nocturia compared to placebo [62]. As mentioned earlier, of these 17 placebo-controlled studies, eight were categorized as non-evaluable due to a short study duration in five (Champault, Descotes, Emili, Mandressi, Löbelenz) [53,55,56,63], too few patient numbers in two (Boccafoschi, Tasca), [52,54] or unclear data in one (Reece Smith) [57]. Excluding these arbitrarily defined non-evaluable studies did not alter the results from our analysis of the evaluable studies in supporting the efficacy of LSESr vs. the key study endpoints such as IPSS, QoL, and Qmax. Moreover, of the seven studies considered non-evaluable due to low patient number (<20 patients) or short duration (<2 months), clinical endpoints such as urgency decreased by 65%, nocturia decreased by 55%, and Qmax improved by 3.0 mL/s (Table 9). These findings further support to the body of literature attesting to LSESr efficacy vs. LUTS.

5. Extract Quality May Affect LSESr Efficacy

Major differences between extraction technology and composition of finished Serenoa products have been identified, and which have been stated to significantly impact the ability of the supplement to ameliorate LUTS [65,66,67,68]. Research suggests that there is a “fingerprint” of saw palmetto that represents a quality standardized profile. A key element of this quality standardized profile is the ratio and content of fatty acids, which can vary dramatically across products [67]. Both the EU monograph and USP standards established the minimum level of total fatty acids (TFA) that are needed for a quality Serenoa repens extract. The USP also established ratios of the key fatty acids compared with lauric acid that are required to meet the established chemical profile [69]. The USP stated the chemical profile for a quality Serenoa repens extract would have a minimum of 80% total fatty acids and have a fatty acid composition of oleic acid (30–35%), lauric acid (26–32%), myristic acid (10–12%), palmitic acid (8.5–9.2%), and linoleic acid (4.3–6.0%) [69]. This fatty acid profile distinguishes quality saw palmetto extracts from vegetable oils, adulterated products, and dried saw palmetto berry powders that are deficient in fatty acid amount and/or composition. Key issues remain whether or not the content of total vs. free fatty acids (FFA) of LSESr correlates with efficacy in treating LUTS or does a particular fatty acid account for LSESr activity, and also, whether or not one extraction process is better than another.

6. The Extraction Process Does Not Correlate with the Efficacy of LSESr Products vs. LUTS

An analysis of 20 commercially available Serenoa repens products using a gas chromatography-flame identification detector (GC-FID) and gas chromatography-mass spectrometry (GC-MS) showed considerable variability in TFA and phytosterol content among preparations [68]. In another gas chromatography study involving 19 different Serenoa repens mono-preparations, the fatty acid content varied from one-tenth to greater than 4.6-times the fatty acid mg/day dose stated on the supplement product package insert [65]. The mean FFA content in 14 different Serenoa repens products available in Europe has ranged from as high as 80.7% to as low as 40.7% [67]. Additionally, only 9 of the 19 mono-preparations evaluated contained the recommended daily dosage of 320 mg LSESr per day, consisting of 70% to 95% fatty acids (range, 224–304 mg) [65]. With consideration of this huge variability in the quality of Serenoa repens products, the hexanic lipidosterolic extract Permixon has been found to have the highest percentage of FFA, and this finding has been attributed to the therapeutic efficacy of this LSESr.
Studies suggest that hexane, supercritical CO2, and ethanol extraction technologies lead to different fatty acid and phytonutrient profiles. However, commercial Serenoa repens extract-containing products made from any of the extraction technologies are said to have demonstrated activity against 5α-reductase and/or to have an impact on symptoms of LUTS/BPH [50,70]. At a biological level, the pharmacologic activity of 10 lipidosterolic extracts of Serenoa repens differed in the degree to which they inhibited fibroblast proliferation as well as 5α-reductase Types 1 and 2 [71]. It was the hexanic lipidosterolic extract that most actively inhibited enzyme activity and fibroblast-induced cell proliferation. Data on supercritical CO2 extracts are more limited, but it is reported that LSESr extracts using ultrahigh-pressure supercritical CO2 have a fatty acid profile similar to hexanic lipidosterolic extracts (Valensa International. 2021. Comparable fatty acid profile of LSESr products. Data on file). Because ethanol has a different polarity than hexane, this may contribute to the differences in the extract profiles of hexane vs. ethanol lipidosterolic products. Do such differences in fatty acid profiles or biochemical actions translate to marked differences in the clinical efficacy of LSESr vs. LUTS? Our review of the peer-reviewed Serenoa repens literature, with evaluability requirements for LSESr monotherapies, and known extraction modality yielded findings that failed to show any obvious relationships between extract type and the degree of clinical effect. The pooled results of the IPSS, QoL, and Qmax for 24, 21, and 13 evaluable studies using hexane, ethanol, and CO2 extraction, respectively, show very similar results. These findings were presented previously (Table 7). Neither FFA content nor extraction modality have any bearing on clinical efficacy.
Unfortunately, none of the clinical studies of LSESr products have involved head-to-head comparisons of one extraction process vs. another [72]. Although it would seem apparent that every peer-reviewed study should detail the extraction process and the details of dosing, it is disappointing that some publications omitted such crucial information. Concerning the clinical endpoints of IPSS, QoL and Qmax, when the extraction method is one of the standard LSESr processes, be it hexane, ethanol, or carbon dioxide, there is no evidence of the superiority of one process over another. The biological differences seen in non-clinical studies dealing with various pharmacologic actions in vitro seem to have minimal relevance to what is seen in vivo in human clinical studies. The major challenge is to educate both physicians and the lay public that products labeled as “saw palmetto” or “Serenoa repens” are not equivalent to a standardized lipidosterolic extract of Serenoa repens (LSESr) with a product profile that meets an established definition, and that only the use of the latter is acceptable.

7. Milieu Factors Are Important When Assessing LSESr vs. LUTS

The demographics of the population under study cannot be ignored when evaluating clinical trial results. Strong epidemiologic data demonstrate that lifestyle factors such as obesity, diet, alcohol intake, stress, and physical activity, play a role in LUTS etiology and progression [73,74,75,76,77]. In fact, such lifestyle issues interact with each other and are consequential in the processes of inflammation, aging, and cancer. The patient’s medical history, the presence of co-existent chronic inflammation, the proper assessment of diabetes, metabolic syndrome, and details about medications and supplements and possible drug interactions need to be taken into account when determining the clinical efficacy of LSESr vs. LUTS [78,79,80,81,82,83,84,85]. Study design should stratify patients into subsets of those who may have lingering pharmacologic effects of alpha-blockers vs. those who were never on them. Inflammation is of such paramount importance that a more substantive assessment of the patient’s inflammatory status must be routine in any analysis of LUTS. Despite significant technological advances in the biological sciences, the current testing of inflammation remains inadequate and should be addressed in future trials of LSESr given the widely recognized role of inflammation in the development of LUTS [86,87,88,89,90,91,92,93]. The anti-inflammatory properties of Serenoa repens are detailed in many reports [11,93,94,95,96,97,98]. Lifestyle modifications aimed at reducing inflammation should help modulate LUTS symptoms and possibly prevent progression [73]. Investigators should consider stratifying the patient population using an “inflammation index”, and further interpreting clinical data by comparing an “inflammation index” with a quality of life indicator such as the bother question of the AUASI [99,100] or the BPHII [16].
Simple measures such as restricting fluid intake 4 h before bedtime, routinely attempting to void prior to sleep, limiting or omitting caffeinated beverages, and avoiding salt in the diet can significantly affect a key symptom such as nocturia [101,102,103,104]. Principal investigators should evaluate study participants based on these lifestyle factors to clarify the potential beneficial effects of LSESr relative to the possible lifestyle modifications known to affect LUTS. In an article published 25 years ago [55], Descotes et al. referred to an article written by Castro still 23 years earlier. In that publication from nearly a half-century ago, Castro remarked on the challenges faced when evaluating patients undergoing treatment for LUTS. “The clinical symptoms of BPH are also labile, and can vary with time, seasons, stress, medication, changes in sympathetic activity, bladder training, sedentary activity, and irregular voiding. Spontaneous variation in disease symptoms and the degree of dynamic obstruction, coupled with a pronounced placebo effect, clearly complicate any assessment of drug efficacy in BPH” [55,105]. Forty-two years after the 1972 publication by Castro, Vaughan shared his views about the clinical lability of LUTS. “To this, I would add my 47 years in frankly discussing LUTS with thousands of patients. Not only is there variability in nocturia, but also in symptoms of hesitancy, weak stream, incomplete emptying, urgency, and terminal dribbling” [106]. At age 78 years (SBS), and despite being a non-smoker, non-drinker, following a low sodium diet, and not having sleep apnea or obesity as problems, I too echo these observations about the day-to-day variations in LUTS. Such variable symptomatology, combined with issues such as the quality of the Serenoa repens product, and patient compliance with medications, contribute to the difficulty in our understanding of LUTS and its optimal approach to prevention and treatment.

8. Clinical Perspective

A review and network meta-analysis (NMA) of randomized placebo-controlled trials on Serenoa repens vs. placebo vs. alpha-blockers in the treatment of LUTS was reported by Russo et al. [107]. Twenty-two trials were identified by the authors for data investigation using this NMA methodology. The outcomes of IPSS and peak flow were considered across the 22 studies, including 10 randomized trials comparing LSESr to a placebo (five studies), or an alpha-blocker (five studies). For the LSESr studies, two used a HESr product that was compared to tamsulosin, and eight other studies used a non-hexane Serenoa product, with five being placebo-controlled and three studies using a prescription drug as a comparator. From the NMA, Russo concluded that HESr and non-HESr did not demonstrate clinically meaningful improvement in LUTS and peak flow over placebo. De Nunzio et al. [108] published a response to Russo, criticizing the NMA methodology for being inappropriate to ascertain clinical efficacy, and that patient cohorts were unbalanced and affected the validity of the conclusions reached. De Nunzio et al. also noted that a large number of randomized clinical trials were either not identified or were excluded. They concluded that these shortcomings could lead to false conclusions [108]. Many criticisms similar to those voiced by De Nunzio et al. of the Russo review are to be found in our global review presented herein. This includes failure to retrieve all eligible publications for analysis, including early science that represents the body of literature relevant to modern medicine, and failure to establish strict evaluability criteria for the studies to be reviewed as opposed to relying on methodology for analysis at the expense of clinical relevance. In fact, of the 22 studies cited by Russo, four did not indicate the extraction process, and three of those four were combinations of Serenoa repens with other products, and one study involved only 13 patients. If clinical data are used to present an opinion only, rather than to enhance medical practice, then the data do not have value. The same could be said for the methodology for both reviews and meta-analyses, and the failure to consider non-native language publications. Dated science that is well done builds a foundation for clinical practice and allows patient care to be improved.
The clinical literature on Serenoa repens for the treatment of LUTS is extensive. In some studies, important variables are often inadequately controlled. This has resulted in inconsistent findings and controversy concerning what benefit may result from the use of commercially available Serenoa products. The most important variable repeatedly presented in this multi-part report is whether the product is a lipidosterolic extract of Serenoa repens (LSESr) vs. a crude product such as crushed dried saw palmetto berry powder. In addition to the use of a high-quality standardized LSESr, other factors to consider when evaluating the Serenoa clinical literature are the dosage of LSESr, the criteria used to select patients, the exclusion of products combining Serenoa with other agents, and the clinical study design. Based on the evidence presented, a standardized LSESr, given as monotherapy, and that has an established profile defined by the EMA or USP, at a dosage of 320 mg/day, either in divided doses or as a single daily dose, may contribute to the alleviation of LUTS. The clinically significant endpoints include a decrease in the IPSS score, an improvement in the QoL score, and an increase in peak urinary flow (Qmax), with all parameters achieved in association with a high therapeutic index. LSESr’s very favorable safety profile includes a negligible impact on sexual function [50,51,109,110,111,112,113,114,115]. Despite achieving the desired endpoints mentioned, the American Urological Association (AUA) and the European Association of Urology (EAU) treatment guidelines have downplayed the efficacy of LSESr therapy [118,117]. In contrast, meta-analyses published in 2016 and 2018 support the use of LSESr in men with mild-to-moderate LUTS/BPH [50,51].
In this clinician’s perspective, based upon nearly 40 years of clinical data, some conclusions are clear. First, LSESr has a definite role in the treatment of LUTS. It has a high safety profile with relatively few adverse side effects. It does not cause sexual dysfunction such as ejaculatory disorders seen with 5α-reductase inhibitors, nor hypotension with some α-blockers such as tamsulosin (Flomax®). LSESr does not alter PSA expression and therefore, does not interfere with the monitoring of men at risk of developing prostate cancer. LSESr using hexane, ethanol, or carbon dioxide extraction have all shown efficacy in published studies. The onset of action may be as early as two weeks but is clearly established by 3 months. Of importance is the durability of efficacy seen with long-term treatment of LUTS with LSESr. Of greater significance is the finding of slowing and even halting the progression of LUTS/BPH during prolonged studies using LSESr, with some trials extending 10 to 15 years. Such studies may indicate that LSESr is affecting the pathological processes i.e., pathobiology or etiopathogenesis, that lead to LUTS/BPH. Patient selection is important, and those patients with severe LUTS, and at high risk for acute urinary retention are not optimal candidates and warrant careful observation relating to the need for surgical intervention. The most critical issue in the use of Serenoa repens in treating LUTS is the need to educate physicians that crude herbal products are never to be equated with standardized LSESr products that have a profile established by the EMA or USP and that the former products have no role to play in LUTS treatment. An unresolved issue in the use of LSESr relates to the lack of head-to-head studies to ascertain any difference in the hexane vs. ethanol vs. carbon dioxide extracts, but the results presented in this report would indicate that no particular extraction process is superior to another. An additional issue relates to regulatory agencies and their role in monitoring the quality of products such as LSESr. Why is LSESr available by prescription in some countries, OTC in others, and is inadequately regulated concerning product quality in others, the latter especially in the United States? We should never confuse the business of medicine with the practice of principled medicine. The former has led to the deterioration of medicine as a profession and has diminished the quality of care to patients while increasing the risk of adverse events.
In summary, LSESr (lipidosterolic extract formulations of Serenoa repens) show efficacy in treating LUTS ± BPH, and the results discussed herein provide a rationale for conducting larger, better-controlled studies using such formulations in men with mild-to-moderate LUTS. These studies should quantitate change in IPSS, QoL, Qmax. An evaluation of the inflammatory status of the patient and the effect of long-term use of LSESr on halting the progression of LUTS/BPH should provide further confirmation that LSESr alters the natural history of this affliction of great “bother” in the adult male.

9. Addendum

During the review process of Part III, the response to a discussion of the ethanolic lipidosterolic product Prostagood led to the finding of an additional peer-reviewed paper by Akbulut et al. that had been overlooked during the initial search [118]. In their study of 106 patients, ages 45 years or older and with a baseline IPSS greater than 10, the use of Prostagood for three months resulted in an IPSS decrease of −6.4 (35% improvement). This result is consistent with the findings of our global review.

Funding

The author received a consulting fee for researching and composing this review from U.S. Nutraceuticals, dba Valensa International and funding by an education grant from the Life Extension® group.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

This is a review of the literature and did not require ethical board approval.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author serves as a paid consultant for Valensa International. He receives a consulting fee from Life Extension.

References

  1. Bent, S.; Kane, C.; Shinohara, K.; Neuhaus, J.; Hudes, E.S.; Goldberg, H.; Avins, A.L. Saw palmetto for benign prostatic hyperplasia. N. Engl. J. Med. 2006, 354, 557–566. [Google Scholar] [CrossRef] [Green Version]
  2. Barry, M.J.; Meleth, S.; Lee, J.Y.; Kreder, K.J.; Avins, A.L.; Nickel, J.C.; Roehrborn, C.G.; Crawford, E.D.; Foster, H.E., Jr.; Kaplan, S.A.; et al. Effect of increasing doses of saw palmetto extract on lower urinary tract symptoms: A randomized trial. JAMA 2011, 306, 1344–1351. [Google Scholar] [CrossRef] [Green Version]
  3. Tacklind, J.; Macdonald, R.; Rutks, I.; Stanke, J.U.; Wilt, T.J. Serenoa repens for benign prostatic hyperplasia. Cochrane Database Syst. Rev. 2012, 12, CD001423. [Google Scholar] [CrossRef]
  4. Derakhshani, P.; Geerke, H.; Böhnert, K.J.; Engelmann, U. Influencing the international prostate symptom score during therapy with saw palmetto fruit extract with a single daily dose. Der. Urol. B 1997, 37, 384–391. (In German) [Google Scholar] [CrossRef]
  5. Andriole, G.L.; McCullum-Hill, C.; Sandhu, G.S.; Crawford, E.D.; Barry, M.J.; Cantor, A.; Group, C.S. The effect of increasing doses of saw palmetto fruit extract on serum prostate specific antigen: Analysis of the CAMUS randomized trial. J. Urol. 2013, 189, 486–492. [Google Scholar] [CrossRef]
  6. Barry, M.J.; Williford, W.O.; Chang, Y.; Machi, M.; Jones, K.M.; Walker-Corkery, E.; Lepor, H. 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? J. Urol. 1995, 154, 1770–1774. [Google Scholar] [CrossRef]
  7. Fawzy, A.; Braun, K.; Lewis, G.P.; Gaffney, M.; Ice, K.; Dias, N. Doxazosin in the treatment of benign prostatic hyperplasia in normotensive patients: A multicenter study. J. Urol. 1995, 154, 105–109. [Google Scholar] [CrossRef]
  8. Lepor, H.; Williford, W.O.; Barry, M.J.; Brawer, M.K.; Dixon, C.M.; Gormley, G.; Haakenson, C.; Machi, M.; Narayan, P.; Padley, R.J. The efficacy of terazosin, finasteride, or both in benign prostatic hyperplasia. Veterans Affairs Cooperative Studies Benign Prostatic Hyperplasia Study Group. N. Engl. J. Med. 1996, 335, 533–539. [Google Scholar] [CrossRef]
  9. Roehrborn, C.G. Current medical therapies for men with lower urinary tract symptoms and benign prostatic hyperplasia: Achievements and limitations. Rev. Urol. 2008, 10, 14–25. [Google Scholar]
  10. Debruyne, F.; Koch, G.; Boyle, P.; Da Silva, F.C.; Gillenwater, J.G.; Hamdy, F.C.; Perrin, P.; Teillac, P.; Vela-Navarrete, R.; Raynaud, J.P. Comparison of a phytotherapeutic agent (Permixon) with an alpha-blocker (Tamsulosin) in the treatment of benign prostatic hyperplasia: A 1-year randomized international study. Eur. Urol. 2002, 41, 497–506, discussion 506–507. [Google Scholar] [CrossRef]
  11. Latil, A.; Petrissans, M.T.; Rouquet, J.; Robert, G.; de la Taille, A. Effects of hexanic extract of Serenoa repens (Permixon(R) 160 mg) on inflammation biomarkers in the treatment of lower urinary tract symptoms related to benign prostatic hyperplasia. Prostate 2015, 75, 1857–1867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Alcaraz, A.; Rodriguez-Antolin, A.; Carballido-Rodriguez, J.; Castro-Diaz, D.; Esteban-Fuertes, M.; Cozar-Olmo, J.M.; Ficarra, V.; Medina-Lopez, R.; Fernandez-Gomez, J.M.; Angulo, J.C.; et al. Clinical Benefit of Tamsulosin and the Hexanic Extract of Serenoa Repens, in Combination or as Monotherapy, in Patients with Moderate/Severe LUTS-BPH: A Subset Analysis of the QUALIPROST Study. J. Clin. Med. 2020, 9, 2909. [Google Scholar] [CrossRef]
  13. Carraro, J.C.; Raynaud, J.P.; Koch, G.; Chisholm, G.D.; Di Silverio, F.; Teillac, P.; Da Silva, F.C.; Cauquil, J.; Chopin, D.K.; Hamdy, F.C.; et al. Comparison of phytotherapy (Permixon) with finasteride in the treatment of benign prostate hyperplasia: A randomized international study of 1,098 patients. Prostate 1996, 29, 231–240, discussion 241–232. [Google Scholar] [CrossRef]
  14. Hizli, F.; Uygur, M.C. A prospective study of the efficacy of Serenoa repens, tamsulosin, and Serenoa repens plus tamsulosin treatment for patients with benign prostate hyperplasia. Int. Urol. Nephrol. 2007, 39, 879–886. [Google Scholar] [CrossRef]
  15. Argirović, A.; Argirović, Đ. Does the addition of Serenoa repens to tamsulosin improve its therapeutical efficacy in benign prostatic hyperplasia? Vojnosanit. Pregl. 2013, 70, 1091–1096. [Google Scholar] [CrossRef]
  16. Alcaraz, A.; Carballido-Rodriguez, J.; Unda-Urzaiz, M.; Medina-Lopez, R.; Ruiz-Cerda, J.L.; Rodriguez-Rubio, F.; Garcia-Rojo, D.; Brenes-Bermudez, F.J.; Cozar-Olmo, J.M.; Baena-Gonzalez, V.; et al. Quality of life in patients with lower urinary tract symptoms associated with BPH: Change over time in real-life practice according to treatment–the QUALIPROST study. Int. Urol. Nephrol. 2016, 48, 645–656. [Google Scholar] [CrossRef]
  17. Debruyne, F.; Boyle, P.; Calais Da Silva, F.; Gillenwater, J.G.; Hamdy, F.C.; Perrin, P.; Teillac, P.; Vela-Navarrete, R.; Raynaud, J.P.; Schulman, C.C. Evaluation of the clinical benefit of permixon and tamsulosin in severe BPH patients-PERMAL study subset analysis. Eur. Urol. 2004, 45, 773–779, discussion 779–780. [Google Scholar] [CrossRef] [PubMed]
  18. Breza, J.; Kliment, J.; Valansky, L.; Capova, G. Prostamol uno (alcohol extract of the fruits of Serenoa repens) in the treatment of symptomatic benign prostatic hyperplasia. Lek. Obz. 2005, 54, 139–144. (In Slovakian) [Google Scholar]
  19. Barry, M.J.; Cantor, A.; Roehrborn, C.G.; Group, C.S. Relationships among participant international prostate symptom score, benign prostatic hyperplasia impact index changes and global ratings of change in a trial of phytotherapy in men with lower urinary tract symptoms. J. Urol. 2013, 189, 987–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Eickenberg, H.U. Treatment of benign prostatic hyperplasia with a lipophilic extract from saw palmetto fruits (Sita). Der. Urol. B 1997, 37, 130–133. (In German) [Google Scholar] [CrossRef]
  21. Willetts, K.E.; Clements, M.S.; Champion, S.; Ehsman, S.; Eden, J.A. Serenoa repens extract for benign prostate hyperplasia: A randomized controlled trial. BJU Int. 2003, 92, 267–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. El-Demiry, M. Serenoa repens in the treatment of patients with symptomatic benign prostatic hyperplasia. BJU Int. Suppl. 2004, 94, 146–147. [Google Scholar]
  23. Roehrborn, C.G.; Oesterling, J.E.; Auerbach, S.; Kaplan, S.A.; Lloyd, L.K.; Milam, D.E.; Padley, R.J. The Hytrin Community Assessment Trial study: A one-year study of terazosin versus placebo in the treatment of men with symptomatic benign prostatic hyperplasia. HYCAT Investigator Group. Urology 1996, 47, 159–168. [Google Scholar] [CrossRef]
  24. Cirillo-Marucco, E.; Pagliarulo, A.; Tritto, G.; Piccinno, A.; Di Rienzo, U. Serenoa repens extract (Permixon®) in the early treatment of prostatic hypertrophy. Urol. J. 1983, 50, 1269–1277. (In Italian) [Google Scholar] [CrossRef]
  25. Cukier; Ducassou; Guillou, L. Permixon versus placebo. CR Pharm. Clin. 1985, 4, 15–21. (In French) [Google Scholar]
  26. Tosto, A.; Rovereto, B.; Paoletti, M.C.; Rizzo, M.; Nicolucci, A.; Costantini, A. Serenoa Repens Extract in the Treatment of Functional Disorders Secondary to Adenoma of the Prostate: Considerations on 20 Cases. Urol. J. 1985, 52, 536–542. (In Italian) [Google Scholar] [CrossRef]
  27. Pannunzio, E.; D’Ascenzo, R.; Giardinetti, F.; Civili, P.; Persichelli, E. Serenoa Repens vs. Gestonorone Caproato in the Treatment of Benign Prostatic Hypertrophy: Randomized Study. Urol. J. 1986, 53, 696–705. (In Italian) [Google Scholar] [CrossRef]
  28. Pescatore, D.; Calvi, P.; Michelotti, P. Urodynamic assessment of treatment in patients with prostatic adenoma with Serenoa repens extract. Urol. J. 1986, 53, 894–897. (In Italian) [Google Scholar] [CrossRef]
  29. Authie, D.; Cauquil, J. Assessment of the effectiveness of Permixon* in daily practice multicentric study. CR Pharm. Clin. 1987, 5, 3–13. (In French) [Google Scholar]
  30. Ollé Carreras, J. Our experience with hexane extract from Serenoa repens in the treatment of benign prostatic hypertrophy. Arch. Esp. Urol. 1987, 40, 310–313. (In Spanish) [Google Scholar] [PubMed]
  31. Orfei, S.; Grumelli, B.; Galetti, G. Clinical and uroflowimetric evaluation of Permixon® in geriatrics. Urol. J. 1988, 55, 373–381. (In Italian) [Google Scholar] [CrossRef]
  32. Dathe, G.; Schmid, H. Phytotherapy for benign prostatic hyperplasia (BPH) with Extractum Serenoa repens (Permixon). Urologe. Ausg. B 1991, 31, 223–330. (In German) [Google Scholar]
  33. Aliaev, G.; Vinarov, A.Z.; Lokshin, K.L.; Spivak, L.G. Five-year experience in treating patients with prostatic hyperplasia patients with permixone (Serenoa repens “Pierre Fabre Medicament). Urologiia 2002, 23–25. (In Russian) [Google Scholar]
  34. Foroutan, F. Effectiveness and tolerability of Permixon in a larger patient population (592 patients) under practical conditions. J. Urol. Urogynäkol. 1997, 2, 17–21. (In German) [Google Scholar]
  35. Medeiros, A.S.; Verona, C.B.M.; Mattos, D., Jr.; Silva, E.G.; Fonseca, G.N.; Begliomini, H.; Pous, J.H.; Cury, J.; Costa, M.M.; Prado, M.J.; et al. Efficacy and tolerability of the extract of Serenoa repens in a multicentric study in patients with symptomatic benign prostatic hyperplasia. Rev. Bras. Med. 2000, 57, 321–324. (In Portuguese) [Google Scholar]
  36. Redecker, K.D.; Funk, P. Sabal-Extrakt WS 1473 bei benigner Prostatahyperplasie. Extr. Urol. 1998, 21, 23–25. [Google Scholar]
  37. Ziegler, H.; Holscher, U. Efficacy of saw palmetto fruit special extract WS 1473 in patients with Alken stage I-II benign prostatic hyperplasia-open multicentre study. Jatros Urol. 1998, 14, 34–43. (In German) [Google Scholar]
  38. Aliaev, Y.G.; Apolikhin, O.I.; Mazo, E.B.; Vinarov, A.Z.; Lokshin, K.L.; Medvedev, A.A.; Permyakova, O.V.; Spivak, L.G.; Shkol’nikov, M.E. First results of a clinical trial of the efficacy and safety of Prostamol® Uno in patients with the early signs of prostatic hyperplasia. Eff. Pharm. Urol. 2007, 8, 11. (In Russian) [Google Scholar]
  39. Razumov, S.V.; Egorov, A.A. Expediency of switching from combined therapy with prostamol Uno and alpha-1-adrenoblockers to monotherapy with prostamol Uno in patients with prostatic adenoma. Urologiia 2007, 3, 47–50. (In Russian) [Google Scholar]
  40. Aliaev, G.; Apolikhin, O.I.; Mazo, E.B.; Vinarov, A.Z.; Lokshin, K.L.; Medvedev, A.A.; Permiakova, O.V.; Spivak, L.G.; Shkol’nikov, M.E. Efficacy and safety of Prostamol-UNO in the treatment of patients with initial symptoms of prostatic adenoma and risk of progression: 2 years of investigations. Urologia 2009, 4, 36–40. (In Russian) [Google Scholar]
  41. Vinarov, A.Z.; Aliaev Yu, G.; Apolikhin, O.I.; Mazo, E.B.; Darenkov, S.P.; Demidko Iu, L.; Lokshin, K.L.; Medvedev, A.A.; Permiakova, O.V.; Spivak, L.G.; et al. Results of three-year clinical study of Prostamol Uno efficacy and safety in patients with initial symptoms of prostatic adenoma and risk of its progression. Urologiia 2010, 6, 3–10. (In Russian) [Google Scholar]
  42. Aliaev, G.; Vinarov, A.Z.; Demidko Iu, L.; Spivak, L.G. The results of the 10-year study of efficacy and safety of Serenoa repens extract in patients at risk of progression of benign prostatic hyperplasia. Urologiia 2013, 4, 32–36. (In Russian) [Google Scholar]
  43. Mattei, F.M.; Capone, M.; Acconcia, A. Medicamentous therapy of benign prostatic hyperplasia with an extract of the sagebrush. TW Urol. Nephrol. 1990, 2, 346–350. (In German) [Google Scholar]
  44. Vahlensieck, W., Jr.; Volp, A.; Lubos, W.; Kuntze, M. Benign prostatic hyperplasia--treatment with sabal fruit extract. A treatment study of 1334 patients. Fortschritte der Medizin 1993, 111, 323–326. (In German) [Google Scholar]
  45. Vahlensieck, W.; Völp, A.; Kuntze, M.; Lubos, W. Changes in micturition in patients with benign prostatic hyperplasia under sabal fruit treatment. Urologe. Ausg. B 1993, 33, 380–383. (In German) [Google Scholar]
  46. Fabricius, P.G.; Vahlensieck, W., Jr. Therapy for benign prostatic hyperplasia: Sabal fruit extract: One dose is enough! Therapiewoche 1993, 43, 1616–1620. (In German) [Google Scholar]
  47. Bauer, H.W.; Casarosa, C.; Cosci, M.; Fratta, M.; Blessmann, G. Saw palmetto fruit extract for treatment of benign prostatic hyperplasia. Results of a placebo-controlled double-blind study. MMW Med. 1999, 141, 62. (In German) [Google Scholar]
  48. Frater-Schröder, M. When a=b and a=c, then b=c. ARS MEDICI Thema Phytother. 2009, 9, 2. (In German) [Google Scholar]
  49. ESCOP Monographs. Serenoae Repentis Fructus (Sabal Fructus) Saw Palmetto Fruit, 2nd ed.; George Thieme Verlag: New York, NY, USA, 2003. [Google Scholar]
  50. Novara, G.; Giannarini, G.; Alcaraz, A.; Cozar-Olmo, J.M.; Descazeaud, A.; Montorsi, F.; Ficarra, V. Efficacy and Safety of Hexanic Lipidosterolic Extract of Serenoa repens (Permixon) in the Treatment of Lower Urinary Tract Symptoms Due to Benign Prostatic Hyperplasia: Systematic Review and Meta-analysis of Randomized Controlled Trials. Eur. Urol. Focus 2016, 2, 553–561. [Google Scholar] [CrossRef]
  51. Vela-Navarrete, R.; Alcaraz, A.; Rodriguez-Antolin, A.; Minana Lopez, B.; Fernandez-Gomez, J.M.; Angulo, J.C.; Castro Diaz, D.; Romero-Otero, J.; Brenes, F.J.; Carballido, J.; et al. Efficacy and safety of a hexanic extract of Serenoa repens (Permixon((R))) for the treatment of lower urinary tract symptoms associated with benign prostatic hyperplasia (LUTS/BPH): Systematic review and meta-analysis of randomised controlled trials and observational studies. BJU Int. 2018, 122, 1049–1065. [Google Scholar] [PubMed] [Green Version]
  52. Boccafoschi, C.; Annoscia, S. Comparison of Serenoa repens extract and placebo in controlled clinical trial in patients with prostatic adenomatosis. Urologiia 1983, 50, 1–14. (In Italian) [Google Scholar]
  53. Emili, E.; Lo Cigno, M.; Petrone, U. Clinical results on a new drug in prostate hypertrophy therapy (Permixon). Nefrol. Chir. 1983, 50, 1042–1048. (In Italian) [Google Scholar]
  54. Tasca, A.; Barulli, M.; Cavazzana, A.; Zattoni, F.; Artibani, W.; Pagano, F. Treatment of obstructive symptomatology caused by prostatic adenoma with an extract of Serenoa repens. Double-blind clinical test v. placebo. Minerva Urol. Nefrol. 1985, 37, 87–91. (In Italian) [Google Scholar]
  55. Descotes, J.L.; Rambeaud, J.J.; Deschaseaux, P.; Faure, G. Placebo-Controlled Evaluation of the Efficacy and.Tolerability of Permixon® in Benign Prostatic Hyperplasia after Exclusion of Placebo Responders. Clin. Drug Invest. 1995, 9, 291–297. [Google Scholar] [CrossRef]
  56. Mandressi, A.; Tarallo, U.; Maggioni, A.; Tombolini, P.; Rocco, F.; Quadraccia, S. Medical treatment of benign prostatic hyperplasia: Efficacy of the extract of Serenoa repens (Permixon) compared to that of the extract of Pygeum africanum and a placebo. Urologia 1983, 50, 752–758. (In Italian) [Google Scholar] [CrossRef]
  57. Reece Smith, H.; Memon, A.; Smart, C.J.; Dewbury, K. The value of permixon in benign prostatic hypertrophy. Br. J. Urol. 1986, 58, 36–40. [Google Scholar] [CrossRef]
  58. Glemain, P.; Coulange, C.; Billebaud, T.; Gattegno, B.; Muszynski, R.; Loeb, G. Tamsulosin with or without Serenoa repens in benign prostatic hyperplasia: The OCOS trial. Prog. Urol. 2002, 12, 395–403. discussion 404(In French) [Google Scholar]
  59. Ryu, Y.W.; Lim, S.W.; Kim, J.H.; Ahn, S.H.; Choi, J.D. Comparison of tamsulosin plus serenoa repens with tamsulosin in the treatment of benign prostatic hyperplasia in Korean men: 1-year randomized open label study. Urol. Int. 2015, 94, 187–193. [Google Scholar] [CrossRef]
  60. Champault, G.; Bonnard, A.M.; Cauquil, J.; Patel, J.C. Medical treatment of prostatic adenoma. Controlled trial: PA 109 vs placebo in one hundred and ten patients. Ann. Urol. (Paris) 1984, 18, 407–410. (In French) [Google Scholar] [PubMed]
  61. MacDonald, R.; Tacklind, J.W.; Rutks, I.; Wilt, T.J. Serenoa repens monotherapy for benign prostatic hyperplasia (BPH): An updated Cochrane systematic review. BJU Int. 2012, 109, 1756–1761. [Google Scholar] [CrossRef] [PubMed]
  62. Boyle, P.; Robertson, C.; Lowe, F.; Roehrborn, C. Updated meta-analysis of clinical trials of Serenoa repens extract in the treatment of symptomatic benign prostatic hyperplasia. BJU Int. 2004, 93, 751–756. [Google Scholar] [CrossRef] [PubMed]
  63. Champault, G.; Patel, J.C.; Bonnard, A.M. A double-blind trial of an extract of the plant Serenoa repens in benign prostatic hyperplasia. Br. J. Clin. Pharm. 1984, 18, 461–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lobelenz, J. Extractum Sabal fructus for benign prostatic hyperplasia (BPH). Clinical trial in stages I and II. Therapeutikon 1992, 6, 34–37. (In German) [Google Scholar]
  65. Booker, A.; Suter, A.; Krnjic, A.; Strassel, B.; Zloh, M.; Said, M.; Heinrich, M. A phytochemical comparison of saw palmetto products using gas chromatography and (1) H nuclear magnetic resonance spectroscopy metabolomic profiling. J. Pharm. Pharmacol. 2014, 66, 811–822. [Google Scholar] [CrossRef] [Green Version]
  66. Feifer, A.H.; Fleshner, N.E.; Klotz, L. Analytical accuracy and reliability of commonly used nutritional supplements in prostate disease. J. Urol. 2002, 168, 150–154, discussion 154. [Google Scholar] [CrossRef]
  67. Habib, F.K.; Wyllie, M.G. Not all brands are created equal: A comparison of selected components of different brands of Serenoa repens extract. Prostate Cancer Prostatic Dis. 2004, 7, 195–200. [Google Scholar] [CrossRef]
  68. Penugonda, K.; Lindshield, B.L. Fatty acid and phytosterol content of commercial saw palmetto supplements. Nutrients 2013, 5, 3617–3633. [Google Scholar] [CrossRef] [Green Version]
  69. USP. Saw Palmetto Extract. Available online: https://online.uspnf.com/uspnf/document/1_GUID-53E14A4F-6F17-4CF1-852C-C6547F5A79DB_5_en-US (accessed on 1 November 2020).
  70. Suter, A.; Saller, R.; Riedi, E.; Heinrich, M. Improving BPH symptoms and sexual dysfunctions with a saw palmetto preparation? Results from a pilot trial. Phytother. Res. 2013, 27, 218–226. [Google Scholar] [CrossRef]
  71. Scaglione, F.; Lucini, V.; Pannacci, M.; Dugnani, S.; Leone, C. Comparison of the potency of 10 different brands of Serenoa repens extracts. Eur. Rev. Med. Pharm. Sci. 2012, 16, 569–574. [Google Scholar]
  72. Ooi, S.L.; Pak, S.C. Serenoa repens for Lower Urinary Tract Symptoms/Benign Prostatic Hyperplasia: Current Evidence and Its Clinical Implications in Naturopathic Medicine. J. Altern. Complement. Med. 2017, 23, 599–606. [Google Scholar] [CrossRef]
  73. Lin, P.H.; Freedland, S.J. Lifestyle and lower urinary tract symptoms: What is the correlation in men? Curr. Opin. Urol. 2015, 25, 1–5. [Google Scholar] [CrossRef] [Green Version]
  74. Raheem, O.A.; Parsons, J.K. Associations of obesity, physical activity and diet with benign prostatic hyperplasia and lower urinary tract symptoms. Curr. Opin. Urol. 2014, 24, 10–14. [Google Scholar] [CrossRef] [Green Version]
  75. Sanford, M.T.; Rodriguez, L.V. The role of environmental stress on lower urinary tract symptoms. Curr. Opin. Urol. 2017, 27, 268–273. [Google Scholar] [CrossRef] [PubMed]
  76. Ullrich, P.M.; Lutgendorf, S.K.; Kreder, K.J. Physiologic reactivity to a laboratory stress task among men with benign prostatic hyperplasia. Urology 2007, 70, 487–491, discussion 491–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Zhang, L.G.; Chen, J.; Meng, J.L.; Zhang, Y.; Liu, Y.; Zhan, C.S.; Chen, X.G.; Zhang, L.; Liang, C.Z. Effect of alcohol on chronic pelvic pain and prostatic inflammation in a mouse model of experimental autoimmune prostatitis. Prostate 2019, 79, 1439–1449. [Google Scholar] [CrossRef]
  78. Gharaee-Kermani, M.; Rodriguez-Nieves, J.A.; Mehra, R.; Vezina, C.A.; Sarma, A.V.; Macoska, J.A. Obesity-induced diabetes and lower urinary tract fibrosis promote urinary voiding dysfunction in a mouse model. Prostate 2013, 73, 1123–1133. [Google Scholar] [CrossRef] [Green Version]
  79. Hammarsten, J.; Hogstedt, B. Clinical, anthropometric, metabolic and insulin profile of men with fast annual growth rates of benign prostatic hyperplasia. Blood Press. 1999, 8, 29–36. [Google Scholar]
  80. Russo, G.I.; Regis, F.; Spatafora, P.; Frizzi, J.; Urzi, D.; Cimino, S.; Serni, S.; Carini, M.; Gacci, M.; Morgia, G. Association between metabolic syndrome and intravesical prostatic protrusion in patients with benign prostatic enlargement and lower urinary tract symptoms (MIPS Study). BJU Int. 2018, 121, 799–804. [Google Scholar] [CrossRef]
  81. Sarma, A.V.; Parsons, J.K.; McVary, K.; Wei, J.T. Diabetes and benign prostatic hyperplasia/lower urinary tract symptoms--what do we know? J. Urol. 2009, 182, S32–S37. [Google Scholar] [CrossRef] [PubMed]
  82. Sebastianelli, A.; Russo, G.I.; Kaplan, S.A.; McVary, K.T.; Moncada, I.; Gravas, S.; Chapple, C.; Morgia, G.; Serni, S.; Gacci, M. Systematic review and meta-analysis on the efficacy and tolerability of mirabegron for the treatment of storage lower urinary tract symptoms/overactive bladder: Comparison with placebo and tolterodine. Int. J. Urol. 2018, 25, 196–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Vignozzi, L.; Gacci, M.; Maggi, M. Lower urinary tract symptoms, benign prostatic hyperplasia and metabolic syndrome. Nat. Rev. Urol. 2016, 13, 108–119. [Google Scholar] [CrossRef] [PubMed]
  84. Yap, T.L.; Brown, C.T.; Emberton, M. Self-management in lower urinary tract symptoms: The next major therapeutic revolution. World J. Urol. 2006, 24, 371–377. [Google Scholar] [CrossRef] [PubMed]
  85. Yoo, S.; Oh, S.; Suh, J.; Park, J.; Cho, M.C.; Jeong, H.; Won, S.; Son, H. Optimal high-density lipoprotein cholesterol level for decreasing benign prostatic hyperplasia in men not taking statin medication: A historical cohort study. Prostate 2020, 80, 570–576. [Google Scholar] [CrossRef] [PubMed]
  86. Gandaglia, G.; Briganti, A.; Gontero, P.; Mondaini, N.; Novara, G.; Salonia, A.; Sciarra, A.; Montorsi, F. The role of chronic prostatic inflammation in the pathogenesis and progression of benign prostatic hyperplasia (BPH). BJU Int. 2013, 112, 432–441. [Google Scholar] [CrossRef]
  87. Kramer, G.; Mitteregger, D.; Marberger, M. Is benign prostatic hyperplasia (BPH) an immune inflammatory disease? Eur. Urol. 2007, 51, 1202–1216. [Google Scholar] [CrossRef]
  88. Kwon, Y.K.; Choe, M.S.; Seo, K.W.; Park, C.H.; Chang, H.S.; Kim, B.H.; Kim, C.I. The effect of intraprostatic chronic inflammation on benign prostatic hyperplasia treatment. Korean J. Urol. 2010, 51, 266–270. [Google Scholar] [CrossRef] [Green Version]
  89. Lee, C.L.; Kuo, H.C. Pathophysiology of benign prostate enlargement and lower urinary tract symptoms: Current concepts. Tzu-Chi Med. J. 2017, 29, 79–83. [Google Scholar]
  90. Robert, G.; Descazeaud, A.; Nicolaiew, N.; Terry, S.; Sirab, N.; Vacherot, F.; Maille, P.; Allory, Y.; de la Taille, A. Inflammation in benign prostatic hyperplasia: A 282 patients’ immunohistochemical analysis. Prostate 2009, 69, 1774–1780. [Google Scholar] [CrossRef] [Green Version]
  91. Steiner, G.E.; Newman, M.E.; Paikl, D.; Stix, U.; Memaran-Dagda, N.; Lee, C.; Marberger, M.J. Expression and function of pro-inflammatory interleukin IL-17 and IL-17 receptor in normal, benign hyperplastic, and malignant prostate. Prostate 2003, 56, 171–182. [Google Scholar] [CrossRef]
  92. Zlotta, A.R.; Egawa, S.; Pushkar, D.; Govorov, A.; Kimura, T.; Kido, M.; Takahashi, H.; Kuk, C.; Kovylina, M.; Aldaoud, N.; et al. Prevalence of inflammation and benign prostatic hyperplasia on autopsy in Asian and Caucasian men. Eur. Urol. 2014, 66, 619–622. [Google Scholar] [CrossRef]
  93. Ficarra, V.; Rossanese, M.; Zazzara, M.; Giannarini, G.; Abbinante, M.; Bartoletti, R.; Mirone, V.; Scaglione, F. The role of inflammation in lower urinary tract symptoms (LUTS) due to benign prostatic hyperplasia (BPH) and its potential impact on medical therapy. Curr. Urol Rep. 2014, 15, 463. [Google Scholar] [CrossRef]
  94. Bernichtein, S.; Pigat, N.; Camparo, P.; Latil, A.; Viltard, M.; Friedlander, G.; Goffin, V. Anti-inflammatory properties of Lipidosterolic extract of Serenoa repens (Permixon(R)) in a mouse model of prostate hyperplasia. Prostate 2015, 75, 706–722. [Google Scholar] [CrossRef]
  95. Gravas, S.; Samarinas, M.; Zacharouli, K.; Karatzas, A.; Tzortzis, V.; Koukoulis, G.; Melekos, M. The effect of hexanic extract of Serenoa repens on prostatic inflammation: Results from a randomized biopsy study. World J. Urol. 2019, 37, 539–544. [Google Scholar] [CrossRef]
  96. Latil, A.; Libon, C.; Templier, M.; Junquero, D.; Lantoine-Adam, F.; Nguyen, T. Hexanic lipidosterolic extract of Serenoa repens inhibits the expression of two key inflammatory mediators, MCP-1/CCL2 and VCAM-1, in vitro. BJU Int. 2012, 110, E301–E307. [Google Scholar] [CrossRef]
  97. Sirab, N.; Robert, G.; Fasolo, V.; Descazeaud, A.; Vacherot, F.; de la Taille, A.; Terry, S. Lipidosterolic extract of serenoa repens modulates the expression of inflammation related-genes in benign prostatic hyperplasia epithelial and stromal cells. Int. J. Mol. Sci. 2013, 14, 14301–14320. [Google Scholar] [CrossRef] [Green Version]
  98. Vela Navarrete, R.; Garcia Cardoso, J.V.; Barat, A.; Manzarbeitia, F.; López Farré, A. BPH and Inflammation: Pharmacological Effects of Permixon on Histological and Molecular Inflammatory Markers. Results of a Double Blind Pilot Clinical Assay. Eur. Urol. 2003, 44, 549–555. [Google Scholar] [CrossRef]
  99. O’Leary, M.P. Validity of the “bother score” in the evaluation and treatment of symptomatic benign prostatic hyperplasia. Rev. Urol. 2005, 7, 1–10. [Google Scholar]
  100. O’Leary, M.P.; Wei, J.T.; Roehrborn, C.G.; Miner, M.; Registry, B.P.H.; Patient Survey Steering, C. Correlation of the International Prostate Symptom Score bother question with the Benign Prostatic Hyperplasia Impact Index in a clinical practice setting. BJU Int. 2008, 101, 1531–1535. [Google Scholar] [CrossRef] [PubMed]
  101. Brown, C.T.; van der Meulen, J.; Mundy, A.R.; O’Flynn, E.; Emberton, M. Defining the components of a self-management programme for men with uncomplicated lower urinary tract symptoms: A consensus approach. Eur. Urol. 2004, 46, 254–262, discussion 263. [Google Scholar] [CrossRef]
  102. Hashim, H.; Abrams, P. How should patients with an overactive bladder manipulate their fluid intake? BJU Int. 2008, 102, 62–66. [Google Scholar] [CrossRef] [Green Version]
  103. Weiss, J.P.; Juul, K.V.; Wein, A.J. Management of nocturia: The role of antidiuretic pharmacotherapy. Neurourol. Urodyn. 2014, 33 (Suppl. S1), S19–S24. [Google Scholar] [CrossRef]
  104. Yap, T.L.; Brown, C.; Cromwell, D.A.; van der Meulen, J.; Emberton, M. The impact of self-management of lower urinary tract symptoms on frequency-volume chart measures. BJU Int. 2009, 104, 1104–1108. [Google Scholar] [CrossRef] [PubMed]
  105. Castro, J.E. Pills for the benign prostate. Trial designs. Proc. R Soc. Med. 1972, 65, 126–127. [Google Scholar]
  106. Vaughan, C.P.; Johnson, T.M., II; Haukka, J.; Cartwright, R.; Howard, M.E.; Jones, K.M.; Markland, A.D.; Goode, P.S.; Burgio, K.L.; Tikkinen, K.A. The fluctuation of nocturia in men with lower urinary tract symptoms allocated to placebo during a 12-month randomized, controlled trial. J. Urol. 2014, 191, 1040–1044. [Google Scholar] [CrossRef] [PubMed]
  107. Russo, G.I.; Scandura, C.; Di Mauro, M.; Cacciamani, G.; Albersen, M.; Hatzichristodoulou, G.; Fode, M.; Capogrosso, P.; Cimino, S.; Marcelissen, T.; et al. Clinical Efficacy of Serenoa repens Versus Placebo Versus Alpha-blockers for the Treatment of Lower Urinary Tract Symptoms/Benign Prostatic Enlargement: A Systematic Review and Network Meta-analysis of Randomized Placebo-controlled Clinical Trials. Eur. Urol. Focus 2021, 7, 420–431. [Google Scholar] [CrossRef] [PubMed]
  108. De Nunzio, C.; Novara, G.; Damiano, R.; Bartoletti, R.; Tubaro, A.; Ficarra, V.; members of the Research Urological Network; Russo, G.I.; Scandura, C.; Di Mauro, M.; et al. Clinical Efficacy of Serenoa repens Versus Placebo Versus Alpha-blockers for the Treatment of Lower Urinary Tract Symptoms/Benign Prostatic Enlargement: A Systematic Review and Network Meta-analysis of Randomized Placebo-controlled Clinical Trials. Eur Urol Focus. In press. https://doi.org/10.1016/j.euf.2020.01.002: New Evidence Changing Clinical Practice or Misunderstanding of Statistical Analyses? The Case of Serenoa repens and alpha-Blockers. Eur. Urol. Focus 2020. [Google Scholar] [CrossRef]
  109. Agbabiaka, T.B.; Pittler, M.H.; Wider, B.; Ernst, E. Serenoa repens (saw palmetto): A systematic review of adverse events. Drug Saf. 2009, 32, 637–647. [Google Scholar] [CrossRef]
  110. Aliaev Yu, G.; Vinarov, A.Z.; Lokshin, K.L.; Spivak, L.G. Efficiency and safety of prostamol-Uno in patients with chronic abacterial prostatitis. Urologiia 2006, 1, 47–50. (In Russian) [Google Scholar]
  111. Avins, A.L.; Bent, S.; Staccone, S.; Badua, E.; Padula, A.; Goldberg, H.; Neuhaus, J.; Hudes, E.; Shinohara, K.; Kane, C. A detailed safety assessment of a saw palmetto extract. Complement. Med. 2008, 16, 147–154. [Google Scholar] [CrossRef] [Green Version]
  112. Bach, D.; Ebeling, L. Long-term drug treatment of benign prostatic hyperplasia-results of a prospective 3-year multicenter study using Sabal extract IDS 89. Phytomedicine 1996, 3, 105–111. (In German) [Google Scholar] [CrossRef]
  113. Braeckman, J.; Bruhwyler, J.; Vanderkerckhove, K.; Géczy, J. Efficacy and safety of the extract of Serenoa repens in the treatment of benign prostatic hyperplasia: Therapeutic equivalence between twice and once daily dosage forms. Phytother. Res. 1997, 11, 558–563. [Google Scholar] [CrossRef]
  114. Pytel, Y.A.; Lopatkin, N.A.; Gorilovskii, L.M.; Vinarov, A.Z.; Sivkov, A.V.; Medvedev, A.A. The results of long-term permixon treatment in patients with symptoms of lower urinary tracts dysfunction due to benign prostatic hyperplasia. Urologiia 2004, 2, 3–7. (In Russian) [Google Scholar]
  115. Sinescu, I.; Geavlete, P.; Multescu, R.; Gangu, C.; Miclea, F.; Coman, I.; Ioiart, I.; Ambert, V.; Constantin, T.; Petrut, B.; et al. Long-term efficacy of Serenoa repens treatment in patients with mild and moderate symptomatic benign prostatic hyperplasia. Urol. Int. 2011, 86, 284–289. [Google Scholar] [CrossRef] [PubMed]
  116. Gratzke, C.; Bachmann, A.; Descazeaud, A.; Drake, M.J.; Madersbacher, S.; Mamoulakis, C.; Oelke, M.; Tikkinen, K.A.O.; Gravas, S. EAU Guidelines on the Assessment of Non-neurogenic Male Lower Urinary Tract Symptoms including Benign Prostatic Obstruction. Eur. Urol. 2015, 67, 1099–1109. [Google Scholar] [CrossRef]
  117. Laekeman, G.; Vlietinck, A. Assessment Report on Serenoa Repens (W. Bartram) Small, Fructus. Available online: http://www.ema.europa.eu/docs/en_GB/document_library/Herbal_-_HMPC_assessment_report/2014/12/WC500179593.pdf (accessed on 14 November 2020).
  118. Akbulut, Z.; Tekdoğan, Ü.Y.; Tekin, A.; Gürbüz, C.; Atan, A.; Şengör, F.; Çaşkurlu, T.; Balbay, M.D. Efficacy and tolerability of Serenoa repens extract (Prostagood) in patients wlth lower urinary tract symptoms due to symptomatic benign prostatic hyperplasia. Turk. J. Urol. 2009, 35, 4. [Google Scholar]
Figure 1. The Duck Principle: “But when I see a bird that quacks like a duck, walks like a duck, has feathers and webbed feet and associates with ducks, I’m certainly going to assume that he IS a duck.”—Emil Mazey Secretary-Treasurer UAW Labor leader 1946.
Figure 1. The Duck Principle: “But when I see a bird that quacks like a duck, walks like a duck, has feathers and webbed feet and associates with ducks, I’m certainly going to assume that he IS a duck.”—Emil Mazey Secretary-Treasurer UAW Labor leader 1946.
Uro 01 00017 g001
Table 1. The Bent (STEP) and Barry (CAMUS) studies are the preeminent placebo-controlled trials of LSESr vs. LUTS with negative results.
Table 1. The Bent (STEP) and Barry (CAMUS) studies are the preeminent placebo-controlled trials of LSESr vs. LUTS with negative results.
Author (Lead)YearExtraction ProcessPt. #Study (mos)IPSSBPHIIQmaxFatty Acids %
Δ%Δ%Δ%
Bent2006CO210212−0.74−0.310+0.4492 TFA
Placebo104−0.75−0.13−0.00
Barry2011Ethanol15118−2.215−0.824−0.2−154 FFA
Placebo155−3.020−1.033−0.8−5
Values for IPSS and BPHII are rounded off to one decimal point; percentages for all are rounded off to the nearest whole number. IPSS, International Prostate Symptom Score; BPHII, BPH impact index; Qmax, peak urinary flow (mL/s); Δ, mean change; mos., months; #, number; Pt., patients; %, percent; TFA, total fatty acids; FFA, free fatty acids.
Table 2. The striking difference in the endpoints of IPSS, QoL and Qmax for Barry [2] and Derakhshani [4] using the identical ethanolic extraction product of Serenoa repens (Prosta Urgenin® Uno).
Table 2. The striking difference in the endpoints of IPSS, QoL and Qmax for Barry [2] and Derakhshani [4] using the identical ethanolic extraction product of Serenoa repens (Prosta Urgenin® Uno).
Author
(Lead)
YearExtraction ProcessPt. #Study
(mos)
IPSS *QoLQmaxFatty Acids %
Δ%Δ%Δ%
Barry2011Ethanol15118−2.215−0.411−0.2−154 FFA
Placebo155−3.020−0.515−0.8−5
Derakhshani1997Ethanol14613−7.440−1.646+3.73154 FFA
No Placebo
* Values for IPSS, BPHII and Qmax are rounded off to one decimal point and percentages to the nearest whole number. IPSS, International Prostate Symptom Score; QoL, quality of life; Qmax, peak urinary flow (mL/s); Δ, mean change; mos, months; #, number; Pt., patient; %, percent; FFA, free fatty acids.
Table 3. Clinical evaluation of HESr or EESr vs. Tamsulosin (Flomax®) in five studies and HESr vs. Finasteride (Proscar®) in one study.
Table 3. Clinical evaluation of HESr or EESr vs. Tamsulosin (Flomax®) in five studies and HESr vs. Finasteride (Proscar®) in one study.
Lead Author
Year, Ref. [#]
Study Duration (mos)Study ArmPatients (#) aIPSS *QoL *Qmax *
Δ%Δ%mL/s%
Debruyne 2002 [10] 12HESr350−4.428NRNR+1.917
Tam354−4.429NRNR+1.816
Latil 2015 [11]3HESr83−4.525−0.923+1.715
Tam86−6.539−1.334+2.120
Alcaraz 2020 [12] ‡6HESr222−5.630−1.334+3.325
Tam222−5.932−1.436+2.823
Combo159−7.337−1.846+2.116
Carraro 1996 [13]6.5HESr467 −5.8 37−1.438+2.725
Fin484 −6.1 39−1.541+3.230
Hizli 2007 [14]6EESr20 −6.1 34−2.662+3.234
Tam20 −4.6 28−2.160+3.735
Combo20 −4.9 31−2.263+4.242
Argirovic 2013 [15]6EESr97 −6.1 34−2.638+3.234
Tam87 −4.6 28−2.140+3.735
Combo81 −4.9 31−2.237+4.245
a Number of patients at study end. * Values rounded off to one decimal point. Percentages are rounded off to the nearest whole number. ‡ In Alcaraz 2020 the number of patients for Qmax were 43, 33 and 46 for HESr, tamsulosin and the combination, respectively. Data is per protocol (PP) obtained from supplementary material available at https://www.mdpi.com/2077-0383/9/9/2909/s1, accessed on 30 July 2021). Δ, mean change; −, negative change; #, number; %, percent change; +, positive change; AUA, American Urological Association; IPSS, International Prostate Symptom Score; EESr, ethanolic extract of Serenoa repens; Fin, finasteride; HESr, hexanic extract of Serenoa repens; LUTS, lower urinary tract symptoms; mL/s, milliliters per second; NR, not reported; mos, months; QoL, quality of life; Qmax, peak urinary flow (mL/s); Ref., reference citation; Tam, tamsulosin.
Table 4. Comparative changes in IPSS with Permixon vs. tamsulosin from PERMAL (adapted after Debruyne 2002 [10] and Debruyne 2004 [17]).
Table 4. Comparative changes in IPSS with Permixon vs. tamsulosin from PERMAL (adapted after Debruyne 2002 [10] and Debruyne 2004 [17]).
IPSS ValuePERMAL 2002PERMAL 2004
Δ (%) IPSSΔ (%) IPSS
PermixonTamsulosinPermixonTamsulosin
>10−4.4 (28%)−4.4 (29%)
>19 a −7.8 (35%)−5.8 (25%)
=20–21 (all)−6.9−5.5
=20–21 irritative−2.5−2.0
=20–21 obstructive−4.4−3.5
>21 (all)−9.3−6.0
>21 irritative−3.5−1.9
>21 obstructive−5.8−4.1
a For patients with a baseline IPSS > 19, Permixon was superior to tamsulosin (p = 0.051). The greatest change in IPSS (e.g., ≥9 points) was seen in patients receiving Permixon vs. tamsulosin (41.5% vs. 25.4%), respectively. Δ, mean change in IPSS from baseline to study end; −, decrease in IPSS from baseline; %, percent improvement in IPSS from baseline; IPSS, International Prostate Symptom Score.
Table 5. Thirty-one LSESr vs. LUTS English-language studies meeting evaluability criteria. The three negative studies are shown in red font.
Table 5. Thirty-one LSESr vs. LUTS English-language studies meeting evaluability criteria. The three negative studies are shown in red font.
Author
(Lead)
YearExtraction
(Method)
Pt. #Duration
(mos)
IPSSQoLQmaxFatty Acids%
Δ%Δ%Δ%
Carraro α1996Hexane4676−5.837−1.438+2.72581
Stepanov β1999Hexane923−6.433−1.026+1.61881
Al-Shukri2000Hexane572−2.2 27−0.618+0.7681
Debruyne γ2002Hexane35012−4.428 +1.91781
Giannakopoulos δ 2002Hexane1006−8.040−0.617+3.74081
Pytel ε2002Hexane11624−5.342−1.340+1.21081
Debruyne θ2004Hexane12412−7.835−1.229+1.21181
El-Demiry ι2004Hexane1906−11.451 +4.44581
Djavan Ω2005Hexane8824−1.0 17−0.419+1.81581
Giulianelli 2012Hexane 5916-5.632 +3.02881
Latil κ2015Hexane833−4.525−0.923+1.71581
Robert 2015Hexane1022−4.525 81
Alcaraz 2020Hexane2226−5.630−1.334+3.32581
Hexane Averages n= 122079−5.533−0.926+2.22181
Gerber λ1998Ethanol466−7.637 −0.7−540
Hizli μ2007Ethanol206−6.134−2.662+3.23481
Barry Ϸ 2011Ethanol15118−2.215 54
Gerber ν2001Ethanol396−4.426−0.721+1.01041
Sinescu π 2011Ethanol12024−5.540−1.850+5.65459
Argirovic ρ2013Ethanol976−6.134−2.638+3.23459
Cai2013Ethanol463−3.118 +0.54--
Suter2013Ethanol692 −7.552 95
Saidi2019Ethanol4012−2.118 +0.86 59
Vinarov 2019Ethanol30180−6.050−3.060+5.04559
Ye2019Ethanol1596−4.429−1.226+4.13668
ETOH Averages n= 117425−5.032−1.843+2.52762
Romics1993CO2 3112 +4.33955
Bach φ1996CO2 31536 73 +6.14655
Kondas χ1996CO2 386 +4.13955
Braeckman 1994CO2 3053−6.635−1.542+2.12674
Braeckman ω 1997CO2 6712−10.260−1.542+2.62474
Braeckman ψ1997CO2 1253 64 3074
Willetts 2003CO2 463−1.18−0.513.0+2.4----
Bent2006CO2 10212−0.74 +0.4492
CO2 Averages n= 812910.9−4.741−1.232+3.23068
Averages All 19415.0−5.135−1.334+2.62670
IPSS, QoL & Qmax values are rounded off to one decimal point. Percentages are rounded off to the nearest whole number. The Hutchison 2007 study was not shown because it was a group analysis, but it is a valuable study. Since Latil 2015 and Robert 2015 contain identical content that was reported in two different journals, Robert 2015 was arbitrarily excluded from the table.
α Carraro study of Permixon vs. finasteride. HESr showed equivalent efficacy to 5ARI with fewer side effects.
β Stepanov study comparing Permixon at 160 mg bid vs. 160 mg ×2 once a day. Average results used.
γ Debruyne 2002 study of Permixon vs. tamsulosin study with 4-week run-in phase. No significant differences in the effect of Permixon vs. tamsulosin 0.4 mg/day.
δ Giannakopoulos study compared 160 mg bid vs. 160 tid. The results shown are the average of both findings. Qmax with 480 mg/day +4.54 vs. +2.8 for 320 mg/day.
ε Pytel reported that 46–69% of patients reported improvement in obstructive and irritative symptoms from month-6 to the study’s end at 2 years.
θ Debruyne 2004 subset analysis of high >19 IPSS patients with randomization between Permixon vs. tamsulosin.
ι El-Demiry is an abstract but with solid data. Ω Djavan study on prevention of progression of LUTS from mild to greater than mild; Permixon vs. WW.
Κ Latil study comparing Permixon vs. tamsulosin and correlations with inflammation.
λ Gerber 1998 noted improvement at 2 months. At 6 months, 46% of patients with ≥50% (21/46) improvement.
μ Hizli 2007 study comparing Prostagood® (ethanol extraction) vs. tamsulosin vs. Prostagood + tamsulosin. All groups with no significant differences in efficacy; Prostagood + tamsulosin did not increase efficacy.
Ϸ Barry study is a negative study and the placebo group had Δ in IPSS of −2.99 or 20% improvement.
ν Gerber 2001 study a with one-month placebo run-in for all patients.
π Sinescu used Prostamol Uno.
ρ Argirovic study compared Prostamol Uno 320 mg/day vs. tamsulosin vs. tamsulosin + Prostamol uno; percentage improvements were 33.9% vs. 28.4% vs. 31.4%, respectively for IPSS. Results were 38% vs. 40% vs. 37% for QoL; and for Qmax they were 34% vs. 35% vs. 44.5%, respectively.
φ Bach 1996 3-year study quantitated nocturia, frequency, and incomplete emptying. Nocturia improved 73%, and no nocturia or nocturia ×1 increased from 33% to 85%. Improvements in frequency and incomplete emptying of 54% and 76%, respectively.
χ Kondas used Strogen® Forte, aka Sabal IDS 89, (Strathmann GmbH & Co. KG, Hamburg, Germany). The authors stated they measured IPSS but did not report results.
ω Braeckman 1997 study #1 used Prostaserene® as LSESr. QoL is not from IPSS but only a rating scale. Only 67 patients completed the study, with 34 patients receiving LSESr at 160 mg bid and 33 patients receiving 320 mg/day.
ψ Braeckman 1997 study with calculations done by SBS. For placebo, IPSS improved 25% and Qmax improved 10%.
Table 6. Evaluable non-English-language papers (27 studies) categorized by extraction method. The mean number of patients, duration of the study, and the key clinical outcome assessments are detailed. The extensive footnotes report key findings of individual studies.
Table 6. Evaluable non-English-language papers (27 studies) categorized by extraction method. The mean number of patients, duration of the study, and the key clinical outcome assessments are detailed. The extensive footnotes report key findings of individual studies.
Lead AuthorRef. [#]YearExtraction
Method
Serenoa
Patients (#) a
Study Duration(mos)IPSSQoLQmax
Δ% bΔ%Δ%
Cirillo-Marucco[24]1983Hexane474 56 ε +4.650 ε
Cukier ψ[25]1985Hexane732 33 λ
Tosto[26]1985Hexane203−5.028 Ω
Pannunzio[27]1986Hexane302 +5.174
Pescatore[28]1986Hexane303 +2.527
Authie[29]1987Hexane5003 78 π
Ollé Carreras[30]1987Hexane402 68 φ
Orfei[31]1988Hexane303 50 χ−2.2 +0.00.2
Dathe[32]1991Hexane496 +5.949
Aliaev[33]2002Hexane2660−8.876−1.353+4.135
Foroutan[34]1997Hexane5923−6.538−1.545+5.966
Medeiros α[35]2000Hexane1303−6.537−1.439+2.022
Totals Hexane (12) Averages1317.8−6.752−1.646+3.840
Derakhshani[4]1997Ethanol10473−7.440−1.646+3.731
Eickenberg *[20]1997Ethanol 69676−8.044−1.838+3.023
Redecker **[36]1998Ethanol503 48 ν +3.424
Ziegler ** β[37]1998Ethanol1093 36+3.729
Breza[18]2005Ethanol59612−5.936−1.754+2.319
Aliaev[38]2007Ethanol506−3.026−1.843+1.714
Razumov[39]2007Ethanol306−6.943−2.768+2.823
Aliaev γ[40]2009Ethanol5024−4.237−2.252+2.721
Vinarov[41]2010Ethanol5036−6.050−2.050+4.539
Aliaev[42]2013Ethanol38120−1.312−1.135+3.326
Totals Ethanol (10) Averages89922−5.337−1.947+3.125
Mattei ψ[43]1990CO2203 55 ω
Vahlensieck[44]1993CO213344 39;55 Ϸ
Vahlensieck[45]1993CO24003 94 θ +5.852
Fabricius δ[46]1993CO21536 39;58 δ
Bauer ψ[47]1999CO21016 37 ρ 16
Totals CO2 (5) Averages4024.4 34
Mean Across All Studies (n = 27)
Hexane extraction (n = 12)
Ethanol extraction (n = 10)
Carbon dioxide extraction (n = 5)
47712−6.045%−1.747%+3.533%
The clinical endpoints of IPSS, QoL and Qmax are rounded off to two significant digits. Percentages are rounded off to the nearest whole number.
≈, approximately; Δ, mean change; −, negative change; #, number; %, percent change; +, positive change; CO2, carbon dioxide; IPSS, International Prostate Symptom Score; mos, months; QoL, quality of life; Qmax, peak urinary flow (mL/s); Ref., citation reference.
a The number of patients at study end, or as reported.
ψ Placebo-controlled study. The study by Bauer was also double-blinded and randomized.
α Medeiros study used a QoL scale 6 (worst) to 1 (best) rather than 6 (worst) and 0 (best).
* Eickenberg used a 96% EESr.
** Redecker & Ziegler used a 90% EESr.
β Ziegler did not use IPSS, so his reported symptoms were based on % improvement involving weak stream, hesitancy, incomplete emptying, frequency, and nocturia.
γ Aliaev 2009 is a 2-year extension of the 6-month 2007 paper.
δ Fabricius 1993 reported decreases in frequency and nocturia of 39%, and 58%, respectively. Nocturia ≤ 1 in 16% pre-LSESr vs. 79% at end of study (n = 153).
ε Cirillo-Marucco study done before IPSS; raw data on nocturia; the study also included Qmax results.
λ Cukier study done before IPSS; only raw data on nocturia.
Ω Tosto study done before IPSS; authors used a unique point scoring to evaluate frequency, nocturia, incomplete emptying, weak stream.
π Authie study before IPSS use; nocturia, frequency, and urgency improvements were 82%, 67%, and 85.3%, respectively (average improvement 78.1%); average complete resolution of these symptoms was 43.5%.
φ Ollé Carreras did not use IPSS. The number shown is based on the change in frequency with complete resolution in 27 out of 40 patients.
χ Orfei used scores from frequency, nocturia, urgency, weak stream, and straining at the beginning and end of the study
ν Redecker data evaluated nocturia before and after LSESr.
ω Mattei used scores from frequency, nocturia, and incomplete emptying. For these three endpoints, average improvement 55% vs. placebo average improvement of 1.4%.
Ϸ Vahlensieck did not use IPSS. The data reflects the change in frequency and nocturia before and after LSESr. Frequency improved by 39% and nocturia by 55%.
Θ Vahlensieck 2nd study reported an average decrease in frequency episodes of 94%. For nocturia, 59.5% of patients had ≤ 1 episode at end of the study vs. 9.7% at the start of the study.
Ρ Bauer only indicated percentage improvement. Talso® Uno with 37% vs. 13% for placebo.
Table 7. Summary of mean outcome for evaluable studies grouped by extraction technology for IPSS, QoL, and Qmax. Data are shown for all studies, and also for only the positive studies. All 24 HESr studies were positive. The three negative studies were Willetts 2003 (CO2) [21], Bent 2006 (CO2) [1], and Barry 2019 (Ethanol) [2].
Table 7. Summary of mean outcome for evaluable studies grouped by extraction technology for IPSS, QoL, and Qmax. Data are shown for all studies, and also for only the positive studies. All 24 HESr studies were positive. The three negative studies were Willetts 2003 (CO2) [21], Bent 2006 (CO2) [1], and Barry 2019 (Ethanol) [2].
Extraction
Technology
Mean Patients #Mean Study
Duration (mos)
Included StudiesIPSSQoLQmaxTypical FFA %
Δ%Δ%mL/s%
Hexane n = 242458.5 All positive−5.841−1.132+2.929Min ≈ 80
Ethanol n = 21
(1 negative)
47723 All studies †−5.134−1.845+2.825Min ≈ 70
Positive only−5.336
CO2 n = 13
(2 negative)
2288.4 All studies−4.643−1.232+3.531Min ≈ 65–70
Positive only−8.453−1.542+4.234
† The one negative ethanol study (Barry) did not significantly alter the IPSS outcome. The free fatty acid (FFA) minimums are typical values for lipidosterolic products (data on file 2021, Valensa International). The clinical endpoints of IPSS, QoL and Qmax are rounded off to two significant digits. Percentages are rounded off to the nearest whole number. ≈, approximately; Δ, mean change; −, negative change; %, percent; +, positive change; CO2, carbon dioxide; FFA, free fatty acid; IPSS, International Prostate Symptom Score; mL/s, milliliters per second; mos, months; QoL, quality of life; Qmax, peak urinary flow (mL/s).
Table 8. Summary of 17 placebo-controlled LSESr clinical trials. Eight studies (bolded) did not meet our criteria for evaluability for LSESr vs. LUTS/BPH.
Table 8. Summary of 17 placebo-controlled LSESr clinical trials. Eight studies (bolded) did not meet our criteria for evaluability for LSESr vs. LUTS/BPH.
Lead AuthorYearStudyExtractionProductSerenoa Patients δPlacebo Patients δStudy Duration
(mos)
Boccafoschi1983D, PHexanePermixon11112
Emili1983D, PHexanePermixon15151
Mandressi1983D, PHexanePermixon 19151
Champault1984D, PHexanePermixon50441
Tasca1985D, PHexanePermixon 14132
Reece Smith1986D, PHexanePermixon 33373
Löbelenz ‡1992PEthanolSabal Extract30301.5
Descotes1995D, PHexanePermixon 82941
Cukier1985D, PHexanePermixon71762.5
Mattei1990D, PCO2Talso®20203
Braeckman1997D, R, PCO2Prostaserene1251133
Bauer *1999D, RCO2Talso® Uno1016
Gerber2001D, REthanolSolaray®39406
Willetts2003R, CCO2Proseren®46473
Bent2006D, PCO2Not stated10210412
Barry2011D, PEthanolProsta Urgenin Uno15117018
Ye2019D, PEthanolProstess® Uno1591696
IPSS, QoL & Qmax values are rounded off to one decimal point. Percentages are rounded off to the nearest whole number. δ The number of patients at study end, or as reported. ‡ Löbelenz study was only six weeks in duration. * Bauer study presented the sum of the Talso Uno and placebo patients. mos, months; D, double-blind; R, randomized; P, placebo-controlled; CO2, carbon dioxide.
Table 9. Efficacy of HESr (Permixon) vs. placebo in seven arbitrarily defined “non-evaluable” studies in Cochrane 2012. All seven studies reported clinical improvement in symptoms vs. placebo.
Table 9. Efficacy of HESr (Permixon) vs. placebo in seven arbitrarily defined “non-evaluable” studies in Cochrane 2012. All seven studies reported clinical improvement in symptoms vs. placebo.
Author
(Lead)
YearRef. [#]Serenoa
Patients (#) δ
Study Duration (mos)Key Results for Serenoa vs. Placebo or Comparator
Boccafoschi1983[52]112Qmax +4.2 (42%) vs. placebo +2.1 (20.6%)
Emili1983[53]151Qmax +3.56 (34.5%) vs. placebo +0.20 (2.2%)
Mandressi1983[56]191Serenoa vs. Pygeum vs. placebo; urgency 70% vs. 62% vs. 24%; frequency 30% vs. 22% vs. 10%; nocturia 42% vs. 38% vs. −4%
Champault1984[63]501Qmax +2.7 (50.5%) vs. placebo +0.25 (5%); nocturia −1.53 (49%) vs. placebo −0.48 (15%)
Tasca1985[54]142Qmax +3.3 (25.6%) vs. placebo −0.6 (−5%); nocturia 74.3% vs. 38.7%; urgency 60% vs. 20%; weak stream 50% vs. 16.6%
Löbelenz 1992[64]301.5Qmax +1.2 (9.8%) vs. placebo +0.6 (4.6%)
Descotes1995[55]821Qmax +3.4 (28.9%) vs. placebo +1.1 (8.9%)
Mean Across All Studies for Clinical OutcomeQmax +3.0 (32%); ↓ nocturia 55%; ↓ urgency 65%
δ The number of patients at study end, or as reported. Löbelenz study did not specify how many patients in LSESr cohort at the end of the study. ↓, decreased; −, negative change; #, number; +, positive change; mos, months; Qmax, peak urinary flow (mL/s); Ref., reference citation; vs., versus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Strum, S.B. Serenoa Repens (Saw Palmetto) for Lower Urinary Tract Symptoms (LUTS): The Evidence for Efficacy and Safety of Lipidosterolic Extracts. Part III. Uro 2021, 1, 155-179. https://doi.org/10.3390/uro1030017

AMA Style

Strum SB. Serenoa Repens (Saw Palmetto) for Lower Urinary Tract Symptoms (LUTS): The Evidence for Efficacy and Safety of Lipidosterolic Extracts. Part III. Uro. 2021; 1(3):155-179. https://doi.org/10.3390/uro1030017

Chicago/Turabian Style

Strum, Stephen B. 2021. "Serenoa Repens (Saw Palmetto) for Lower Urinary Tract Symptoms (LUTS): The Evidence for Efficacy and Safety of Lipidosterolic Extracts. Part III" Uro 1, no. 3: 155-179. https://doi.org/10.3390/uro1030017

Article Metrics

Back to TopTop