Skip to main content
  • Research article
  • Open access
  • Published:

RETRACTED ARTICLE: Observational study: daily treatment with a new compound “tradamixina” plus serenoa repens for two months improved the lower urinary tract symptoms

This article was retracted on 28 April 2016

Abstract

Background

Lower urinary tract symptoms (LUTS) are associated with great emotional costs to individuals and substantial economic costs to society. This study seeks to evaluate the effect of a new natural compound “Tradamixina plus Serenoa Repens” in order to improve lower urinary tract symptoms.

Methods

100 patients (≥45years) who had had LUTS/BPH for >6 mo at screening and with IPSS -The international Prostate symptom scores- ≥13 and maximum urinary flow rate (Qmax) ≥4 to ≤15 ml/s. were recruited. The compound “Tradamixina plus Serenoa Repens” (80 mg of Alga Ecklonia Bicyclis, 100 mg of Tribulus Terrestris and 100 mg of D-Glucosamine and N-Acetyl-D-Glucosamine plus 320 mg of Serenoa Repens) was administered daily for 2 months. At visit and after 60 days of treatment patients were evaluated by means of detailed medical urological history, clinical examination, laboratory investigations (total PSA), and instrumental examination like urolfowmetry. Efficacy measures included IPSS-International Prostate Sympto, BPH Impact Index (BII), Quality-of-Life (QoL) Index. Measures were assessed at baseline and end point (12 wk or end of therapy) and also at screening, 1 and 4 wk for IPSS, and 4 wk for BII. Statistical significance was interpreted only if the results of the preceding analysis were significant at the 0.05 level.

Results

After 2 months of treatment the change from baseline to week 12 relative to “Tradamixina plus Seronea Repens” in total IPSS and Qol was statistically significant. Differences from baseline in BII were statistically significant for “Tradamixina plus Seronea Repens” above all differences in BII were also significant at 4 wk (LSmean ± SE: -0.8 ± 0.2). In the distribution of subjects over the PGI-I and CGI-I response categories were significant for”Tradamixina plus Seronea Repens” (PGI-I: p = 0.001; CGI-I). We also observed a decrease of total PSA.

Conclusion

The daily treatment with a new compound “Tradamixina plus Serenoa Repens” for 2 months improved the male sexual function , it improved the bother symptoms which affect the patient’s quality of life , improved uroflowmetric parameters, and we also observed a decrease of serum PSA level.

Background

Lower urinary tract symptoms (LUTS) are associated with great emotional costs [1] to individuals and substantial economic costs to society [2]. The prevalence and severity of LUTS increases with age [3], and the progressive growth of the aged population group has broadened the societal impact of LUTS. LUTS comprise storage symptoms (daytime urinary frequency, nocturia, urgency, urinary incontinence) voiding symptoms (slow stream, splitting or spraying, intermittency, hesitancy, straining, terminal dribble), and post micturition symptoms (sensation of incomplete emptying, post micturition dribble) [4] In EPIC, 62.5% of 7210 men in the five countries surveyed reported one or more LUTS; the prevalence of individual LUTS in men increased with age. A greater percentage of men reported storage symptoms (51.3%) vs. voiding (25.7%) or post micturition (16.9%) symptoms. Nocturia, defined by the ICS as waking one or more times to urinate during the night, was the most commonly reported symptom (48.6% of men); terminal dribble (14.2% of men) and sensation of incomplete emptying (13.5% of men) were the most commonly reported voiding and post micturition symptoms, respectively [5]. A large-scale multinational study revealed that 90%of men aged 50 to 80 suffer from potentially troublesome LUTS [3]. Questionnaire data from 1,271 men with LUTS indicated that many men have storage and voiding symptoms [6]. The same study demonstrated that voiding symptoms were the most common male LUTS, but that storage symptoms made up four of the five most bothersome LUTS. Although LUTS are also highly prevalent in women, their frequent comorbidity with prostatic disease in men adds complexity to the management of male LUTS [6]. Benign prostatic hyperplasia (BPH) is a pathologic process that contributes to, but is not the sole cause of, lower urinary tract symptoms (LUTS) in aging men. Despite intense research efforts in the past five decades to elucidate the underlying etiology of prostatic growth in older men, cause-and effect relationships have not been established. For example, androgens are a necessary but not a clearly causative aspect of BPH. Previously held notions that the clinical symptoms of BPH are due simply to a mass-related increase in urethral resistance are too simplistic. It is now clear that a significant portion of LUTS is due to age –related detrusor dysfunction. Bladder outlet obstruction itself may induce a variety of neural alteration in the bladder, which contributes to symptomatology. Moreover bothersome LUTS may be seen on men with polyuria, sleep disorders, and a variety of systemic medical conditions unrelated to the prostate- bladder unit. BPH is but one cause of the LUTS in aging men commonly, and probably incorrectly, referred to as prostatism. BPH is a classical age-related disease and present in 20% of men at the age of 40 years, with progression to 70% at the age of 60 years [7]. The clinical relevance of this disorder is underscored by the fact that up to 50% of elderly men develop lower urinary tract symptoms (LUTS) due to BPH/BPE, and that transurethral resection of the prostate remains one of the most frequent interventions in elderly men, with a lifetime risk for surgery of around 25–30% [7]. Histopathologically, BPH is characterized by an increased number of epithelial and stromal cells around the urethra with an excessive nodular growth localized to the points where ejaculatory ducts enter into the transitional or periurethral zones of the prostate. At the cellular level, alterations including basal cell hyperplasia (enlarged, hypertrophic basal cells), increased stromal mass (particularly the amount of smooth muscle cells), enhanced extracellular matrix deposition, reduced elastic tissue, more infiltrating lymphocytes around ducts, acinar hypertrophy and more luminal corpora amylacea and calcifications in the form of prostatic calculi [8]. Periurethral nodules in BPH compress the urethra and may cause urodynamic obstruction. Such an obstruction can lead to LUTS as well as secondary changes that may ultimately require surgical intervention, such as bladder hypertrophy, urinary tract infection development of post-void residual volume, upper urinary tract changes and urinary retention. The observed increase in cell number may be due to epithelial and stromal proliferation or to impaired programmed cell death leading to cellular accumulation. Androgens, estrogens, stroaml, epithelial interactions, growth factors, and neurotransmitters may play a role, either singly or in combination in the etiology of the hyperplastic process. The prostate receives innervations from the sympathetic and the parasympathetic nerve system. The sympathetic system (noradrenergic nerves) is responsible for expelling prostatic fluid into the urethra during ejaculation [9], and the parasympathetic system (cholinergic nerves) increases the rate of secretion [10]. Moreover, the neuronal system has been shown to regulate prostatic function and growth [11]. Neuronal systems with effects on the prostate include the alpha adrenergic [12], the beta adrenergic [13] the cholinergic [14], the enkephalinergic [15], the peptidergic [16] and the nitrinergic system [13]..Sympathetic signaling pathways are important in the pathophysiology of LUTS, as reviewed subsequently. In addition, there is increasing evidence that sympathetic pathways may be important in the pathogenesis of the hyperplastic growth process. Alpha blockade, in some model systems can induce apoptosis [17, 18] .α –adrenergic pathways can also modulate the smooth muscle cell phenotype in the prostate[19]. All the components of the rennin-angiotensin system are present in prostatic tissue and may be active in BPH [20]. The alpha-1-adrenoreceptor is the prime determinant for urethral resistance causing outflow obstruction and LUTS [21]. Based on this observation, an important cornerstone of medical management of LUTS due to BPH/BPE is based on alpha-1-adrenergic-receptor (adrenoreceptor) blockade to reduce urethral resistance. This study seeks to evaluate the effect of a new natural compound “tradamixina “ made of Alga Ecklonia Bicyclis, Tribulus Terrestris and D-Glucosamine plus Serenoa Repens in order to improve lower urinary tract symptoms. Seven phlorotannins were isolated and characterized from an edible marine brown alga Ecklonia Bicyclis, along with three common sterol derivatives (fucosterol, ergosterol, and cholesterol) according to the comprehensive spectral analysis of MS and NMR data. Compounds 7-phloro eckoland ,6,6′-bieckoll in phlorotannin derivatives were obtained for the first time with their respective high yields. Any bioactive report of compound Fucodiphloroethol G was found nowhere up to date [22]. Ecklonia bicyclis has radical scavenger activity 10-100 times more powerful than any other polifenol terrestris plants, which have only 3-4 fenolic and rings that are commonly considered among the most effective antioxidant molecules. Common polyphenols are soluble in water also and have a relatively short half-life introduced into the body. All phlorotannins had antioxidant properties in vitro, especially, compounds 6,6′-bieckol which showed significant activities compared to the other phlorotannins [23]. In the Ecklonia bicyclis there are molecules that are able to reduce the inflammatory response, partially neutralizing the inflammatory damage caused by ROS and in part by slowing the gaming lipoxygenase and inhibiting the formation of prostaglandin E2, a powerful inflammatory mediator. Jung Wk [24] revealed that Ecklonia bicyclis inhibits LPS-induced NO and PGE2 production in a concentration-dependent manner and inhibits inducible iNOS and COX-2 in BV2 microglia without significant cytotoxicity. Ecklonia bicyclis treatment significantly reduced NF-kB translocation and DNA-binding in LPS-stimulated BV2 microglia. This effect was mediated through the inhibition of the degradation of the inhibitor kappaB and by inhibition of the mitogen-activated protein kinase (MAPK) phosphorylation, at least in part by inhibiting the generation of reactive oxygen species. This data also indicate that Ecklonia bicyclis extracts exert anti-inflammatory effects by suppressing proinflammatory cytokines. Collectively, these results suggest that Ecklonia bicyclis suppresses the induction of cytokines by LPS, as well as iNOS and COX-2 expression, by blocking NF-kappaB and MAPK activation. These findings provide mechanistic insights into the anti-inflammatory and neuroprotective actions of Ecklonia bicyclis in BV2 microglia [24]. Won -Kyo Jung[25] revealed that also only dieckol suppresses LPS-induced production of nitric oxide (NO) and prostaglandin E2 (PGE2) and expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in a dose-dependent manner, without causing cytotoxicity. It also significantly reduced the generation of proinflammatory cytokines, such as interleukin (IL)-1β and tumor necrosis factor (TNF)-α. Moreover, dieckol significantly reduced LPS-induced nuclear factor κB (NF-κB) and p38 mitogen-activated protein kinases (MAPKs) activation, as well as reactive oxygen species (ROS) production. Taken together, the inhibition of LPS-induced NO and PGE2 production might be due to the suppression of NF-κB and p38 MAPK signal pathway and, at least in part, by inhibiting the generation of ROS. (25) S.-K. Kim revealed extracts reduced the concentrations of IL-4 and IL-5 by 66% and 84%, respectively, and resulted in a 73% reduction in the secretion of TNF-α . Ecklonia bicyclis extracts enhanced IFN-γ production, but not to a significant degree [26]. The protodioscin is a steroidal saponin, which is about 45% of the extract obtained from aerial parts of Tribulus Terrestris. Its particular steroidal structure has an androgen-mimetic action, binding and activating the receptor of testosterone. So this substance is able to increase the endogenous production of testosterone, dihydrotestosterone, a hormone luteinizing hormone (LH), dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEA-S). Because of these effects in experimental animals there is an increase in spermatogenesis and the frequency of matches. In the rabbit in particular it has been shown that the compound stimulates the release of nitric oxide (NO) by vascular endothelium of the corpora cavernosa thereby having a pro-erectile effect. The mechanism behind this effect appears to involve the pathway of steroid hormones. Although in humans protodioscin is used for the treatment of erectile dysfunction [27, 28]. In a placebo-controlled study on a group of young volunteers serum levels of testosterone, androstenedione and luteinizing hormone were detected after administration of Tribulus Terrestris at doses of 10 and 20 mg / kg. After 4 weeks of treatment, these values were similar to those of untreated [29]. Biovis contains polymers of d-glucosamine and n-acetyl-d-glucosamine that act on both the non-adrenergic and non-colinergic system (NANC) and on the endothelial cell system as a strong nitric oxide synthetase (NOS) stimulator [30]. Serenoa Repens has been approved in France and Germany for treatment of BPH [31]. The mechanism of action of Serenoa Repens has been investigated in several in vitro or indirect in vivo studies and has not been definitively defined. The mechanism may include alteration of cholesterol metabolism, anti-oestrogenic, anti-androgenic (including 5a-reductase inhibitor activity), anti-inflammatory effects, and a decrease in available sex hormone binding globuline [32, 34].

Methods

An observational study was conducted from May 2011 to May 2012, at our Department of Urology. We enrolled 100 patients. Eligible men were ≥45 yrs. of age who had had LUTS/BPH for >6 mo at screening and with IPSS (The international Prostate symptom scores) ≥13 and maximum urinary flow rate (Qmax) ≥4 to ≤15 ml/s. Exclusion criteria were: drug assumption :α-blocker , Serenoa Repens , dutasteride, finasteride; hyper- and hypothyroidism, neurogenic syndrome (multiple sclerosis, multiple atrophy, Parkinson’s disease, tumors, stroke, disk disease, spinal cord disorders, polyneuropathy, uraemia), lower pelvic surgery (oncological pelvic surgery, lower urinary and genital tract surgery), prostate cancer. The compound “Tradamixina plus Serenoa Repens” (80 mg of Alga Ecklonia Bicyclis, 100 mg of Tribulus Terrestris and 100 mg of D-Glucosamine and N-Acetyl-D-Glucosamine plus 320 mg of Serenoa Repens) was administered daily for two months. At visit patients were evaluated by means of detailed medical urological history, clinical examination, laboratory investigations (total PSA), and instrumental examination like uroflowmetry. Efficacy measures included IPSS-International Prostate Symptom Scores (primary measure), BPH Impact Index (BII), and week 1 IPSS (mIPSS) questions beginning with ‘‘Since your last visit.’’ IPSS storage and voiding subscores, nocturia question (question 7), and IPSS Quality-of-Life (QoL) Index were also assessed. Measures were assessed at baseline and end point (12 wk or end of therapy) and also at screening, 1 and 4 wk for IPSS, and 4 wk for BII. The Patient and Clinician Global Impression of Improvement (PGI-I and CGI-I, respectively) instruments and the subject-rated Treatment Satisfaction Scale–BPH (TSS-BPH) [22], evaluated from 0% (greater) to 100% (lower) satisfaction, were administered at end point. Uroflowmetry was performed using standard calibrated devices at the screening, baseline, and end point visits. Valid Qmax measurements required prevoid total bladder volume (assessed by ultrasound) of ≥150 to ≤550 ml and voided volume (Vvoid) of ≥125 ml. Bladder capacity was calculated post hoc as the sum of Vvoid and postvoid residual (PVR) volume. Safety was evaluated based on subject-reported adverse events (AE), PVR, clinical laboratory parameters (total PSA). The primary efficacy objective was evaluating the change in total IPSS from baseline to end point. Efficacy was analysed in all subjects. At last continuous efficacy measures, uroflowmetry, and PVR were evaluated as a change from baseline to week 12/last-observation carried-forward end point. All other efficacy analyses (including all tradamixina results) were assessed at the 0.05 significance level without adjustment for multiplicity. A fixed-sequence testing procedure was implemented to control type I error in analyses of primary and key secondary outcomes for “Tradamixina plus Serenoa Repens” using the following pre-specified order: total IPSS at end point, total IPSS after 4 wk, BII at end point, mIPSS after 1 wk, and BII after 4 wk. Statistical significance was interpreted only if results of the preceding analysis were significant at the 0.05 level. Results were presented independent of the fixed sequence, as all end points tested under this procedure achieved statistical significance. All other efficacy analyses were assessed at the 0.05 significance level without adjustment for multiplicity. PGI-I- Patient Global Impression of Improvement- was analysed using the Cochran –Mantel-Haenszel test adjusted for baseline LUTS severity .Changes from baseline to end of therapy in Qmax, PVR, and clinical laboratory parameters were analysed using a ranked analysis of variance (ANOVA) with a term for treatment group. Treatment group differences for average urinary flow rate (Qave), Vvoid, and bladder capacity were performed as post hoc analyses.

Results and discussion

Mean age was 64 yrs. of age (10.2% ±75 yr of age). At randomisation, IPSS was ≥20 in 30% of subjects, and Qmax was <10 ml/s in 54%. The change from baseline to week 12 relative to “Tradamixina plus seronea Repens” in total IPSS was statistically significant p = 0.001 (Table 1). Least squares mean (LSmean) plus or minus standard error (SE) differences in IPSS were significant for “Tradamixina plus Serenoa Repens” at 1 wk (mIPSS: -1.5 ± 0.5; p = 0.003 ) and 4 wk (-2.2 ± 0.6; p < 0.001 ). Changes in IPSS subscores and nocturia are shown in table 1. Differences from baseline in BII were statistically significant for “Tradamixina plus Seronea Repens” ( p = 0.003), above all differences in BII were also significant at 4 wk (LSmean ± SE: -0.8 ± 0.2;p < 0.001 ). For the IPSS QoL Index, significant improvements at 12 wk were reported ( p = 0.022).(Table 2). The TSSBPH overall satisfaction score at end point was significantly low (indicating higher satisfaction) ( p = 0.005).In the distribution of subjects over the PGI-I and CGI-I response categories were significant for “Tradamixina plus Seronea Repens” (PGI-I: p = 0.001; CGI-I: p = 0.004). More subjects and their clinicians perceived improvements in LUTS at end point . Improvements in Qmax were significantly great (p = 0.009) (Table 3). For PVR, mean reductions from were observed with “Tradamixina plus Serenoa Repens”, but were not statistically significant. We also observed a decrease of total PSA (Table 4). “Tradamixina plus Serenoa Repens” improved total IPSS score and PGI-I and CGI-I scores because the Ecklonia bicyclis with its anti-inflammatory action and antioxidant effects, suppress LPS-induced production of nitric oxide (NO) and prostaglandin E2 (PGE2) and expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in a dose-dependent manner, without causing cytotoxicity. It also significantly reduced the generation of proinflammatory cytokines, such as interleukin (IL)-1β and tumor necrosis factor (TNF)-α. Moreover, dieckol significantly reduced LPS-induced nuclear factor κB (NF-κB) and p38 mitogen-activated protein kinases (MAPKs) activation, as well as reactive oxygen species (ROS) production [25]. It improved the bother symptoms. BPH is associated with changes of innervations, and biological mediator production and release in the central zone of the prostate [35]. In particular, there is a decrease of nerves of the enkephalinergic [36] and nitrinergic systems [37, 38] both mediating smooth muscle cell relaxation [37]. Compared to normal tissue, nNOS expression has been observed to decrease in the transitional zone of BPH tissue. On the other hand, inducible NOS (iNOS), produced after stimulation by immune and inflammatory cytokines and free radicals, has been reported to increase selectively in the stroma of patients suffering from BPH [36]. This observation is probably due to increased proinflammatory conditions in BPH. The importance of the NOS system for the prostate stroma is further supported by experiments with constitutive active NOS dependent guanylylcyclases. cGMP analogues have been shown to activate the Proteinkinase G II and to inhibit proliferation of human smooth muscle cells in vitro [37]. Two enzymes, the neuronal (NOS-I) and the endothelial (NOS-III) isoforms, are Ca2_-dependent and constitutively expressed.The third enzyme is an inducible Ca2_-independent isoform (NOS-II), expressed after stimulation with Escherichia coli lipopolysaccharide (LPS) and/or different cytokines, such as interferon-γ (IFNγ), interleukin-1, or tumor necrosis factor- NOS-II induction occurs at the transcriptional level and is mediated by the early activation of some nuclear transcriptional factors, including NFkB [38]. A large body of experimental evidence suggests that physiological levels of NO, similar to those produced by the basal activity of NOS-I or NOS-III, prevent induction of NOS-II mRNA expression through the suppression of NFkB activation [39, 40]. As a consequence, NOS-II gene expression takes place after LPS/cytokine stimulation, provided that the NOS-I- or NOS-III-generated NO is reduced below a threshold value in a short time [41]. A recent report [42, 43] shows that NOS-II inducers (e.g. LPS and IFNγ) consistently elicit a rapid inactivation of NOS-I by tyrosine phosphorylation, an event leading to a decrease of basal NO levels. A recent study [44] reports that inhibition of NOS-I can be achieved via activation of cytosolic phospholipase A2 (cPLA2), a large molecular mass member of the family of PLA2 enzymes. The activities of NOS-I and cPLA2 are both regulated by increases in the intracellular concentration of free Ca2 ([Ca2]i). Not surprisingly, enhancing the [Ca2]i caused a parallel increase in both activities and accumulation of respective products, NO and arachidonic acid (AA). Inducible nitric oxide synthase (iNOS) is expressed in a variety of cell types, in particular in inflammatory cells, in response to diverse pro-inflammatory stimuli. This process requires critical levels of arachidonic acid (AA), generated by constitutive phospholipase A2 (PLA2), promoting tyrosine kinase-dependent phosphorylation, and inhibition, of constitutive NOS. Lowering basal NO levels is indeed critical for the activation of NF-κB, and thus for the expression of genes (e.g. iNOS) regulated by this transcription factor. It is interesting to note that NO and AA, two small lipid soluble molecules, rapidly cross the plasma membrane thereby allowing the triggering of the above responses in distal cells. That is, constitutive NO might inhibit NF-κB activity in the same cells (e.g. astrocytes) in which it is generated, as well as in other cells that do not express constitutive NOS (e.g. microglia). NO from cells unable to respond to pro-inflammatory stimuli (e.g. neurons) will also contribute to these effects. Along the same line, AA released by pro-inflammatory molecules in specific cell types (e.g. astrocytes) might suppress constitutive NOS activity in the same cells as well as in other cells (e.g. neurons). Thus, AA produced at the very early stages of the inflammatory response is a likely critical signal switching the regulation of the “NO tone” from physiological (i.e. mediated by constitutive NOS) to pathological (i.e. mediated by iNOS). This second phase of the inflammatory response is often accompanied by the onset of deleterious effects in the tissue in which a critical role is played by iNOS-derived NO (directly or indirectly, i.e. via formation of peroxynitrite) as well as by products of the AA cascade. In summary, the relative amounts of NO and AA, released by their constitutive enzymes, produce autocrine and paracrine effects regulating the onset of an inflammatory response in which, in addition to other factors, NO and AA are extensively released by their inducible enzymes [45]. Biovis contains polymers of d-glucosamine and n-acetyl-d-glucosamine that act both on the non-adrenergic and non-colinergic system (NANC) and on the endothelial cell system as a strong nitric oxide synthetase (NOS) stimulator [46]. This explains why there is an improvement of Qave and Qmax. In fact while α-blocker drugs facilitated the opening of the bladder neck thanks to the presence of alpha receptors, Tradamixina improved the NOS action on its receptor, so for the reasons above also improved LUTS. Serenoa Repens also has anti-inflammatory effects. As described above, α1-adrenoceptor antagonists are commonly used in the treatment of men with voiding symptoms (urinary obstruction, pollakiuria and urinary incontinence) secondary to BPH. Goepel et. Al. have shown that Serenoa Repens might have α1-adrenoceptor inhibitory properties. Serenoa Repens significantly affects pharmacological receptors, such as the α1-adrenoceptor and the muscarinic receptor in the lower urinary tract, to relieve the irritative and obstructive symptoms of dysuria due to BPH and LUTS [34]. In addition to traditionally used medications, like α1-adrenoceptor antagonists, antimuscarinics, 5α-reductase inhibitors, and phytotherapy, several new therapeutic agents, such as selective β3-adrenoceptor agonists, are potentially useful for treating LUTS suggestive of BPH, particularly for storage symptoms secondary to outflow obstruction [47]. Thus, the effects of SPE on these receptors in the lower urinary tract might be pharmacologically relevant. To date, more than 11 placebo-controlled trials and 4 active-controlled trials with SPE in men with BPH have been conducted. Patient numbers were usually limited and the evaluation periods were relatively short, so it would be difficult to evaluate the effect of SPE and ascertain the efficacy of SPE in BPH patients. However, some placebo-controlled studies and comparisons to α1-blockers have recently been conducted with relatively long-term treatments and sufficient numbers of patients [4850]. BPH causes dysuria and residual urine via a mechanical stoppage due to hypertrophy of prostatic tissue and via a functional stoppage caused by α1-adrenoceptor hypertonia of prostatic smooth muscle. Previous studies have demonstrated that Serenoa Repens had a number of pharmacological effects: 1) an antiandrogenic effect — inhibition of 5α-reductase I and II and inhibition of binding of dihydrotestosterone (DHT) to the cytosolic androgen receptors, 2) an anti-inflammatory effect, 3) an anti-proliferative effect, and 4) significant binding of pharmacological receptors existing in the lower urinary tract. In BPH there is a decreased ratio between androgen and estrogen, and tribulus terrestris by increasing total testosterone serum level restores the ratio.

Table 1 International prostate symptom score (IPSS) total,storage and voiding subscores and nocturia question (IPSS question 7).
Table 2 International Prostate Symptom Score Quality of Life Index, Treatment Satisfaction scale, and Patient or Clinician Global Impression of Improvement Scores at 12 wks. compared to baseline
Table 3 Uroflowmetry and postvoid residual volume
Table 4

Conclusion

The daily treatment with a new compound “Tradamixina” (100 mg of Alga Ecklonia Bicyclis, 80 mg of Tribulus Terrestris and 80 mg of D-Glucosamine and N-Acetyl-D-Glucosamine) plus 320 mg Serenoa Repens for two months improved the male sexual function. It improved the bother symptoms which affect the patient’s quality of life, improved uroflowmetric parameters, and we observed a decrease of serum PSA level. These effects are due to its antioxidant, anti-aging action, and the bother symptoms due to its anti-inflammatory action. Infact it neutralizes the action of ROS, LPS, COX2, NFkβ, probably also reducing the concentrations of TNF-α, MMP-1. The decrease of PSA is due to the anti-inflammatory action. This result can be the basis for future research.

Abbreviations

AA:

Arachidonic acid

AE:

Adverse events

BII:

BPH Impact Index

BPH:

Benign prostatic hyperplasia

CGI-I:

The Clinician Global Impression of Improvement

COX-2:

Cyclooxygenase-2

cPLA2:

Cytosolic phospholipase A2

DHEA:

Dehydroepiandrosterone

DHEA-S:

Dehydroepiandrosterone sulfate

IL-1β:

Interleukin

iNOS:

Inducible nitric oxide synthase

IPSS:

International Prostate Symptom

IPSS:

International Prostate Symptom

LH:

Hormone luteinizing hormone

LUTS:

Lower urinary tract symptoms

MAPKs:

Mitogen-activated protein kinases

MMP-1:

Matrix metalloproteinase-1

NANC:

Non-adrenergic and non-colinergic system

NF-κB:

Nuclear factor κB

NO:

Nitric oxide

NOS:

Nitric oxide synthetase

NOS-I:

Neuronal Nitric oxide synthetase isoforms I

NOS-III:

Endothelial Nitric oxide synthetase isoforms III

PGE2 :

Prostaglandin E2

PGI-I:

Patient Global Impression of Improvement

PGI-I:

The Patient and Clinician Global Impression of Improvement

PVR:

Postvoid residual

Qave:

Urinary flow rate

QoL:

Quality-of-Life

ROS:

Reactive oxygen species

TNF-α:

Tumor necrosis factor

TSS-BPH:

Treatment Satisfaction Scale–BPH

Vvoid:

Voided volume

References

  1. Engstrom G, Henningsohn L, Steineck G, Leppert J: Selfassessed health, sadness and happiness in relation to the total burden of symptoms from the lower urinary tract. BJU Int. 2005, 95: 810-5. 10.1111/j.1464-410X.2005.05406.x.

    Article  PubMed  Google Scholar 

  2. Hu TW, Wagner TH, Bentkover JD, et al: Estimated economic costs of overactive bladder in the United States. Urology. 2003, 61: 1123-8. 10.1016/S0090-4295(03)00009-8.

    Article  PubMed  Google Scholar 

  3. Rosen R, Altwein J, Boyle P, et al: Lower urinary tract symptoms and male sexual dysfunction: the Multinational Survey of the Aging Male (MSAM-7). Eur Urol. 2003, 44: 637-49. 10.1016/j.eururo.2003.08.015.

    Article  PubMed  Google Scholar 

  4. Irwin DE, Milsom I, Hunskaar S, et al: Population-based survey of urinary incontinence, overactive bladder, and other lower urinary tract symptoms in five countries: results of the EPIC study. Eur Urol. 2006, 50: 1306-15. 10.1016/j.eururo.2006.09.019.

    Article  PubMed  Google Scholar 

  5. Abrams P, Cardozo L, Fall M, et al: The standardisation of terminology in lower urinary tract function: report from the standardisation sub-committee of the International Continence Society. Urology. 2003, 61: 37-49. 10.1016/S0090-4295(02)02243-4.

    Article  PubMed  Google Scholar 

  6. Peters TJ, Donovan JL, Kay HE, et al: The International Continence Society ‘‘Benign Prostatic Hyperplasia’’ Study:the bothersomeness of urinary symptoms. J Urol. 1997, 157: 885-9. 10.1016/S0022-5347(01)65075-4.

    Article  CAS  PubMed  Google Scholar 

  7. Isaacs JT: Etiology Of Benign Prostatic Hyperplasia. Eur Urol. 1994, 25 (Suppl.1): 6-9.

    PubMed  Google Scholar 

  8. Bostwick DG, Cooner WH, Denis L, Jones GW, Scardino PT, Murphy GP: The Association Of Benign Prostatic Hyperplasia And Cancer Of The Prostate. Cancer. 1992, 70: 291-301. 10.1002/1097-0142(19920701)70:1+<291::AID-CNCR2820701317>3.0.CO;2-4.

    Article  CAS  PubMed  Google Scholar 

  9. Bruschini H, Schmidt RA, Tanagho EA: Neurologic Control Of Prostatic Secretion In The Dog. Invest. Urol. 1978, 15: 288-290.

    CAS  PubMed  Google Scholar 

  10. Smith ER, Lebeaux MI: The Mediation Of The Canine Prostatic Secretion Provoked By Hypogastric Nerve Stimulation. Invest. Urol. 1970, 7: 313-318.

    CAS  PubMed  Google Scholar 

  11. Burnett AL, Maguire MP, Chamness SL, Ricker DD, Takeda M, Lepor H, Chang TS: Characterization And Localization Of Nitric Oxide Synthase In The Human Prostate. Urology. 1995, 45: 435-439. 10.1016/S0090-4295(99)80012-0.

    Article  CAS  PubMed  Google Scholar 

  12. Higgins JR, Gosling JA: Studies On The Structure And Intrinsic Innervation Of The Normal Human Prostate. Prostate Suppl. 1989, 2: 5-16.

    Article  CAS  PubMed  Google Scholar 

  13. Tsujii T, Azuma H, Yamaguchi T, Oshima H: A Possible Role Of Decreased Relaxation Mediated By Beta-Adrenoceptors In Bladder Outlet Obstruction By Benign Prostatic Hyperplasia. Br. J. Pharmacol. 1992, 107: 803-807. 10.1111/j.1476-5381.1992.tb14527.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Caine M, Raz S, Zeigler M: Adrenergic And Cholinergic Receptors In The Human Prostate, Prostatic Capsule And Bladder Neck. Br. J. Urol. 1975, 47: 193-202. 10.1111/j.1464-410X.1975.tb03947.x.

    Article  CAS  PubMed  Google Scholar 

  15. Jungblut T, Aumuller G, Malek B, Melchior H: Age dependency And Regional Distribution Of Enkephalinergic Nerves In Human Prostate. Urol. Int. 1989, 44: 352-356. 10.1159/000281539.

    Article  CAS  PubMed  Google Scholar 

  16. Lange W, Unger J: Peptidergic Innervation Within The Prostate Gland And Seminal Vesicle. Urol. Res. 1990, 18: 337-340. 10.1007/BF00300783.

    Article  CAS  PubMed  Google Scholar 

  17. Anglin Ie, Glassman Dt, Kyprianou N, et al: Induction Of Prostate Apoptosis By Alpha1- Adrenoceptor Antagonists: Mechanistic Significance Of The Quinazoline Component. Prostate Cancer Prostatic Dis. 2002, 5: 88-95. 10.1038/sj.pcan.4500561.

    Article  CAS  PubMed  Google Scholar 

  18. Partin Jv, Anglin Ie, Kyprianou N: Quinazoline-Based Alpha 1-Adrenoceptor Antagonists Induce Prostate Cancer Cell Apoptosis Via Tgf-Beta Signalling And I Kappa B Alpha Induction. Br J Cancer. 2003, 88: 1615-1621. 10.1038/sj.bjc.6600961.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lin Vk, Benaim Ea, Mcconnell Jd: Alpha-Blockade Downregulates Myosin Heavy Chain Gene Expression In Human Benign Prostatic Hyperplasia. Urology. 2001, 57: 170-175. 10.1016/S0090-4295(00)00842-6.

    Article  CAS  PubMed  Google Scholar 

  20. Dinh Dt, Frauman Ag, Sourial M, et al: Identification, Distribution, And Expression Of Angiotensin Ii Receptors In The Normal Human Prostate And Benign Prostatic Hyperplasia. Endocrinology. 2001, 142: 1349-1356. 10.1210/en.142.3.1349.

    CAS  PubMed  Google Scholar 

  21. Lepor H, Shapiro E: Characterization Of Alpha1 Adrenergic Receptors In Human Benign Prostatic Hyperplasia. J. Urol. 1984, 132: 1226-1229.

    CAS  PubMed  Google Scholar 

  22. Yong Li, Zhong-Ji Qian, Bomi Ryu, Sang-Hoon Lee, Moon-Moo Kim, Se-Kwon Kim: Chemical Components And Its Antioxidant Properties In Vitro: An Edible Marine Brown Alga, Ecklonia Cava. Bioorganic & Medicinal Chemistry. 2009, 17 (5): 1963-1973. 10.1016/j.bmc.2009.01.031.

    Article  Google Scholar 

  23. Li Y, Qian Zj, Ryu B, Lee Sh, Kim Mm, Kim Sk: Chemical Components And Its Antioxidant Properties In Vitro: An Edible Marine Brown Alga, Ecklonia Cava. Bioorg Med Chem. 2009, 17 (5): 1963-73. 10.1016/j.bmc.2009.01.031. Epub 2009 Jan 21

    Article  CAS  PubMed  Google Scholar 

  24. Jung Wk, Ahn Yw, Lee Sh, Choi Yh, Kim Sk, Yea Ss, Choi I, Park Sg, Seo Sk, Lee Sw, Choi Iw: Ecklonia Cava Ethanolic Extracts Inhibit Lipopolysaccharide-Induced Cyclooxygenase-2 And Inducible Nitric Oxide Synthase Expression In Bv2 Microglia Via The Map Kinase And Nf-Kappab Pathways. Food Chem Toxicol. 2009, 47 (2): 410-7. 10.1016/j.fct.2008.11.041. Epub 2008 Dec 11

    Article  CAS  PubMed  Google Scholar 

  25. Won-Kyo Jung, Soo-Jin Heo§, You-Jin Jeon, Chang-Min Lee, Yeong-Min Park, Hee-Guk Byun, Yung Hyun Choi, Sae-Gwang Parkδ, Il-Whan Choi: Inhibitory Effects And Molecular Mechanism Of Dieckol Isolated From Marine Brown Alga On Cox-2 And Inos In Microglial Cells. J. Agric. Food Chem. 2009, 57 (10): 4439-4446. 10.1021/jf9003913.

    Article  Google Scholar 

  26. Kim S-K, et al: / Biomedicine & Pharmacotherapy. 2008, 62: 289e296-

    Google Scholar 

  27. Gauthman K, Adaikan Pg, Prasad Rn: Aphrodisiac Properties Of Tribulus Terrestris Extract (Protodioscin) In Normal And Castrated Rats. Life Sci. 2002, 71: 1385-1396. 10.1016/S0024-3205(02)01858-1.

    Article  Google Scholar 

  28. Adaikan Pg, Gauthaman K, Prasad Rnv, Ng Sc: Proerectile Pharmacological Effects Of Tribulus Terrestris Extract On The Rabbit Corpus Avernosum. Ann Acad Med Singapore. 2000, 29: 22-26.

    CAS  PubMed  Google Scholar 

  29. Neychev Vk, Mitev Vi: The Aphrodisiac Herb Tribulus Terrestris Does Not Influence The Androgen Production In Young Men. Jethnopharmacol. 2005, 101: 319-323. 10.1016/j.jep.2005.05.017.

    Article  CAS  Google Scholar 

  30. Chung H, Park Sl, Kim Sc, Sung Cw, Lee Jw: Effect Of Kp 2647tm In Patients With Erectile Dysfunction. Chung Euro & Derma Clinic, Seoul, Korea - Central Research Institute Of Kunpoong Bio, Korea

  31. Dreikorn K, Richter R, Ackerman R, Schroeder FH: Conservative nonhormonal treatment of patients with benign prostatic hyperplasia. New Developments in Biosciences 5, Prostatic Hyperplasia. 1989, Berlin: Walter de Gruyter & Co, 109±31

    Google Scholar 

  32. Marwick C: Growing use of medicinal botanicals forces assessment by drug regulators. JAMA. 1995, 273: 607±9

    Google Scholar 

  33. McGuire E: Detrusor response to obstruction. 1987, Department of Health and Human Services, Rockville, MD, 221-

    Google Scholar 

  34. Di Silverio F, D'Eramo G, Lubrano C, Flammia GP, Sciarra A, Palma E, et al: Evidence that Serenoa repens extract displays an antiestrogenic activity in prostatic tissue of benign prostatichypertrophy patients. Eur. Urol. 1992, 2: 309±14

    Google Scholar 

  35. Chapple CR, Crowe R, Gilpin SA, Gosling J, Burnstock G: The Innervation Of The Human Prostate Gland—The Changes Associated With Benign Enlargement. J. Urol. 1991, 146: 1637-1644.

    CAS  PubMed  Google Scholar 

  36. Caine M, Raz S, Zeigler M: Adrenergic And Cholinergic Receptors In The Human Prostate, Prostatic Capsule And Bladder Neck. Br. J. Urol. 1975, 47: 193-202. 10.1111/j.1464-410X.1975.tb03947.x.

    Article  CAS  PubMed  Google Scholar 

  37. Jungblut T, Aumuller G, Malek B, Melchior H: Age dependency And Regional Distribution Of Enkephalinergic Nerves In Human Prostate. Urol. Int. 1989, 44: 352-356. 10.1159/000281539.

    Article  CAS  PubMed  Google Scholar 

  38. Takeda M, Tang R, Shapiro E, Burnett AL, Lepor H: Effects Of Nitric Oxide On Human And Canine Prostates. Urology. 1995, 45: 440-446. 10.1016/S0090-4295(99)80013-2.

    Article  CAS  PubMed  Google Scholar 

  39. Gradini R, Realacci M, Ginepri A, Naso G, Santangelo C, Cela O, Sale P, Berardi A, Petrangeli E, Gallucci M, Di Silverio F, Russo MA: Nitric Oxide Synthases In Normal And Benign Hyperplastic Human Prostate:. Immunohistochemistry And Molecular Biology. J. Pathol. 1999, 189: 224-229.

    CAS  PubMed  Google Scholar 

  40. Colasanti M, Suzuki H: Trends Pharmacol. Sci. 2000, 21: 249-252. 10.1016/S0165-6147(00)01499-1.

    Article  CAS  PubMed  Google Scholar 

  41. Colasanti M, Cavalieri E, Persichini T, Mollace V, Mariotto S, Suzuki H, Lauro GM: J. Biol. Chem. 1997, 272: 7582-75857. 10.1074/jbc.272.12.7582.

    Article  CAS  PubMed  Google Scholar 

  42. Colasanti M, Persichini T, Cavalieri E, Fabrizi C, Mariotto S, Menegazzi M, Lauro GM, Suzuki H: J. Biol. Chem. 1999, 274: 9915-9917. 10.1074/jbc.274.15.9915.

    Article  CAS  PubMed  Google Scholar 

  43. Palomba L, Bianchi M, Persichini T, Magnani M, Colasanti M, Cantoni O: Free Radic. Biol. Med. 2004, 36: 319-329. 10.1016/j.freeradbiomed.2003.10.026.

    Article  CAS  PubMed  Google Scholar 

  44. Leslie CC: J. Biol. Chem. 1997, 272: 16709-16712. 10.1074/jbc.272.27.16709.

    Article  CAS  PubMed  Google Scholar 

  45. Mariotto S, Suzuki Y, Persichini T, Colasanti M, Suzuki H, Cantoni O: Cross-Talk Between No And Arachidonic Acid In Inflammation. Curr Med Chem. 2007, 14 (18): 1940-4.

    Article  CAS  PubMed  Google Scholar 

  46. Chung H, Park Sl, Kim Sc, Sung Cw, Lee Jw: Effect Of Kp 2647tm In Patients With Erectile Dysfunction. Chung Euro & Derma Clinic, Seoul, Korea - Central Research Institute Of Kunpoong Bio, Korea

  47. Andersson KE: LUTS treatment: future treatment options. Neurourol Urodyn. 2007, 26: 934-47. 10.1002/nau.20500.

    Article  CAS  PubMed  Google Scholar 

  48. Bent S, Kane C, Shinohara K, Neuhaus J, Hudes ES, Goldberg H, et al: Saw palmetto for benign prostatichyperplasia. N Engl J Med. 2006, 354: 557-66. 10.1056/NEJMoa053085.

    Article  CAS  PubMed  Google Scholar 

  49. Debruyne F, Koch G, Boyle P, Da Silva FC, Gillenwater JG, Hamdy FC, et al: 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. 10.1016/S0302-2838(02)00066-0. discussion 506-7

    Article  CAS  PubMed  Google Scholar 

  50. Willetts KE, Clements MS, Champion S, Ehsman S, Eden JA: Serenoa repens extract for benign prostate hyperplasia: a randomized controlled trial. BJU Int. 2003, 92: 267-70. 10.1046/j.1464-410X.2003.04316.x.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This article has been published as part of BMC Surgery Volume 12 Supplement 1, 2012: Selected articles from the XXV National Congress of the Italian Society of Geriatric Surgery. The full contents of the supplement are available online at http://www.biomedcentral.com/bmcsurg/supplements/12/S1.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ester Illiano.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

FI, DP: conception and design, interpretation of data, given final approval of the version to be published; EI, AR, GR: acquisition of data, drafting the manuscript, given final approval of the version to be published; BA: critical revision, interpretation of data, given final approval of the version to be published.

This article has been retracted by the authors because it contains large portions of text that were duplicated from a number of pervious publications including Chapple et al., 2006 , Untergasser et al., 2005 and Mariotto et al., 2007 . The authors apologise for failing to cite these articles.

An erratum to this article can be found online athttp://dx.doi.org/10.1186/s12893-016-0139-0.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

About this article

Cite this article

Iacono, F., Prezioso, D., Illiano, E. et al. RETRACTED ARTICLE: Observational study: daily treatment with a new compound “tradamixina” plus serenoa repens for two months improved the lower urinary tract symptoms. BMC Surg 12 (Suppl 1), S22 (2012). https://doi.org/10.1186/1471-2482-12-S1-S22

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/1471-2482-12-S1-S22

Keywords