Journal List > Korean J Urol > v.56(10) > 1006325

Andersson: Drug therapy of overactive bladder - What is coming next?

Abstract

After the approval and introduction of mirabegron, tadalafil, and botulinum toxin A for treatment of lower urinary tract symptoms/overactive bladder, focus of interest has been on their place in therapy versus the previous gold standard, antimuscarinics. However, since these agents also have limitations there has been increasing interest in what is coming next - what is in the pipeline? Despite progress in our knowledge of different factors involved in both peripheral and central modulation of lower urinary tract dysfunction, there are few innovations in the pipe-line. Most developments concern modifications of existing principles (antimuscarinics, β3-receptor agonists, botulinum toxin A). However, there are several new and old targets/drugs of potential interest for further development, such as the purinergic and cannabinoid systems and the different members of the transient receptor potential channel family. However, even if there seems to be good rationale for further development of these principles, further exploration of their involvement in lower urinary tract function/dysfunction is necessary.

INTRODUCTION

The places in therapy for the three new drug principles recently approved for treatment of lower urinary tract symptoms/overactive bladder (LUTS/OAB), the β3-adrenoceptor (AR) agonist, mirabegron, the phosphodiesterase 5-inhibitor tadalafil, and the blocker of afferent and efferent neutotransmission, botulinum toxin (BoNT), still have to be established [1234]. Even if these agents compared to animuscarinics may have advantages, they are not effective in all patients, and alternatives are continuously being explored. There has been increasing interest in both old and new therapeutic principles and in what is currently in the pipeline. Much nonclinical and clinical research is ongoing, both involving modifications of existing options and directed at identifying novel pharmacological principles involved in LUTS/OAB pathophysiology, and this has been extensively discussed in several excellent reviews [45678].
The aims of this review is to briefly comment on what is ongoing and then speculate on the future of some of the many targets and drugs theoretically attractive for development.

OAB DRUGS IN THE PIPELINE

1. Antimuscarinics

Several antimuscarinics with different profiles are in development and have been reviewed previously [6]. These include tarafenacin which is a novel potent antimuscarinic agent highly selective for M3 over M2 receptors [9]. In a mouse model, the drug was reported to have functional selectivity for bladder over atrial tissues in the order of 200 folds, which may be of interest from a cardiac safety point of view. In a multicenter, randomized controlled 2b trial (235 patients), Song et al. [10] showed that tarafenacin at doses of 0.2 and 0.4 mg was superior to placebo after 4 weeks in reducing the number of micturitions per day (primary endpoint) and showed an good safety profile. Interestingly, there were very few cases of constipation. However, the most common side effect was dry mouth, which at a dose of 0.4 mg occurred in 52 out of 76 randomized patients. Considering this, it is hard to believe that this drug, even if proven efficacious in future studies, will offer any advantages over existing options. OAB, defined either based on symptoms (OAB syndrome) or urodynamically (detrusor overactivity, DO), is a filling disorder, and even if it is well established that M3 receptors are involved in detrusor muscle contraction, it is not necessarily by inhibition this contraction that the beneficial effects of antimuscarinics are exerted [11].
To specifically reduce the adverse effect of tolterodine-induced dry mouth, THVD-201 (Tolenix, twice daily formulation) and THVD-202 (once daily formulation) were designed Both drugs are a combination of the muscarinic antagonist tolterodine with modified-release formulations of the muscarinic receptor agonist, pilocarpine, as a salivary stimulant. Tolenix is advancing into phase III studies and has demonstrated efficacy comparable to twice-daily tolterodine; however, the combination showed statistically significant and clinically meaningful improvements in saliva production and dry mouth, as compared to active control tolterodine [12]. It is possible, but has to be demonstrated in further trials, that this advantage over tolterodine alone will be sufficient to motivate marketing of the drug.
Another antimuscarinic claimed to have a different profile is afacifenacin (SMP-986), which combines the dual pharmacological actions of nonselective muscarinic receptor antagonism and inhibition of bladder afferent pathways through Na+ channel blockade [6]. Theoretically, Na+ channel blockade would produce a local anesthetic effect that might increase the risk for cardiac side effects [1314]. This would not be in favor of the drug. However, since no clinical studies have been published, no efficacy or saftety profile is available.

2. β3-Adrenoceptor agonists

The approval and clinical success of mirabegron has focused interest on β3-AR agonism and on how to modify and further improve this therapeutic principle. β3-AR agonists have generally been considered to relieve OAB symptoms by relaxing detrusor muscle, inhibiting spontaneous contractile activity in the detrusor (in vitro: microcontractions; in vivo: nonvoiding contractions), and reducing bladder afferent activity [1516171819]. In vitro, Biers et al. [15] demonstrated that the β3-AR agonist, solabegron, concentration-dependently inhibited microcontractions in strips of human detrusor muscle, and in vivo, several investigators have shown that β3-AR agonists can decrease nonvoiding contractions in the obstructed bladder [10]. Effects on afferent bladder activity was shown by e.g., Aizawa et al. [20] who demonstrated that single-unit afferent activities of both Aδ-fibers and C-fibers in response to bladder filling significantly and dose-dependently decreased after mirabegron administration, the effect being more conspicuous for Aδ-fibers. However, in a series of studies, Gillespie and colleagues [21222324] have questioned the accepted view on the mode and site of action of β3-AR agonists, and suggested that effects on neither spontaneous microcontractions, nor on nonvoiding contractions in e.g., obstructed rats, can fully explain the effects of mirabegron. Supporting the view that other mechanisms than effects on detrusor muscle may contribute, recent evidence showed that activation of prejunctional β3-AR may result in down-regulation of ACh released from cholinergic terminals thereby exerting an additional inhibitory control of parasympathetic activity [2526].
In addition to the only marketed β3-AR agonist, mirabegron, there are reports on other β3-AR agonists in development, e.g., ritobegron and solabegron [718]. Phases II and III randomised, double blind, placebo controlled studies of ritobegron in patients with OAB has been initiated and completed, but the results of this study have not been published and it seems that the primary efficacy endpoint of the studies was not met [7]. Since preclinical studies were promising, the findings that ritobegron was not clinically successful are somewhat surprising.
Efficacy and safety of solabegron (GW427353) have been reported in a phase II multicenter, randomized, proof-ofconcept trial in 258 women with wet OAB [27]. Solabegron was well tolerated and at the dose of 125 mg produced a statistically significant difference in percent change from baseline to week 8 in incontinence episodes over 24 hours (primary outcome) when compared with placebo (p=0.025) [27]. Further studies are awaited.
There have been many early investigations of other novel and putative β3-AR agonists for management of OAB, including CL-316243 [28], aryloxypropanolamine [29], conformationally restricted acetanilides [30], TRK-380 [31], AJ-9677 [32], and BRL37344 [33]. These agents have been reported as being in development, but no clinical data have been published. Thus, even if there are a number of β3-AR agonists in the pipeline, it is uncertain which, if any, will come to market and be available for the management of OAB.

3. BoNT A - recent developments

To improve intravesical treatment with BoNTs, novel therapeutic uses and formulations have been reported [34353637]. New formulations seek to improve bioavailability at the site of action while decreasing adverse events.
Liposomes are vesicles composed of concentric phospholipid bilayers separated by aqueous compartments. Because they adsorb onto cell surfaces and fuse with cells, they are being used as vehicles for drug delivery and gene therapy. In order to have the therapeutic effects of BoNT-A on the urothelial afferent nerves without impairing detrusor contractility, and to improve patients' acceptability of the treatment by overcoming the adverse effects of cystoscope-guided needle injections, studies are ongoing exploring if liquid liposomes may deliver BoNT-A (liposome encapsulated BoNT-A or lipotoxin) through the urothelium to the suburothelial space. In a rat model, intravesical lipotoxin cleaved SNAP-25, inhibited calcitonin gene-related peptide release from afferent nerve terminals, and blocked acetic acid-induced DO [38]. Kuo et al. [36] performed a study on 24 patients with OAB, who were nonresponsive to >3 months of therapy with traditional antimuscarinic agents. They were randomized 1:1 to receive intravesical instillation of lipotoxin or saline solution. In the lipotoxin group, 3-day urinary frequency and urgency episodes were significantly decreased at 1 month, whereas no change was reported in the control group. Importantly, no urinary tract infections or large postvoid residuals volumes were reported. However, only 50% of the 12 patients initially treated with lipotoxin showed a response, and only four had a maintained response at 3 months. Furthermore, of 12 nonresponders who were subsequently treated, or retreated with lipotoxin (six from each cohort), only one showed a response at 3 months. Moreover, no change in urgency incontinence was found in either group, although the median baseline frequency was only 0.5 events in the lipotoxin cohort.
Intravesical lipotoxin is an interesting and promising principle, but it may have some limitations. The obvious question is whether BoNT will be transported into the bladder wall far enough to affect not only the afferent nerves in the suburothelium, but also the afferent and efferent nerves in the detrusor muscle. Intravesical liposome carried BoNT may have fewer adverse effects, but is this obtainable at the price of reduced efficacy (compared to bladder wall injection)?
Combination of genetic engineering and molecular biology techniques have enabled the possibility of developing recombinant biotherapeutic proteins incorporating the light chain (endopeptidase) and the HN translocation domain of BoNT, combined with a binding domain that binds to a specific target represented by a cell surface receptor [3437]. A novel targeted BoNT-A (AGN-214868; senrebotase) has already completed phase I studies and entered proof-of-concept phase II studies in postherpetic neuralgia and idiopathic OAB [39]. This is an exciting and promising principle, but the results of clinical studies have to be awaited. Thus, despite encouraging preclinical results, significant technology refinement and clinical testing will be required in order to define the safety and efficacy profile of new BoNT formulations and engineered variants.

4. Combinations

Treatment of disorders with multifactorial pathophysiology with combinations of drugs seems to be a logical approach-not only can more than one underlying mechanism be influenced (if the drugs have different mechanisms of action), but also the doses of drugs can be kept low making it possible to reduce the number of side effects. LUTS/OAB in both men and women are multifactorial, and there are many examples that combined treatment can be superior to monotherapy. However, which combination to which patient? How much can be gained? Is there really a cost/benefit in combining currently approved drugs with respect to efficacy and side effects, or is the field open for introduction of "minor players" i.e., drugs with some efficacy, but not efficacious enough to be given as monotherapy? There are many reviews of combinations used to treat male LUTS, with and without OAB as a dominating symptom [404142]. Combination of mirabegron and solifenacin has shown promising results in a phase 2 study of OAB parients [43], and further phase 3 studies are ongoing, possibly resulting in a fixed combination for clinical use within a reasonable time frame.
Even if drug+drug combinations are promising, combination of drugs with nonpharmacological neuromodulation, for example percutaneous tibial nerve stimulation, may be an interesting alternative, and the many possibilities of combining therapies opens the door for personalized therapy of LUTS/OAB [44].

PROMISING FUTURE TARGETS?

There are many agents with theoretically interesting profiles that have been or still are considered as promising, but currently do not seem to be in active development or where development is slow (Table 1). For example, nerve growth factor (NGF) and other neurotrophins have been suggested to be an interesting target for treatment and a biomarker for diagnosis and evaluation of treatment outcome [454647]. However, even if the effects of a humanized NGF antibody (tanezumab) in patients with interstitial cystitis seemed promising [48], adverse effects found in nonbladder studies stopped further development [49]. However, developments in other areas than LUT, e.g., pain seem to have been resumed [50].
Prostaglandin E2, acting via EP1 receptors, stimulates bladder contractile activity by sensitization of afferent nerves, and is increased in urine from patients with LUTS [51]. Despite promising results in animal experiments, a double-blind, placebo-controlled phase II study in OAB patients concluded that the role of an EP1 receptor antagonist in the management of OAB syndrome is minimal [52].
Rho-kinase inhibition is a theoretically interesting principle for inhibition of bladder overactivity [53], since upregulation of the Rho-kinase pathway has been associated with bladder changes in diabetes, outflow obstruction, and idiopathic DO. The vitamin D3 agonist, elocalcitol, was shown to have an inhibitory effect on the RhoA/Rho kinase pathway [5455], and showed some promising effects in female patients with OAB [56]. However, whether or not vitamin D3 receptor agonism (monotherapy or in combination) will be a useful alternative for the treatment of LUTS/OAB, requires further randomized controlled trials.
K+ channel openers have shown great promise in preclinical experiments [57], but so far the K-channel openers studied clinically-have yielded disappointing results [58]. Injection of naked" Maxi-K DNA directly into detrusor may be an interesting future possibility [59].
Even if not discussed in this review, it should be noted that drugs with a central mode of action, such as neurokinin receptor antagonists, tramadol, and duloxetine have positive proof of concept documented in randomized controlled trials [60]. Currently, most of these drugs cannot for various reasons be recommended for general use in the treatment of LUTS/OAB, but they illustrate that agents with a target in the central nervous system have a potential to be therapeutically useful.
Presently, the most promising targets seem to be the purinergic [61626364] and cannabinoid [656667] systems, and different members of the transient receptor potential (TRP) channel family [686970717273]. However, even if P2X3-receptor antagonists have a good rationale and are currently being developed for treatment of nonbladder diseases, clinical experiences in bladder disorders have not yet been reported. Clinical studies with the use of exocannabinoids on LUTS are scarce and essentially restricted to multiple sclerosis patients, and the results have so far not been convincing. However, amplification of the activity of endocannabinoids by fatty acid amide hydrolase inhibitors, inhibiting their degradation, may be an attractive approach [6774], but again clinical proof of concept is lacking. Several TRP channels, including TRPV1, TRPV2, TRPV4, TRPM8, and TRPA1, are expressed in the bladder and urethra, and may act as sensors of stretch and/or chemical irritation. There seem to be several links between activation of these channels and LUTS/OAB, and the therapeutic potential for TRPV1 channel agonists (capsaicin, resiniferatoxin) has been convincingly demonstrated. However, so far the potential of any of the channel antagonists developed for nonbladder indications has not been explored clinically in lower urinary tract (LUT) dysfunction, and the adverse effect of hyperthermia of the first generation TRPV1 antagonists has delayed development. Nevertheless, TRP channels still may be most exciting targets for future LUT drugs. LUT dysfunction may not have been given the highest priority in TRP drug development, but research carried out for non-bladder diseases may be possible to apply also to LUT disorders.

CONCLUSIONS

Despite the limitations of current OAB treatments, which would be expected to stimulate the search for new alternatives, there are few innovations in the pipe-line, and most developments concern modifications of existing principles (antimuscarinics, β3-receptor agonists, BoNT A). Several new and old targets/drugs of potential interest for further development can be identified, however, further exploration of their involvement in lower urinary tract function/dysfunction is necessary.

Figures and Tables

Table 1

Drugs and targets of potential interest

kju-56-673-i001
Nerve growth factor - Inhibitor
Prostanoid receptors - Antagonists
Rho-kinase - Inhibitors
Vitamin D3 receptor - Agonists
K+ channels - K+ channel openers
Centrally acting drugs
Purinergic system - P2X3 receptor antagonists
Cannabinoid system - exocannabinoids; FAAH inhibitors
TRP channel family - TRP channel antagonists

FAAH, fatty acid amide hydrolase; TRP, transient receptor potential.

Notes

CONFLICTS OF INTEREST Consultancy and Advisory Boards: Allergan, Astellas, Ferring. Except for that, the author has nothing to disclose.

References

1. Andersson KE, Martin N, Nitti V. Selective β3-adrenoceptor agonists for the treatment of overactive bladder. J Urol. 2013; 190:1173–1180.
2. Chapple CR, Cardozo L, Nitti VW, Siddiqui E, Michel MC. Mirabegron in overactive bladder: a review of efficacy, safety, and tolerability. Neurourol Urodyn. 2014; 33:17–30.
3. Mangera A, Apostolidis A, Andersson KE, Dasgupta P, Giannantoni A, Roehrborn C, et al. An updated systematic review and statistical comparison of standardised mean outcomes for the use of botulinum toxin in the management of lower urinary tract disorders. Eur Urol. 2014; 65:981–990.
4. Lythgoe C, McVary KT. The use of PDE-5 inhibitors in the treatment of lower urinary tract symptoms due to benign prostatic hyperplasia. Curr Urol Rep. 2013; 14:585–594.
5. Yeo EK, Hashim H, Abrams P. New therapies in the treatment of overactive bladder. Expert Opin Emerg Drugs. 2013; 18:319–337.
6. Zacche MM, Giarenis I, Cardozo L. Phase II drugs that target cholinergic receptors for the treatment of overactive bladder. Expert Opin Investig Drugs. 2014; 23:1365–1374.
7. Sacco E, Bientinesi R. Innovative pharmacotherapies for women with overactive bladder: where are we now and what is in the pipeline? Int Urogynecol J. 2015; 26:629–640.
8. Bechis SK, Kim MM, Wintner A, Kreydin EI. Differential response to medical therapy for male lower urinary tract symptoms. Curr Bladder Dysfunct Rep. 2015; 10:177–185.
9. Salcedo C, Davalillo S, Cabellos J, Lagunas C, Balsa D, Perez-Del-Pulgar S, et al. In vivo and in vitro pharmacological characterization of SVT-40776, a novel M3 muscarinic receptor antagonist, for the treatment of overactive bladder. Br J Pharmacol. 2009; 156:807–817.
10. Song M, Kim JH, Lee KS, Lee JZ, Oh SJ, Seo JT, et al. The efficacy and tolerability of tarafenacin, a new muscarinic acetylcholine receptor M3 antagonist in patients with overactive bladder; randomised, double-blind, placebo-controlled phase 2 study. Int J Clin Pract. 2015; 69:242–250.
11. Michel MC, Igawa Y. Therapeutic targets for overactive bladder other than smooth muscle. Expert Opin Ther Targets. 2015; 19:687–705.
12. Dmochowski RR, Staskin DR, Duchin K, Paborji M, Tremblay TM. Clinical safety, tolerability and efficacy of combination tolterodine/pilocarpine in patients with overactive bladder. Int J Clin Pract. 2014; 68:986–994.
13. Andersson KE, Campeau L, Olshansky B. Cardiac effects of muscarinic receptor antagonists used for voiding dysfunction. Br J Clin Pharmacol. 2011; 72:186–196.
14. Rosa GM, Bauckneht M, Scala C, Tafi E, Leone Roberti Maggiore U, Ferrero S, et al. Cardiovascular effects of antimuscarinic agents in overactive bladder. Expert Opin Drug Saf. 2013; 12:815–827.
15. Biers SM, Reynard JM, Brading AF. The effects of a new selective beta3-adrenoceptor agonist (GW427353) on spontaneous activity and detrusor relaxation in human bladder. BJU Int. 2006; 98:1310–1314.
16. Takasu T, Ukai M, Sato S, Matsui T, Nagase I, Maruyama T, et al. Effect of (R)-2-(2-aminothiazol-4-yl)-4'-{2-[(2-hydroxy-2-phenylethyl)amino]ethyl} acetanilide (YM178), a novel selective beta3-adrenoceptor agonist, on bladder function. J Pharmacol Exp Ther. 2007; 321:642–647.
17. Hatanaka T, Ukai M, Watanabe M, Someya A, Ohtake A, Suzuki M, et al. In vitro and in vivo pharmacological profile of the selective β3-adrenoceptor agonist mirabegron in rats. J Naunyn Schmiedebergs Arch Pharmacol. 2013; 386:247–253.
18. Igawa Y, Michel MC. Pharmacological profile of β3-adrenoceptor agonists in clinical development for the treatment of overactive bladder syndrome. Naunyn Schmiedebergs Arch Pharmacol. 2013; 386:177–183.
19. Gillespie JI, Palea S, Guilloteau V, Guerard M, Lluel P, Korstanje C. Modulation of non-voiding activity by the muscarinergic antagonist tolterodine and the β(3)-adrenoceptor agonist mirabegron in conscious rats with partial outflow obstruction. BJU Int. 2012; 110(2 Pt 2):E132–E142.
20. Aizawa N, Homma Y, Igawa Y. Effects of mirabegron, a novel β3-adrenoceptor agonist, on primary bladder afferent activity and bladder microcontractions in rats compared with the effects of oxybutynin. Eur Urol. 2012; 62:1165–1173.
21. Eastham JE, Gillespie JI. The concept of peripheral modulation of bladder sensation. Organogenesis. 2013; 9:224–233.
22. Gillespie JI, Rouget C, Palea S, Granato C, Korstanje C. Beta adrenergic modulation of spontaneous microcontractions and electrical field-stimulated contractions in isolated strips of rat urinary bladder from normal animals and animals with partial bladder outflow obstruction. Naunyn Schmiedebergs Arch Pharmacol. 2015; 388:719–726.
23. Gillespie JI, Rouget C, Palea S, Granato C, Birder L, Korstanje C. The characteristics of intrinsic complex micro-contractile activity in isolated strips of the rat bladder. Naunyn Schmiedebergs Arch Pharmacol. 2015; 388:709–718.
24. Granato C, Korstanje C, Guilloteau V, Rouget C, Palea S, Gillespie JI. Prostaglandin E2 excitatory effects on rat urinary bladder: a comparison between the β-adrenoceptor modulation of non-voiding activity in vivo and micro-contractile activity in vitro. Naunyn Schmiedebergs Arch Pharmacol. 2015; 388:727–735.
25. Rouget C, Rekik M, Camparo P, Botto H, Rischmann P, Lluel P, et al. Modulation of nerve-evoked contractions by β3-adrenoceptor agonism in human and rat isolated urinary bladder. Pharmacol Res. 2014; 80:14–20.
26. D'Agostino G, Maria Condino A, Calvi P. Involvement of β3-adrenoceptors in the inhibitory control of cholinergic activity in human bladder: Direct evidence by [(3)H]-acetylcholine release experiments in the isolated detrusor. Eur J Pharmacol. 2015; 758:115–122.
27. Ohlstein EH, von Keitz A, Michel MC. A multicenter, double-blind, randomized, placebo-controlled trial of the β3-adrenoceptor agonist solabegron for overactive bladder. Eur Urol. 2012; 62:834–840.
28. Leon LA, Hoffman BE, Gardner SD, Laping NJ, Evans C, Lashinger ES, et al. Effects of the beta 3-adrenergic receptor agonist disodium 5-[(2R)-2-[[(2R)-2-(3-chlorophenyl)-2-hydroxyethyl] amino]propyl]-1,3-benzodioxole-2,2-dicarboxylate (CL-316243) on bladder micturition reflex in spontaneously hypertensive rats. J Pharmacol Exp Ther. 2008; 326:178–185.
29. Tasler S, Baumgartner R, Behr-Roussel D, Oger-Roussel S, Gorny D, Giuliano F, et al. An aryloxypropanolamine hβ3-adrenoceptor agonist as bladder smooth muscle relaxant. Eur J Pharm Sci. 2012; 46:381–387.
30. Moyes CR, Berger R, Goble SD, Harper B, Shen DM, Wang L, et al. Design, synthesis, and evaluation of conformationally restricted acetanilides as potent and selective β3 adrenergic receptor agonists for the treatment of overactive bladder. J Med Chem. 2014; 57:1437–1453.
31. Kanie S, Otsuka A, Yoshikawa S, Kobayashi R, Itaba S, Yokokawa H, et al. TRK-380, a novel selective human β3-adrenoceptor agonist, ameliorates formalin-induced pollakiuria in rats and carbachol-induced bladder contraction in dogs. Urology. 2013; 82:975.e7–975.e12.
32. Otsuka A, Shinbo H, Hasebe K, Matsumoto R, Ozono S. Effects of a novel beta(3)-adrenoceptor agonist, AJ-9677, on relaxation of the detrusor muscle: an in vitro study. Int J Urol. 2008; 15:1072–1076.
33. Afeli SA, Rovner ES, Petkov GV. BRL37344, a β3-adrenergic receptor agonist, decreases nerve-evoked contractions in human detrusor smooth muscle isolated strips: role of BK channels. Urology. 2013; 82:744.e1–744.e7.
34. Keith F, John C. Targeted secretion inhibitors-innovative protein therapeutics. Toxins (Basel). 2010; 2:2795–2815.
35. Edupuganti OP, Ovsepian SV, Wang J, Zurawski TH, Schmidt JJ, Smith L, et al. Targeted delivery into motor nerve terminals of inhibitors for SNARE-cleaving proteases via liposomes coupled to an atoxic botulinum neurotoxin. FEBS J. 2012; 279:2555–2567.
36. Kuo HC, Liu HT, Chuang YC, Birder LA, Chancellor MB. Pilot study of liposome-encapsulated onabotulinumtoxina for patients with overactive bladder: a single-center study. Eur Urol. 2014; 65:1117–1124.
37. Kane CD, Nuss JE, Bavari S. Novel therapeutic uses and formulations of botulinum neurotoxins: a patent review (2012 - 2014). Expert Opin Ther Pat. 2015; 25:675–690.
38. Chuang YC, Tyagi P, Huang CC, Yoshimura N, Wu M, Kaufman J, et al. Urodynamic and immunohistochemical evaluation of intravesical botulinum toxin A delivery using liposomes. J Urol. 2009; 182:786–792.
39. ClinicalTrials.gov [Internet]. Bethesda (MD): National Institutes of Health;c2015. [cited 2015 Jul 28]. Available from: https://clinicaltrials.gov/.
40. Füllhase C, Chapple C, Cornu JN, De Nunzio C, Gratzke C, Kaplan SA, et al. Systematic review of combination drug therapy for non-neurogenic male lower urinary tract symptoms. Eur Urol. 2013; 64:228–243.
41. Lee SH, Lee JY. Current role of treatment in men with lower urinary tract symptoms combined with overactive bladder. Prostate Int. 2014; 2:43–49.
42. Schauer I, Madersbacher S. Medical treatment of lower urinary tract symptoms/benign prostatic hyperplasia: anything new in 2015. Curr Opin Urol. 2015; 25:6–11.
43. Abrams P, Kelleher C, Staskin D, Rechberger T, Kay R, Martina R, et al. Combination treatment with mirabegron and solifenacin in patients with overactive bladder: efficacy and safety results from a randomised, double-blind, dose-ranging, phase 2 study (Symphony). Eur Urol. 2015; 67:577–588.
44. Bechis SK, Otsetov AG, Ge R, Olumi AF. Personalized medicine for the management of benign prostatic hyperplasia. J Urol. 2014; 192:16–23.
45. Ochodnicky P, Cruz CD, Yoshimura N, Cruz F. Neurotrophins as regulators of urinary bladder function. Nat Rev Urol. 2012; 9:628–637.
46. Cruz CD. Neurotrophins in bladder function: what do we know and where do we go from here? Neurourol Urodyn. 2014; 33:39–45.
47. Seth JH, Sahai A, Khan MS, van der Aa F, de Ridder D, Panicker JN, et al. Nerve growth factor (NGF): a potential urinary biomarker for overactive bladder syndrome (OAB)? BJU Int. 2013; 111:372–380.
48. Evans RJ, Moldwin RM, Cossons N, Darekar A, Mills IW, Scholfield D. Proof of concept trial of tanezumab for the treatment of symptoms associated with interstitial cystitis. J Urol. 2011; 185:1716–1721.
49. Leite VF, Buehler AM, El Abd O, Benyamin RM, Pimentel DC, Chen J, et al. Anti-nerve growth factor in the treatment of low back pain and radiculopathy: a systematic review and a metaanalysis. Pain Physician. 2014; 17:E45–E60.
50. Sopata M, Katz N, Carey W, Smith MD, Keller D, Verburg KM, et al. Efficacy and safety of tanezumab in the treatment of pain from bone metastases. Pain. 2015; 156:1703–1713.
51. Rahnama'i MS, van Kerrebroeck PE, de Wachter SG, van Koeveringe GA. The role of prostanoids in urinary bladder physiology. Nat Rev Urol. 2012; 9:283–290.
52. Chapple CR, Abrams P, Andersson KE, Radziszewski P, Masuda T, Small M, et al. Phase II study on the efficacy and safety of the EP1 receptor antagonist ONO-8539 for nonneurogenic overactive bladder syndrome. J Urol. 2014; 191:253–260.
53. Christ GJ, Andersson KE. Rho-kinase and effects of Rho-kinase inhibition on the lower urinary tract. Neurourol Urodyn. 2007; 26:6 Suppl. 948–954.
54. Morelli A, Vignozzi L, Filippi S, Vannelli GB, Ambrosini S, Mancina R, et al. BXL-628, a vitamin D receptor agonist effective in benign prostatic hyperplasia treatment, prevents RhoA activation and inhibits RhoA/Rho kinase signaling in rat and human bladder. Prostate. 2007; 67:234–247.
55. Penna G, Fibbi B, Amuchastegui S, Corsiero E, Laverny G, Silvestrini E, et al. The vitamin D receptor agonist elocalcitol inhibits IL-8-dependent benign prostatic hyperplasia stromal cell proliferation and inflammatory response by targeting the RhoA/Rho kinase and NF-kappaB pathways. Prostate. 2009; 69:480–493.
56. Digesu GA, Verdi E, Cardozo L, Olivieri L, Khullar V, Colli E. Phase IIb, multicenter, double-blind, randomized, placebocontrolled, parallel-group study to determine effects of elocalcitol in women with overactive bladder and idiopathic detrusor overactivity. Urology. 2012; 80:48–54.
57. Petkov GV. Role of potassium ion channels in detrusor smooth muscle function and dysfunction. Nat Rev Urol. 2011; 9:30–40.
58. Andersson KE, Chapple CR, Cardozo L, Cruz F, Hashim H, Michel MC, et al. Pharmacological treatment of urinary incontinence.In : Abrams P, Cardozo L, Khoury S, Wein AJ, editors. Incontinence, 5th International Consultation on Incontinence. [Paris]: ICUD-EAU;2013. p. 623–728.
59. Christ GJ, Day NS, Day M, Santizo C, Zhao W, Sclafani T, et al. Bladder injection of "naked" hSlo/pcDNA3 ameliorates detrusor hyperactivity in obstructed rats in vivo. Am J Physiol Regul Integr Comp Physiol. 2001; 281:R1699–R1709.
60. Andersson KE. LUTS treatment: future treatment options. Neurourol Urodyn. 2007; 26:6 Suppl. 934–947.
61. Ford AP, Cockayne DA. ATP and P2X purinoceptors in urinary tract disorders. Handb Exp Pharmacol. 2011; (202):485–526.
62. Ford AP, Undem BJ. The therapeutic promise of ATP antagonism at P2X3 receptors in respiratory and urological disorders. Front Cell Neurosci. 2013; 7:267.
63. North RA, Jarvis MF. P2X receptors as drug targets. Mol Pharmacol. 2013; 83:759–769.
64. Andersson KE. Purinergic signalling in the urinary bladder. Auton Neurosci. 2015; 191:78–81.
65. Tyagi P, Tyagi V, Yoshimura N, Chancellor M. Functional role of cannabinoid receptors in urinary bladder. Indian J Urol. 2010; 26:26–35.
66. Ruggieri MR Sr. Cannabinoids: potential targets for bladder dysfunction. Handb Exp Pharmacol. 2011; (202):425–451.
67. Hedlund P. Cannabinoids and the endocannabinoid system in lower urinary tract function and dysfunction. Neurourol Urodyn. 2014; 33:46–53.
68. Everaerts W, Gevaert T, Nilius B, De Ridder D. On the origin of bladder sensing: Tr(i)ps in urology. Neurourol Urodyn. 2008; 27:264–273.
69. Andersson KE, Gratzke C, Hedlund P. The role of the transient receptor potential (TRP) superfamily of cation-selective channels in the management of the overactive bladder. BJU Int. 2010; 106:1114–1127.
70. Skryma R, Prevarskaya N, Gkika D, Shuba Y. From urgency to frequency: facts and controversies of TRPs in the lower urinary tract. Nat Rev Urol. 2011; 8:617–630.
71. Avelino A, Charrua A, Frias B, Cruz C, Boudes M, de Ridder D, et al. Transient receptor potential channels in bladder function. Acta Physiol (Oxf). 2013; 207:110–122.
72. Deruyver Y, Voets T, De Ridder D, Everaerts W. Transient receptor potential channel modulators as pharmacological treatments for lower urinary tract symptoms (LUTS): myth or reality? BJU Int. 2015; 115:686–697.
73. Franken J, Uvin P, De Ridder D, Voets T. TRP channels in lower urinary tract dysfunction. Br J Pharmacol. 2014; 171:2537–2551.
74. Fullhase C, Russo A, Castiglione F, Benigni F, Campeau L, Montorsi F, et al. Spinal cord FAAH in normal micturition control and bladder overactivity in awake rats. J Urol. 2013; 189:2364–2370.
TOOLS
Similar articles