eISSN: 2299-0038
ISSN: 1643-8876
Menopause Review/Przegląd Menopauzalny
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank


1/2020
vol. 19
 
Share:
Share:
Review paper

An update on treatment options for interstitial cystitis

Simone Garzon
1
,
Antonio Simone Laganà
1
,
Jvan Casarin
1
,
Ricciarda Raffaelli
2
,
Antonella Cromi
1
,
Davide Sturla
1
,
Massimo Franchi
2
,
Fabio Ghezzi
1

1.
Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Varese, Italy
2.
Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Verona, Italy
Menopause Rev 2020; 19(1): 35-43
Online publish date: 2020/04/27
Article file
Get citation
 
PlumX metrics:
 

Introduction

Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic pelvic pain syndrome characterized by pain, pressure, or discomfort perceived to be related to the urinary bladder, that lasts more than 6 months with at least one other urinary symptom such as frequency or persistent urge [1]. Because other diseases can provide same symptoms, the exclusion of infection or other identifiable cause(s) is mandatory, as well as further investigation to document possible overlapping of multiple conditions [1, 2]. In the clinical setting, a shorter period of 6 weeks duration was proposed to allow early diagnosis and treatment [3].
The term BPS alone is considered the most appropriate, because the related clinical diagnostic criteria are more inclusive than those based only on cystoscopy and histological findings, which does not recognize up to the 60% of patients. On that basis, some guidelines does not further require cystoscopy for the diagnosis [1, 3-5]. The BPS is defined by clinical symptoms and is based on the hypothesis that women affected by this condition are a homogenous group having a disease with the same etiopathogenesis, with subgroups distinguished by positive signs [5]. Nevertheless, although in IC/BPS the terms BPS and IC are reported together due to formal/historical reasons, they are not actually the same concept. IC is a subgroup (type) of BPS with cystoscopic and histologic signs of interstitial inflammation fulfilling the diagnostic requirements of the original term “IC” [1]. IC diagnosis requires cystoscopy with bladder hydrodistension and/or some peculiar morphological findings in bladder biopsies. The findings of cystoscopy can be glomerulations or Hunner lesions; meanwhile, histologic evaluation of mucosal biopsy can show inflammatory infiltrates, granulation tissue, detrusor mastocytosis, and/or interfascicular fibrosis [1, 4, 5]. The diagnosis of BPS is actually based on clinical symptoms after the exclusion of other urological and gynecological conditions having similar presentation and/or on the cystoscopic/histologic findings [4]. Noteworthy, often patients refer to the gynecologist based on the chronic pelvic pain that characterizes the disease. In this scenario, an appropriate gynecological evaluation is of paramount importance to exclude other pathologies, such as endometriosis [2, 6-8]. Endometriosis is able to present with similar symptoms and can directly involve the bladder [9], and not rarely the two conditions are overlapping [10]. However, the correct identification of symptoms and the exclusion of other pathologies as well as the suspect of an overlapping condition are important to appropriately refer the patient to a urologist or urogynecologist [11].
The improved understanding of the BPS pathophysiology has raised the numbers of diagnosis, although the variations over time of BPS diagnostic criteria has meant that epidemiological studies reported different statistics on prevalence [5]. Based on the less and the most inclusive definitions, the BPS prevalence in women was reported ranging between 0.83% and 2.71% [12]. Similarly, the 6.53% vs. the 2.70% of women who met symptom criteria, respectively based on high sensitivity vs. high specificity definitions [13].
Although the growing body of evidence about the IC/BPS clinical presentation and pathophysiology, the real etiopathogenesis is not completely understood as well as the role of the different cystoscopic/histologic presentation. These factors limit the available evidence-based treatment options, that are of paramount importance to maximize treatment outcomes since the early stages of the disease [5, 14]. Noteworthy, the early diagnosis has a key role in the syndrome [15], that is a pain syndrome and can determine a pelvic cross-organ sensitization worsening symptoms and hardening management and resolution.

Treatment option overview

The ideal treatment should match as much as possible with the pathophysiologic causes of the IC/BPS, but the scarce evidence limits this approach with the majority of available treatments primarily targeted at symptom control [5]. Treatments strategies have traditionally focused on the bladder, that is considered the primary end organ and source of pain, where most of the IC/BPS symptoms seemed to manifest. Nevertheless, the growing body of evidence on the IC/BPS pathophysiology suggested a multifaceted nature of the disease with systemic components [16]. On that basis, the available treatment options increased from the local (bladder/pelvic) approach to the systemic approach, supporting a multifactorial and comprehensive management [14]. In general, guidelines recommend the personalized and progressive approach, that starts from the more conservative options and then advances toward more invasive and combined treatments [3, 17].

Conservative treatment

The conservative management of IC/BPS represents the first-line treatment option. Behavioral and diet changes, psychological stress management, urogynecological exercises, and heat or cold therapy represent the first therapeutic strategies [3, 17]. All these interventions are available only after adequate patient education, that represents the first actual step. Patient needs to be made aware about the bladder function, what is known about IC/BPS, that multiple trials may be required before acceptable symptom control, that often multiple simultaneous approach are necessary, and that specific behavior might improve or worsen IC/BPS symptoms [18]. Because most of these interventions are inexpensive and risk free, they should be encouraged. Diet restriction with the reduced consumption of coffee, tea, alcohol, chocolate and spicy food are reported improving IC/BPS symptoms. This intervention may allow to identify which specific food or fluid may affect each patient. Additionally, the regulation of diet and fluids intake reduces constipation and normalizes the frequency of urination [18, 19]. Pelvic floor relaxation exercises (placing knees against the chest, reclining with spread legs, or squatting) and bladder training have also been shown to improve symptoms increasing intervals between urinations and void volume [20, 21]. Although all of these approaches are able to improve IC/BPS, the psychological support with appropriate coping strategies and stress management has a key role in these women. IC/BPS symptoms are reported related to stress [22-24] and depression similarly to other chronic pain syndromes [25-28]. Psychological, emotional, and social support resulted in an improved health, quality of life, and symptoms [18].

Non-pharmacological treatment

The utility of pelvic floor physical therapy for the management of BPS is related to the hypertonic pelvic floor muscle dysfunction reported in affected women, although it is not known if these alterations are primary or secondary to IC/BPS [29]. Physical therapy is aimed to release the myofascial trigger points and the connective tissue. Randomized controlled trial reported an improvement in pain, urgency, frequency and quality of life (QoL) for women who underwent myofascial physical therapy as compared to women who underwent general therapeutic massage [30]. These results were confirmed by further studies investigating the effectiveness of physical massage in BPS, that can be usually recommended for most patients [11, 20, 31, 32].
Similarly to physical therapy, because of safety and relatively affordability, acupuncture can be considered an option in women affected by IC/BPS based on studies suggesting the effectiveness of the technique, although the evidence is limited and more data are required [11, 29, 33-35].
Conversely, conflicting results are reported about the effectiveness of transcutaneous nerve stimulation. The transcutaneous stimulation of peripheral sensory nerves aims to modulate the pain impulses and has the advantage of being available in outpatient setting. Nevertheless, it requires continuous and daily use for months to achieve modest symptoms improvement [11, 29].

Oral medications

Oral pharmacotherapy for IC/BPS represents a second line therapy that should be combined with conservative treatments [3, 33, 36].

Pentosan polysulfate

The only oral medication approved by the U.S. Food and Drug Administration (FDA) for treatment of IC/BPS is the pentosan polysulfate (PPS) [36]. PPS is a synthetic sulfated polysaccharide, which therapeutic function is supposed to be the reduction of urothelial permeability by reconstituting the glycosaminoglycan (GAG) layer of the bladder urothelium, that is thought to be compromised in patients affected by IC/BPS [5]. The symptoms of IC/BPS are supposed to be related to urothelium abnormalities caused by the disruption of the GAG layer overlining apical cells, which normally regulate the passage of cations and protects the urothelium from bacteria and toxic substances [37, 38]. PPS is one of the most studied therapies for IC/BPS, and different studies, randomized controlled trials, and meta-analysis supported the improvement of symptoms with PPS treatment as compared to placebo, with the reduction of pain, urgency, and frequency [39, 40]. Nevertheless, more recent randomized controlled trials provided conflicting results, reporting no statistically significant differences of symptoms between PPS treatment and placebo [41, 42]. On that basis, the Royal College of Obstetricians and Gynecologists (RCOG) no longer recommend PPS for treatment of IC/BPS considering the undemonstrated efficacy and the adverse effects of diarrhea, vomiting, rectal bleeding and alopecia [43]. Nevertheless, it is still recommended in other guidelines, although recognizing the limited evidence [3].

Tricyclic antidepressants

Tricyclic antidepressants have a wide range of interactions with different pattern of neurotransmitters receptors. Among them, Amitriptyline is the most studied for the treatment of IC/BPS and act blocking the reuptake of released serotonin and noradrenaline, histamine 1 receptors, and acetylcholine receptors. It is supposed that the urgency and frequency symptoms are alleviated by the anticholinergic effects, meanwhile interaction with neurotransmitters reuptake may have an analgesic effect [14]. Different studies reported an improvement of symptoms ranging between the 50% and 70% in the treated group as compared to placebo, although efficacy is directly related to the dosage as well as side effects, such as nausea, constipation, dry mouth, weight gain, blurred vision, lightheadedness, and sedation [44]. The therapeutic efficacy resulted strictly related to the highest tolerated dose, that, ranging between 25 mg to 100 mg, reported a clinical improvement up to the 63% of patients [45-47]. Nevertheless, the majority of patients are not able to tolerate and achieve the therapeutic dose with a rate of side effects up to the 79% [47].

Histamine and leukotriene receptor inhibitors

Mast cells infiltrates and detrusor mastocytosis are reported in patients affected by BPS with histological signs of IC, and it was supposed that the mediators released in hypersensitivity reaction by these cells may lead to urinary symptoms [48, 49]. On that basis, histamine receptor inhibitors were investigated as possible therapeutic option in these patients as molecules able to reduce the mastocytes activation and subsequently BPS symptoms [14]. Among them, cimetidine and hydroxyzine were the most investigated so far. Cimetidine (H2 antagonist) was related to a significant improvement of suprapubic pain and nocturia as compared to placebo in a randomized controlled trial, although the histology of the bladder biopses was reported unchanged [21, 50, 51]. Hydroxyzine is a H1 antagonist with anticholinergic activity, that reported in different observational studies an improvement of symptoms up to the 90% of treated patients, with sedation representing the primary side effect. Conversely, in a randomized controlled trial these results were not confirmed [42]. Both drugs have limited and conflicting evidence supporting their therapeutic role in IC/BPS, and some guidelines do not recommend their use [43]. On the contrary, others includes histamine receptor inhibitors in second line therapies due to their safety therapeutic profile with few side effects beyond sedation [5].
Other than histamine, leukotriene plays a key role in the activation of mast cells and eosinophils [52]. In patients with IC/BPS higher levels of leukotriene were identified in the urine of women with detrusor mastocytosis, supporting a potential key role of these mediators in the pathophysiology of IC/BPS [53]. In line with this evidence, montelukast, a leukotriene receptor-1 antagonist, was reported able to improve symptoms of patients affected by IC/BPS in a pilot study and in a case report [54, 55].

Immunosuppressants

In addition to mast cells infiltrates, in women affected by IC/BPS infiltration of the bladder mucosa by CD4 T lymphocytes and eosinophilic leukocytes was reported. This inflammatory components suggest an autoimmune pathogenesis of the disease, and humoral or cell-mediated mechanisms targeting the bladder mucosa may cause the episodic exacerbation of the IC/BPS [56, 57]. On that basis, cyclosporine A was investigated as immunosuppressant for the treatment of refractory cases of IC/BPS. Different observational studies reported an improvement of symptoms with reduced pain, increased max bladder capacity, and voided volume [58]. A subsequent randomized trial showed cyclosporine A superior to PPS with higher improved pain, frequency, void volume and nocturia at 6 months [59]. Overall, all the available pieces of evidence summarized in a systematic review support the effectiveness of cyclosporine A in the treatment of IC/BPS, even in patients who had failed one or more oral therapies [60]. Although the promising long-term therapeutic effects, the use of cyclosporin A is not without side effects. Nephrotoxicity, reduced glomerular filtration rate, increased creatinine levels, and hypertension are side effects regularly reported and should be always monitored and weighted with therapeutic benefits [58-61]. On that basis, the better understanding of the mechanism of action of cyclosporine A may allow to identify women who will benefit from the treatment and the minimum dose required to achieve a satisfactory result. Due to these side effects, cyclosporine A is recommended in patients affected by IC/BPS refractory to all other therapies [3, 33].

Rosiptor (AQX-1125)

The novel SH2-containing inositol-5-phosphatase 1 (SHIP1) activator AQX-1125 was recently investigated as potential new oral medication for the treatment of IC/BPS. It was supposed able to modulate the immune/inflammatory response thought the trigger of SHIP1 protein, which modulates phosphoinositide signaling. A phase two randomized double-blind controlled trial investigated the efficacy of a 6 weeks treatment vs. placebo, reporting promising results. Women with moderate to severe IC/BPS reported a significant improvement of pain and urinary symptoms after 6 weeks of treatment with oral AQX-1125 as compared to placebo [62]. Nevertheless, these results were not confirmed by the phase three randomized double-blind controlled trial, that compared 12 weeks of daily 100 mg or 200 mg of oral SHIP1 activator treatment with placebo. The trial concluded that SHIP1 activation is a safe but ineffective therapeutic approach to IC/BPS [63].

Intravesical medications

Treatments based on intravesical medications consist in the direct instillation of the therapeutic substance into the bladder via a catheter, that usually is combined with low-pressure, short-term hydrodistension [64]. These treatments require to rule out other pathologies and are usually recommended when less invasive treatments have failed [3, 33, 65]. In general, a regular maintenance treatment is required and recommended in those patients who report symptoms improvements [3, 33, 43].
The majority of available treatments in this category are aimed to reconstitute the glycosaminoglycan (GAG) layer of the bladder urothelium, that is thought to be compromised in patients affected by IC/BPS [5]. As reported above, the etiopathogenesis of IC/BPS is supposed to be related to urothelium abnormalities caused by the disruption of the GAG layer overlining apical cells [37, 38]. This layer is composed by different GAG, such as dermatan sulfate, chondroitin sulfate, heparan sulfate, keratin sulfate, and hyaluronic acid; and the intravesical instillation of exogenous GAG components, as monotherapy or mixed with other medications, is supposed to be able to restore this impaired layer [14].

Dimethyl sulfoxide

Dimethyl sulfoxide (DMSO) is one of the most common used intravesical medications and the only one approved by the FDA; it is usually instilled weekly for 6 weeks, alone or mixed with other medications [66]. DMSO exerts a combination of anti-inflammatory effects, collagen dissolution, smooth muscle relaxation, and nerve blockade. Three randomized-controlled trials and different cohort studies reported a symptoms improvement in up to the 95% of patients, with particular benefit for patients with ulcerative IC/BPS and without advantage provided by mixing with other medications [67]. Nevertheless, the optimal dose, timing, and type of IC/BPS most likely benefit from DMSO are not established. Of note, in more than the 35% of patients symptoms relapse within 8 weeks, and some patients cannot tolerate the pain after instillation and garlic odor [67, 68]. Although the available evidence, DMSO effectiveness is unclear, and not all guidelines recommend its use [3, 69].

Heparin and pentosan polysulfate

Heparin is a mucopolysaccharide part of the GAG family, that mimics the GAGs lining the urothelium. Moreover, it is supposed able to exert anti-inflammatory effects, promote the urothelium growth, induce fibroblast and smooth muscle proliferation, and favoring angiogenesis, with little systemic absorption [14]. Different observational studies reported a symptoms improvement in the 56-73% of patients after intravesical heparin instillation for three months, with a side effect profile comparable to placebo [69-71]. Although these pieces of evidence, the available randomized controlled trials investigated the intravesical instillation of heparin combined with lidocaine, reporting a significant improvement of IC/BPS symptoms in patients underwent heparin/lidocaine instillation as compared to placebo [72]. Similar results were provided by a later observational study after 12 weeks of therapy [73]. On that basis, the strongest evidence is provided by the use of heparin in combination with other medications, instead of monotherapy [14]. However, a randomized controlled trial comparing heparin/lidocaine with lidocaine alone reported significant better results in the combined therapy, supporting a specific role of heparin [74].
PPS is a heparin analogue, that other than the previously reported oral administration is used as intravesical medication with the aim to directly restore the GAG layer. Different placebo controlled trials reported an improvement of symptoms after the weekly intravesical administration [40, 75], and a combination of both oral and intravesical administration provided further improvement in a randomized control trial [76].

Chondroitin sulfate and hyaluronic acid

Both chondroitin sulfate and hyaluronic acid are components of the GAG layer lining the urothelium, and the intravesical instillation is aimed to restore the protective barrier [14]. Different pieces of evidence support the intravesical administration of chondroitin sulfate and hyaluronic acid alone or in combination as effective treatment for the IC/BPS [77-79]. Two randomized controlled trials comparing the combined intravesical administration of these two GAGs or only chondroitin sulfate vs. DMSO reported a symptoms improvement in both groups with higher pain relief in the chondroitin sulfate alone and plus hyaluronic acid arm [68, 80]. Nevertheless, although promising results, chondroitin sulfate reported a limited magnitude of effect when used in monotherapy [81], as well as the effect of hyaluronic acid were questioned by three unpublished randomized controlled trials [33]. On that basis, the use of these GAGs is not recommended as monotherapy but as part of a multimodal approach [33].

Lidocaine

Lidocaine is a local anesthetic with anti-inflammatory effects administered intravesical in the alkalinized form, that allow a better penetration of the bladder epithelium [14].
Different studies reported a significant improvement of symptoms after lidocaine instillation with a rapid effect, even in monotherapy or combined with other medications [82-84]. Moreover, its use was recently proposed as a method to characterize the peripheral vs. central components of the syndrome in affected patients [85]. The main limit of lidocaine as monotherapy is the short-term effects, that induced the research of new strategies to provide a continuous administration of the drug. On that basis, a water-permeable tube pellet continuously releasing lidocaine was developed and tested as drug delivery device to be introduced in the balder by cystoscopy. In a prospective pilot study, the device resulted well tolerated and effective with the reduction of pain, urgency, and voiding frequency [86]. In general, lidocaine remains as one of the main intravesical instillation option in different guidelines [3, 33, 43].

Liposomes

Liposomes are biocompatible drug carriers composed by phospholipids and sphingomyelins. They are able to adhere onto the membrane surface of the urothelium and to favor the endocytosis with subsequent penetration of drugs, toxins, and oligonucleotides into the epithelium after the intravesical administration [87]. Because sphingomyelin is a phospholipid of cell membranes, it is supposed able to repair and promote healing of the external cell layer and decrease the permeability of urothelium lining. On that basis, empty liposomes composed by sphingomyelin were investigated in a prospective open-label cohort study, that reported an improvement of pain, urinary urgency, and overall symptoms with no side effects [88]. In addition, liposomes were investigated as carrier of botulinum toxin A in patients with refractory IC/BPS as alternative to the needle injection, although no benefit was reported as compared to placebo [89].

Others

Although in general they are not recommended by guidelines, some drugs are intravesically administered for their supposed effects in conjunction with other treatments, such as steroids, capsaicin, resiniferatoxin, and sodium cromoglycate [17, 90]. Among the corticosteroids, triamcinolone was proposed both via intravesical instillation and via submucosal injection through cystoscopy reporting an improvement of symptoms in the 70% of the patients [91].

Intravesical physical treatment

The intravesical physical treatments represent the third line option in the treatment of IC/BPS and consist in the bladder hydrodistension under general anesthesia and in the electrocoagulation of the Hunner’s lesions [3, 17]. Hunner’s lesions and glomerulations are present in the 4-10% of patients with BPS and represent a specific cystoscopic finding of IC [92]. The electrocoagulation of these lesions was associated to an improvement of symptoms in up to the 90% of women [93-95], although a significant proportion of them needs to repeat the procedure after 2-5 years [96]. Usually, Hunner’s lesion ablation is performed associated to the bladder hydrodistension with improvement of results when combined [94]. The bladder hydrodistension under general anesthesia was investigated both alone and combined with other procedures, such as the above reported lesion ablation and the instillation of intravesical medications. The available evidence showed an improvement of symptoms up the 30-55% of patients, although the beneficial effects are variable and decline over time requiring repeated procedures [93-95, 97, 98]. Randomized controlled trials are not available and cases of bladder rupture or necrosis were reported [99, 100]. However, hydrodistension is one of the most commonly performed procedure and is recommended as optional treatment after conservative and medical options have failed [33, 43].

Neuromodulation

The neuromodulation both proximal, by sacral neuromodulation, and distal, with the use of botulinum toxin A, represents a fourth line of treatment limited to patients refractory to other options previously described [17, 33, 43].

Botulinum toxin A

Intravesical botulin toxin A injection for the treatment of IC/BPS was investigated in seven randomized controlled trials and multiple prospective studies [101-103]. The toxin acts inhibiting the acetylcholine release at the motor neurons endplate with muscle relaxation and is supposed providing antinociceptive and anti-inflammatory activity [104]. It is usually utilized for the treatment of overreactive bladder syndrome via cystoscopic injections in the detrusor muscle [14]. The same treatment in patients affected by IC/BPS was reported by different randomized controlled trials associated to a significant improvement of symptoms, pain, frequency of urination, and maximum bladder capacity. Conversely, nocturia, dysuria, and maximal urinary flow rate resulted not improved [101-103]. In general, the intravesical botulin toxin A injection is a safe and effective procedure, even after repeated treatment due the limited duration (9-10 months) of the effect [105-107]. Nevertheless, the cost and the possible side effects, particularly the urinary retention requiring catheterization, suggest performing this treatment with caution and only after other less invasive approaches have failed [3, 33, 43].
Based on these side effects, the noninvasive intravesical instillation of the toxin was investigated as alternative administration route, showing limited efficacy with only a significant improvement of bladder voiding volume [108].

Sacral neuromodulation

Sacral neuromodulation is an invasive treatment approved for other bladder syndromes but not for the management of IC/BPS [109]. Nevertheless, different guidelines consider this treatment an option for patients refractory to other less invasive treatment approaches [3, 33, 43]. Although evidence is based only on non-randomized and non-controlled trials, a growing body of literature shows a significant improvement of symptoms, such as pain, frequency of urination, urgency, and maximum bladder capacity, even after long-term follow-up [110]. The use of this approach as one of the last options is related to different issues. Only the 50-60% of patients are eligible for permanent implantation, and up to the 50% of them require to remove the implant [110-112]. The technique and device is expensive and related to failure in symptoms improvements and to the development of side effects, such as uncomfortable sensations, painful stimulation, seroma, and infections [33].

Surgery

The surgical approach is the last option in case of severe symptoms significantly affecting the quality of life and refractory to all other available treatments [3, 33, 43]. The available surgical procedures are the partial supratrigonal cystectomy with augmentation cystoplasty and the urinary diversion with or without cystectomy. Partial supratrigonal cystectomy was associated to an improvement of pain, urinary symptoms, and quality of life [113, 114]; in addition, urinary diversion was reported effective to improve symptoms in the 75-85% of woman, with required cystectomy in the others for persistent pain [115, 116]. Although a limited and selected number of patients require such treatment, a few of them will still continue to report pain. The patients more likely to have improvement of symptoms were reported those with evident bladder disease, such as Hunner’s lesions and fixed reduced bladder capacity [5]. Considering the radicality of the treatment and the possible failure in symptoms improvements, referring to a specialized center is mandatory.

Conclusions

A wide range of treatment options is available for the management of patients with IC/BPS. After the correct diagnosis, the personalization of treatment with a progressive and multimodal approach represent the key element for the correct management of these patients. However, although the multiple treatment available options, which allow a wide range of possible combinations and a high level of personalization, the optimal treatment is not easy to be found. Because of the limited knowledge about the etiopathogenesis and the nature of the disease, most of the available treatments have a limited effect and are actually aimed to manage symptoms.
On that basis, further investigation is required to clarify the multifactorial etiopathogenetic mechanisms, the differences between possible subgroups, and the interaction between central and peripherical factors. Only this further evidence may allow to provide a real improvement in the treatment and management of these patients.

Disclosure

The authors report no conflict of interest.

References

1. van de Merwe JP, Nordling J, Bouchelouche P, et al. Diagnostic Criteria, Classification, and Nomenclature for Painful Bladder Syndrome/Interstitial Cystitis: An ESSIC Proposal. Eur Urol 2008; 53: 60-67.
2. Triolo O, Laganà AS, Sturlese E. Chronic Pelvic Pain in Endometriosis: An Overview. J Clin Med Res 2013; 5: 153-163.
3. Hanno PM, Erickson D, Moldwin R, Faraday MM. Diagnosis and Treatment of Interstitial Cystitis/Bladder Pain Syndrome: AUA Guideline Amendment. J Urol 2015; 193: 1545-1553.
4. Davis NF, Gnanappiragasam S, Thornhill JA. Interstitial cystitis/painful bladder syndrome: the influence of modern diagnostic criteria on epidemiology and on Internet search activity by the public. Transl Androl Urol 2015; 4: 506-511.
5. Marcu I, Campian EC, Tu FF. Interstitial Cystitis/Bladder Pain Syndrome. Semin Reprod Med 2018; 36: 123-135.
6. Laganà AS, Garzon S, Götte M, et al. The Pathogenesis of Endometriosis: Molecular and Cell Biology Insights. Int J Mol Sci 2019; 20: 5615.
7. Riemma G, Laganà AS, Schiattarella A, et al. Ion Channels in The Pathogenesis of Endometriosis: A Cutting-Edge Point of View. Int J Mol Sci 2020; 21: 1114.
8. Della Corte L, Noventa M, Ciebiera M, et al. Phytotherapy in endometriosis: an up-to-date review. J Complement Integr Med 2019. doi: 10.1515/jcim-2019-0084.
9. Laganà AS, Vitale SG, Trovato MA, et al. Full-Thickness Excision versus Shaving by Laparoscopy for Intestinal Deep Infiltrating Endometriosis: Rationale and Potential Treatment Options. BioMed Res Int 2016; 2016: 3617179.
10. Wu C-C, Chung S-D, Lin H-C. Endometriosis increased the risk of bladder pain syndrome/interstitial cystitis: A population-based study. Neurourol Urodyn 2018; 37: 1413-1418.
11. Huffman MM, Slack A, Hoke M. Bladder Pain Syndrome. Prim Care 2019; 46: 213-221.
12. Clemens JQ, Link CL, Eggers PW, et al. Prevalence of painful bladder symptoms and effect on quality of life in black, Hispanic and white men and women. J Urol 2007; 177: 1390-1394.
13. Berry SH, Elliott MN, Suttorp M, et al. Prevalence of symptoms of bladder pain syndrome/interstitial cystitis among adult females in the United States. J Urol 2011; 186: 540-544.
14. Giusto LL, Zahner PM, Shoskes DA. An evaluation of the pharmacotherapy for interstitial cystitis. Expert Opin Pharmacother 2018; 19: 1097-1108.
15. Forrest JB, Vo Q. Observations on the presentation, diagnosis, and treatment of interstitial cystitis in men. Urology 2001; 57 (Suppl 1): 26-29.
16. Patnaik SS, Laganà AS, Vitale SG, et al. Etiology, pathophysiology and biomarkers of interstitial cystitis/painful bladder syndrome. Arch Gynecol Obstet 2017; 295: 1341-1359.
17. Hanno PM, Burks DA, Clemens JQ, et al. AUA guideline for the diagnosis and treatment of interstitial cystitis/bladder pain syndrome. J Urol 2011; 185: 2162-2170.
18. Bosch PC, Bosch DC. Treating Interstitial Cystitis/Bladder Pain Syndrome as a Chronic Disease. Rev Urol 2014; 16: 83-87.
19. Friedlander JI, Shorter B, Moldwin RM. Diet and its role in interstitial cystitis/bladder pain syndrome (IC/BPS) and comorbid conditions. BJU Int 2012; 109: 1584-1591.
20. Weiss JM. Pelvic floor myofascial trigger points: manual therapy for interstitial cystitis and the urgency-frequency syndrome. J Urol 2001; 166: 2226-2231.
21. Hanley RS, Stoffel JT, Zagha RM, et al. Multimodal therapy for painful bladder syndrome / interstitial cystitis: pilot study combining behavioral, pharmacologic, and endoscopic therapies. Int Braz J Urol Off J Braz Soc Urol 2009; 35: 467-474.
22. Rothrock NE, Lutgendorf SK, Kreder KJ, et al. Stress and symptoms in patients with interstitial cystitis: a life stress model. Urology 2001; 57: 422-427.
23. Lee UJ, Ackerman AL, Wu A, et al. Chronic psychological stress in high-anxiety rats induces sustained bladder hyperalgesia. Physiol Behav 2015; 139: 541-548.
24. Gao Y, Zhang R, Chang HH, Rodríguez LV. The role of C-fibers in the development of chronic psychological stress induced enhanced bladder sensations and nociceptive responses: A multidisciplinary approach to the study of urologic chronic pelvic pain syndrome (MAPP) research network study. Neurourol Urodyn 2018; 37: 673-680.
25. Crawford A, Tripp DA, Nickel JC, et al. Depression and helplessness impact interstitial cystitis/bladder pain syndrome pain over time. Can Urol Assoc J J Assoc Urol Can 2019; 13: 328-333.
26. Laganà AS, La Rosa VL, Rapisarda AM, et al. Anxiety and depression in patients with endometriosis: impact and management challenges. Int J Womens Health 2017; 9: 323-330.
27. Till SR, As-Sanie S, Schrepf A. Psychology of Chronic Pelvic Pain: Prevalence, Neurobiological Vulnerabilities, and Treatment. Clin Obstet Gynecol 2019; 62: 22-36.
28. Laganà AS, Condemi I, Retto G, et al. Analysis of psychopathological comorbidity behind the common symptoms and signs of endometriosis. Eur J Obstet Gynecol Reprod Biol 2015; 194: 30-33.
29. Verghese TS, Riordain RN, Champaneria R, Latthe PM. Complementary therapies for bladder pain syndrome: a systematic review. Int Urogynecology J 2016; 27: 1127-1136.
30. FitzGerald M, Payne C, Lukacz E, et al. Randomized Multicenter Clinical Trial of Myofascial Physical Therapy in Women with Interstitial Cystitis/Painful Bladder Syndrome (IC/PBS) and Pelvic Floor Tenderness. J Urol 2012; 187: 2113-2118.
31. Lukban J, Whitmore K, Kellogg-Spadt S, et al. The effect of manual physical therapy in patients diagnosed with interstitial cystitis, high-tone pelvic floor dysfunction, and sacroiliac dysfunction. Urology 2001; 57 (Suppl 1): 121-122.
32. Moldwin RM, Fariello JY. Myofascial trigger points of the pelvic floor: associations with urological pain syndromes and treatment strategies including injection therapy. Curr Urol Rep 2013; 14: 409-417.
33. Cox A, Golda N, Nadeau G, et al. CUA guideline: Diagnosis and treatment of interstitial cystitis/bladder pain syndrome. Can Urol Assoc J 2016; 10: E136-E155.
34. Sönmez MG, Kozanhan B. Complete response to acupuncture therapy in female patients with refractory interstitial cystitis/bladder pain syndrome. Ginekol Pol 2017; 88: 61-67.
35. Pang R, Ali A. The Chinese approach to complementary and alternative medicine treatment for interstitial cystitis/bladder pain syndrome. Transl Androl Urol 2015; 4: 653-661.
36. Fall M, Baranowski AP, Elneil S, et al. EAU guidelines on chronic pelvic pain. Eur Urol 2010; 57: 35-48.
37. Hurst RE, Moldwin RM, Mulholland SG. Bladder defense molecules, urothelial differentiation, urinary biomarkers, and interstitial cystitis. Urology 2007; 69 (4 Suppl): 17-23.
38. Parsons CL. The role of the urinary epithelium in the pathogenesis of interstitial cystitis/prostatitis/urethritis. Urology 2007; 69 (4 Suppl): 9-16.
39. Nickel JC, Barkin J, Forrest J, et al. Randomized, double-blind, dose-ranging study of pentosan polysulfate sodium for interstitial cystitis. Urology 2005; 65: 654-658.
40. van Ophoven A, Vonde K, Koch W, et al. Efficacy of pentosan polysulfate for the treatment of interstitial cystitis/bladder pain syndrome: results of a systematic review of randomized controlled trials. Curr Med Res Opin 2019; 35: 1495-1503.
41. Nickel JC, Herschorn S, Whitmore KE, et al. Pentosan polysulfate sodium for treatment of interstitial cystitis/bladder pain syndrome: insights from a randomized, double-blind, placebo controlled study. J Urol 2015; 193: 857-862.
42. Sant GR, Propert KJ, Hanno PM, et al. A pilot clinical trial of oral pentosan polysulfate and oral hydroxyzine in patients with interstitial cystitis. J Urol 2003; 170: 810-815.
43. Management of Bladder Pain Syndrome: Green-top Guideline No. 70. BJOG Int J Obstet Gynaecol 2017; 124: e46-e72.
44. Generali JA, Cada DJ. Amitriptyline: interstitial cystitis (painful bladder syndrome). Hosp Pharm 2014; 49: 809-810.
45. van Ophoven A, Pokupic S, Heinecke A, Hertle L. A prospective, randomized, placebo controlled, double-blind study of amitriptyline for the treatment of interstitial cystitis. J Urol 2004; 172: 533-536.
46. Foster HE, Hanno PM, Nickel JC, et al. Effect of amitriptyline on symptoms in treatment naïve patients with interstitial cystitis/painful bladder syndrome. J Urol 2010; 183: 1853-1858.
47. van Ophoven A, Hertle L. Long-term results of amitriptyline treatment for interstitial cystitis. J Urol 2005; 174: 1837-1840.
48. Kim A, Han J-Y, Ryu C-M, et al. Histopathological characteristics of interstitial cystitis/bladder pain syndrome without Hunner lesion. Histopathology 2017; 71: 415-424.
49. Gamper M, Regauer S, Welter J, et al. Are mast cells still good biomarkers for bladder pain syndrome/interstitial cystitis? J Urol 2015; 193: 1994-2000.
50. Dasgupta P, Sharma SD, Womack C, Blackford HN, Dennis P. Cimetidine in painful bladder syndrome: a histopathological study. BJU Int 2001; 88: 183-186.
51. Thilagarajah R, Witherow RO, Walker MM. Oral cimetidine gives effective symptom relief in painful bladder disease: a prospective, randomized, double-blind placebo-controlled trial. BJU Int 2001; 87: 207-212.
52. Theoharides TC, Cochrane DE. Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol 2004; 146: 1-12.
53. Bouchelouche K, Kristensen B, Nordling J, et al. Increased urinary excretion of leukotriene E(4) in patients with interstitial cystitis. Urology 2001; 57 (Suppl 1): 128.
54. Wajih Ullah M, Lakhani S, Sham S, et al. Painful Bladder Syndrome/Interstitial Cystitis Successful Treatment with Montelukast: A Case Report and Literature Review. Cureus 2018; 10: e2876.
55. Bouchelouche K, Nordling J, Hald T, Bouchelouche P. The cysteinyl leukotriene D4 receptor antagonist montelukast for the treatment of interstitial cystitis. J Urol 2001; 166: 1734-1737.
56. van de Merwe JP. Interstitial cystitis and systemic autoimmune diseases. Nat Clin Pract Urol 2007; 4: 484-491.
57. Zhang L, Ihsan AU, Cao Y, et al. An Immunogenic Peptide, T2 Induces Interstitial Cystitis/Painful Bladder Syndrome: an Autoimmune Mouse Model for Interstitial Cystitis/Painful Bladder Syndrome. Inflammation 2017; 40: 2033-2041.
58. Sairanen J, Forsell T, Ruutu M. Long-term outcome of patients with interstitial cystitis treated with low dose cyclosporine A. J Urol 2004; 171: 2138-2141.
59. Sairanen J, Tammela TLJ, Leppilahti M, et al. Cyclosporine A and pentosan polysulfate sodium for the treatment of interstitial cystitis: a randomized comparative study. J Urol 2005; 174: 2235-2238.
60. Wang Z, Zhang L. Treatment effect of cyclosporine A in patients with painful bladder syndrome/interstitial cystitis: A systematic review. Exp Ther Med 2016; 12: 445-450.
61. Crescenze IM, Tucky B, Li J, et al. Efficacy, Side Effects, and Monitoring of Oral Cyclosporine in Interstitial Cystitis-Bladder Pain Syndrome. Urology 2017; 107: 49-54.
62. Nickel JC, Egerdie B, Davis E, et al. A Phase II Study of the Efficacy and Safety of the Novel Oral SHIP1 Activator AQX-1125 in Subjects with Moderate to Severe Interstitial Cystitis/Bladder Pain Syndrome. J Urol 2016; 196: 747-754.
63. Nickel JC, Moldwin R, Hanno P, et al. Targeting the SHIP1 Pathway Fails to Show Treatment Benefit in Interstitial Cystitis/Bladder Pain Syndrome: Lessons Learned from Evaluating Potentially Effective Therapies in This Enigmatic Syndrome. J Urol 2019; 202: 301-308.
64. Zhang W, Deng X, Liu C, Wang X. Intravesical treatment for interstitial cystitis/painful bladder syndrome: a network meta-analysis. Int Urogynecology J 2017; 28: 515-525.
65. Malde S, Palmisani S, Al‐Kaisy A, Sahai A. Guideline of guidelines: bladder pain syndrome. BJU Int 2018; 122: 729-743.
66. Colaco MA, Evans RJ. Current Recommendations for Bladder Instillation Therapy in the Treatment of Interstitial Cystitis/Bladder Pain Syndrome. Curr Urol Rep 2013; 14: 442-447.
67. Rawls WF, Cox L, Rovner ES. Dimethyl sulfoxide (DMSO) as intravesical therapy for interstitial cystitis/bladder pain syndrome: A review. Neurourol Urodyn 2017; 36: 1677-1684.
68. Tutolo M, Ammirati E, Castagna G, et al. A prospective randomized controlled multicentre trial comparing intravesical DMSO and chondroïtin sulphate 2% for painful bladder syndrome/interstitial cystitis. Int Braz J Urol Off J Braz Soc Urol 2017; 43: 134-141.
69. EAU Guidelines: Chronic Pelvic Pain. Uroweb. https://uroweb.org/guideline/chronic-pelvic-pain/#5 (accessed: 20 Sep 2019).
70. Kuo HC. Urodynamic results of intravesical heparin therapy for women with frequency urgency syndrome and interstitial cystitis. J Formos Med Assoc Taiwan Yi Zhi 2001; 100: 309-314.
71. Generali JA, Cada DJ. Intravesical heparin: interstitial cystitis (painful bladder syndrome). Hosp Pharm 2013; 48: 822-824.
72. Parsons CL, Zupkas P, Proctor J, et al. Alkalinized lidocaine and heparin provide immediate relief of pain and urgency in patients with interstitial cystitis. J Sex Med 2012; 9: 207-212.
73. Nomiya A, Naruse T, Niimi A, et al. On- and post-treatment symptom relief by repeated instillations of heparin and alkalized lidocaine in interstitial cystitis. Int J Urol Off J Jpn Urol Assoc 2013; 20: 1118-1122.
74. Parsons CL, Koziol JA, Proctor JG, et al. Heparin and alkalinized lidocaine versus alkalinized lidocaine for treatment of interstitial cystitis symptoms. Can J Urol 2015; 22: 7739-7744.
75. Daha LK, Lazar D, Simak R, Pflüger H. The effects of intravesical pentosanpolysulfate treatment on the symptoms of patients with bladder pain syndrome/interstitial cystitis: preliminary results. Int Urogynecol J Pelvic Floor Dysfunct 2008; 19: 987-990.
76. Davis EL, El Khoudary SR, Talbott EO, et al. Safety and efficacy of the use of intravesical and oral pentosan polysulfate sodium for interstitial cystitis: a randomized double-blind clinical trial. J Urol 2008; 179: 177-185.
77. Pyo J-S, Cho WJ. Systematic Review and Meta-Analysis of Intravesical Hyaluronic Acid and Hyaluronic Acid/Chondroitin Sulfate Instillation for Interstitial Cystitis/Painful Bladder Syndrome. Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol 2016; 39: 1618-1625.
78. Gafni-Kane A, Botros SM, Du H, et al. Measuring the success of combined intravesical dimethyl sulfoxide and triamcinolone for treatment of bladder pain syndrome/interstitial cystitis. Int Urogynecology J 2013; 24: 303-311.
79. Downey A, Hennessy DB, Curry D, et al. Intravesical chondroitin sulphate for interstitial cystitis/painful bladder syndrome. Ulster Med J 2015; 84: 161-163.
80. Cervigni M, Sommariva M, Tenaglia R, et al. A randomized, open-label, multicenter study of the efficacy and safety of intravesical hyaluronic acid and chondroitin sulfate versus dimethyl sulfoxide in women with bladder pain syndrome/interstitial cystitis. Neurourol Urodyn 2017; 36: 1178-1186.
81. Thakkinstian A, Nickel JC. Efficacy of intravesical chondroitin sulphate in treatment of interstitial cystitis/bladder pain syndrome (IC/BPS): Individual patient data (IPD) meta-analytical approach. Can Urol Assoc J 2013; 7: 195-200.
82. Henry RA, Morales A, Cahill CM. Beyond a Simple Anesthetic Effect: Lidocaine in the Diagnosis and Treatment of Interstitial Cystitis/bladder Pain Syndrome. Urology 2015; 85: 1025-1033.
83. Nickel JC, Moldwin R, Lee S, Davis EL, Henry RA, Wyllie MG. Intravesical alkalinized lidocaine (PSD597) offers sustained relief from symptoms of interstitial cystitis and painful bladder syndrome. BJU Int 2009; 103: 910-918.
84. Parsons CL. Successful downregulation of bladder sensory nerves with combination of heparin and alkalinized lidocaine in patients with interstitial cystitis. Urology 2005; 65: 45-48.
85. Offiah I, Dilloughery E, McMahon SB, O’Reilly BA. Prospective comparative study of the effects of lidocaine on urodynamic and sensory parameters in bladder pain syndrome. Int Urogynecology J 2019; 30: 1293-1301.
86. Nickel JC, Jain P, Shore N, et al. Continuous intravesical lidocaine treatment for interstitial cystitis/bladder pain syndrome: safety and efficacy of a new drug delivery device. Sci Transl Med 2012; 4: 143ra100.
87. Tyagi P, Kashyap M, Majima T, et al. Intravesical liposome therapy for interstitial cystitis. Int J Urol Off J Jpn Urol Assoc 2017; 24: 262-271.
88. Peters KM, Hasenau D, Killinger KA, et al. Liposomal bladder instillations for IC/BPS: an open-label clinical evaluation. Int Urol Nephrol 2014; 46: 2291-2295.
89. Chuang Y-C, Kuo H-C. A Prospective, Multicenter, Double-Blind, Randomized Trial of Bladder Instillation of Liposome Formulation OnabotulinumtoxinA for Interstitial Cystitis/Bladder Pain Syndrome. J Urol 2017; 198: 376-382.
90. Guo C, Yang B, Gu W, et al. Intravesical resiniferatoxin for the treatment of storage lower urinary tract symptoms in patients with either interstitial cystitis or detrusor overactivity: a meta-analysis. PloS One 2013; 8: e82591.
91. Cox M, Klutke JJ, Klutke CG. Assessment of patient outcomes following submucosal injection of triamcinolone for treatment of Hunner’s ulcer subtype interstitial cystitis. Can J Urol 2009; 16: 4536-4540.
92. Lamale LM, Lutgendorf SK, Hoffman AN, Kreder KJ. Symptoms and cystoscopic findings in patients with untreated interstitial cystitis. Urology 2006; 67: 242-245.
93. Ens G, Garrido GL. Role of cystoscopy and hydrodistention in the diagnosis of interstitial cystitis/bladder pain syndrome. Transl Androl Urol 2015; 4: 624-628.
94. Niimi A, Nomiya A, Yamada Y, et al. Hydrodistension with or without fulguration of hunner lesions for interstitial cystitis: Long-term outcomes and prognostic predictors. Neurourol Urodyn 2016; 35: 965-969.
95. Tomoe H, Yamashita K. Does repeated hydrodistension with transurethral fulguration for interstitial cystitis with Hunner’s lesion cause bladder contraction? Arab J Urol 2019; 17: 77-81.
96. Hillelsohn JH, Rais-Bahrami S, Friedlander JI, et al. Fulguration for Hunner ulcers: long-term clinical outcomes. J Urol 2012; 188: 2238-2241.
97. Ahmad I, Sarath Krishna N, Meddings RN. Sequential hydrodistension and intravesical instillation of hyaluronic acid under general anaesthesia for treatment of refractory interstitial cystitis: a pilot study. Int Urogynecol J Pelvic Floor Dysfunct 2008; 19: 543-546.
98. Turner KJ, Stewart LH. How do you stretch a bladder? A survey of UK practice, a literature review, and a recommendation of a standard approach. Neurourol Urodyn 2005; 24: 74-76.
99. Platte RO, Parekh M, Minassian VA, Poplawsky D. Spontaneous bladder rupture following cystoscopy with hydrodistention and biopsy in a female patient with interstitial cystitis. Female Pelvic Med Reconstr Surg 2011; 17: 149-152.
100. Zabihi N, Allee T, Maher MG, et al. Bladder necrosis following hydrodistention in patients with interstitial cystitis. J Urol 2007; 177: 149-152; discussion 152.
101. Wang J, Wang Q, Wu Q, et al. Intravesical Botulinum Toxin A Injections for Bladder Pain Syndrome/Interstitial Cystitis: A Systematic Review and Meta-Analysis of Controlled Studies. Med Sci Monit Int Med J Exp Clin Res 2016; 22: 3257-3267.
102. Shim SR, Cho YJ, Shin I-S, Kim JH. Efficacy and safety of botulinum toxin injection for interstitial cystitis/bladder pain syndrome: a systematic review and meta-analysis. Int Urol Nephrol 2016; 48: 1215-1227.
103. Tirumuru S, Al-Kurdi D, Latthe P. Intravesical botulinum toxin A injections in the treatment of painful bladder syndrome/interstitial cystitis: a systematic review. Int Urogynecology J 2010; 21: 1285-1300.
104. Jhang J-F, Kuo H-C. Novel Treatment of Chronic Bladder Pain Syndrome and Other Pelvic Pain Disorders by Onabotulinumtoxin A Injection. Toxins 2015; 7: 2232-2250.
105. Pinto R, Lopes T, Silva J, et al. Persistent therapeutic effect of repeated injections of onabotulinum toxin a in refractory bladder pain syndrome/interstitial cystitis. J Urol 2013; 189: 548-553.
106. Shie J-H, Liu H-T, Wang Y-S, Kuo H-C. Immunohistochemical evidence suggests repeated intravesical application of botulinum toxin A injections may improve treatment efficacy of interstitial cystitis/bladder pain syndrome. BJU Int 2013; 111: 638-646.
107. Lee C-L, Kuo H-C. Long-Term Efficacy and Safety of Repeated Intravescial OnabotulinumtoxinA Injections Plus Hydrodistention in the Treatment of Interstitial Cystitis/Bladder Pain Syndrome. Toxins 2015; 7: 4283-4293.
108. Lee HY, Doo SW, Yang WJ, et al. Efficacy and Safety of Noninvasive Intravesical Instillation of Onabotulinum Toxin-A for Overactive Bladder and Interstitial Cystitis/Bladder Pain Syndrome: Systematic Review and Meta-analysis. Urology 2019; 125: 50-57.
109. Rosa VL, Platania A, Ciebiera M, et al. A comparison of sacral neuromodulation vs. transvaginal electrical stimulation for the treatment of refractory overactive bladder: The impact on quality of life, body image, sexual function, and emotional well-being. Prz Menopauzalny 2019; 18: 89-93.
110. Wang J, Chen Y, Chen J, et al. Sacral Neuromodulation for Refractory Bladder Pain Syndrome/Interstitial Cystitis: a Global Systematic Review and Meta-analysis. Sci Rep 2017; 7: 11031.
111. Gajewski JB, Al-Zahrani AA. The long-term efficacy of sacral neuromodulation in the management of intractable cases of bladder pain syndrome: 14 years of experience in one centre. BJU Int 2011; 107: 1258-1264.
112. Powell CR, Kreder KJ. Long-term outcomes of urgency-frequency syndrome due to painful bladder syndrome treated with sacral neuromodulation and analysis of failures. J Urol 2010; 183: 173-176.
113. Kim HJ, Lee JS, Cho WJ, et al. Efficacy and safety of augmentation ileocystoplasty combined with supratrigonal cystectomy for the treatment of refractory bladder pain syndrome/interstitial cystitis with Hunner’s lesion. Int J Urol Off J Jpn Urol Assoc 2014; 21 (Suppl 1): 69-73.
114. Hanno P. Editorial comment to Efficacy and safety of augmentation ileocystoplasty combined with supratrigonal cystectomy for the treatment of refractory bladder pain syndrome/interstitial cystitis with Hunner’s lesion. Int J Urol Off J Jpn Urol Assoc 2014; 21 (Suppl 1): 74.
115. Andersen AV, Granlund P, Schultz A, et al. Long-term experience with surgical treatment of selected patients with bladder pain syndrome/interstitial cystitis. Scand J Urol Nephrol 2012; 46: 284-289.
116. Rössberger J, Fall M, Jonsson O, Peeker R. Long-term results of reconstructive surgery in patients with bladder pain syndrome/interstitial cystitis: subtyping is imperative. Urology 2007; 70: 638-642.
Copyright: © 2020 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.