1 s2.0 S187952261200053X Main

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Urological Science 23 (2012) 70e77

Contents lists available at SciVerse ScienceDirect

Urological Science
journal homepage: www.urol-sci.com

Mini review

Intravesical therapy for lower urinary tract symptomsq CME


Credits

Jayabalan Nirmal a, Yao-Chi Chuang b, *, Pradeep Tyagi a, Michael B. Chancellor a


a
Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, USA
b
Department of Urology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan

a r t i c l e i n f o a b s t r a c t

Article history: Intravesical (local) therapy for diseases emanating from the bladder is similar to the commonplace
Received 11 September 2011 management of ocular diseases using eyedrops. The existing intravesical therapy involves administering
Received in revised form dosage forms directly into the bladder (through a catheter). This mode of treatment is routine in the
27 October 2011
bladder cancer by instillation of bacillus CalmetteeGuérin to provoke the body’s own natural defenses
Accepted 6 December 2011
Available online 24 August 2012
against invasion by proliferating cancer cells in the bladder. Although convenient, the conventional
routes of administration for treating diseases such as interstitial cystitis/painful bladder syndrome (IC/
PBS) and overactive bladder (OAB) are less acceptable to patients owing to side effects, which may
Keywords:
cystitis
further exacerbate the disease symptoms. Intravesical therapy has demonstrated varying degrees of
intravesical therapy efficacy and safety in treating IC/PBS and OAB. In recent years, intravesical delivery of biotechnological
overactive bladder products including neurotoxins using a liposome platform demonstrated immense promise for lower
urinary tract symptoms. This review considers the current status of intravesical therapy in lower urinary
tract diseases with special attention to novel drug-delivery systems.
Copyright Ó 2012, Taiwan Urological Association. Published by Elsevier Taiwan LLC.
Open access under CC BY-NC-ND license.

1. Introduction intravesical drug delivery (IDD) with respect to specific diseases


and novel drug-delivery systems.
Intravesical therapy/drug delivery is widely used as a first-line
treatment for delaying or preventing the recurrence of bladder
2. LUTS
cancer.1,2 Given the success of this approach in oncology, it would
be wise to apply the lessons learned over the decades in treating
2.1. Interstitial cystitis/painful bladder syndrome
bladder cancer to improve the treatment of lower urinary tract
(LUT) and its symptoms (LUTS). The advertising slogan “applying
Interstitial cystitis/painful bladder syndrome (IC/PBS) is
the medicine directly where it hurts,” heard often in mass media for
a chronic disease characterized by suprapubic/bladder discomfort.
over-the-counter drugs, is apt for promoting wider acceptance of
It is accompanied by urinary frequency, urgency, or nocturia, in the
this line of therapy for LUTS. Instillation of drugs in the bladder
absence of infection or other pathological conditions. Little is
provides a high concentration of drugs locally at the disease site in
known about the pathogenesis of IC; however, it was proposed that
the bladder without an increase in systemic levels, which can
a dysfunctional epithelium allows the transepithelial migration of
explain the low risk of systemic side effects. However, drug delivery
solutes, such as potassium, which can depolarize subepithelial
to bladder tissues by the intravesical routes is constrained by the
afferent nerves and provoke sensory symptoms. In addition,
impermeability of urothelial cells, a short duration of action, and
localization of a high number of activated mast cells in the bladder
the need for frequent administration.3e5 The lining of the urinary
and dysfunction of the superficial layer of the extracellular matrix,
bladder has the most impermeable barrier in the human body. The
the glycosaminoglycan (GAG) layer, were demonstrated to be
following review of the literature describes the status of
related to IC.6,7 The result of these potential etiologies involves
neurogenic inflammation, primary afferent nerve overactivity, and
central nervous system sensitization, all of which interact to
perpetuate pain. Pain-sensing C fibers are located within the
* Corresponding author. Department of Urology, Kaohsiung Chang Gung
uroepithelium and submucosa of the bladder and can be activated
Memorial Hospital, Chang Gung University College of Medicine, 123 Ta-Pei Road,
Niao-Song, Kaohsiung 833, Taiwan.
by either a GAG layer deficiency or release of histamine via mast
E-mail address: chuang82@ms26.hinet.net (Y.-C. Chuang). cells. Because of the multifactorial nature of the disease, improved
q There are 2 CME questions based on this article. therapeutic outcomes can be achieved using multimodal treatment

1879-5226 Copyright Ó 2012, Taiwan Urological Association. Published by Elsevier Taiwan LLC. Open access under CC BY-NC-ND license.
http://dx.doi.org/10.1016/j.urols.2012.07.005
J. Nirmal et al. / Urological Science 23 (2012) 70e77 71

through pharmacological and nonpharmacological approaches increases side effects. IDD involves the direct administration of
acting via different mechanisms.8,9 bioactive agents into the urinary bladder through a catheter (Fig. 1).
The anatomy of the urinary bladder allows relatively uncompli-
2.2. Overactive bladder cated access and manipulation with a catheter, and hence localized
treatment options have enormous potential. Delivery of a thera-
Overactive bladder (OAB) is defined as a symptom complex peutic agent directly into the bladder greatly improves the expo-
comprised of urinary urgency, with or without urgency inconti- sure of the affected bladder lining to the agent, and this is of even
nence, usually with frequency and nocturia.10 There are four main greater significance in the case of drug-resistant targets that
hypotheses to explain the pathophysiological basis for OAB. The require a much higher dose of the drug.17,18
neurogenic hypothesis is based on observations that altered neural
control induces bladder overactivity by altering sensory activity, 3.2. Constraints to intravesical delivery
reducing inhibition, or inappropriately triggering the voiding reflex
and increasing the efferent drive.11 The myogenic hypothesis states IDD is associated with some intrinsic limitations. Periodic
that myocytes from the urinary bladder of patients have increased voiding of urine washes out drug solutions instilled in the bladder.
spontaneous activity that allows excitation to propagate widely This greatly reduces the residence time of the drug in the bladder
through the bladder wall.12 The basis for the peripheral autonomy and hence requires frequent dosing. A further complication of IDD
theory is that cellular interactions in the LUT determine the blad- is the progressive dilution of the instilled drug solution due to
der’s functional state; both spontaneous excitation and wider filling up of urine in the bladder. To overcome this problem, strat-
propagation can result from activity in various cell types such as egies such as emptying the bladder before drug instillation,19
muscle, interstitial, urothelial, and neuronal cells.13 The afferent suppressing the rate of urine production by the kidneys, and
hypothesis focuses on the sensory basis of OAB, suggesting that regulating fluid intake before and after drug administration can be
alterations in the central transfer or sensory transduction leads to utilized. In addition, the drug’s molecular weight, ionization,
increased afferent “noise,” and therefore, an increased awareness of concentration, and exposure time of treatment were proposed as
bladder filling.14 In OAB, the release of acetylcholine from urothe- major factors for determining the rate of transurothelial drug
lium during the storage phase of micturition can activate musca- absorption. An ideal intravesical agent was postulated to have
rinic receptors in the urothelium. Activated muscarinic receptors a molecular weight of >200 amu, negligible ionization between pH
trigger the release of urothelial ATP and nitric oxide, which results 6 and 7, and a partition coefficient in the critical ranges of either
in the activation of the afferent pathway. Thus, blockade of uro- approximately 0.4e0.2 or 7.5e8.0. Recent phase III randomized
thelial muscarinic receptors could indirectly act to reduce afferent clinical trials for intravesical mitomycin demonstrated that elimi-
nerve activation and therefore decrease OAB symptoms. Oral nating the residual volume, overnight fasting, doubling the mito-
antimuscarinics are the mainstay of pharmaceutical management mycin concentration to 40 mg in 20 mL, and urinary alkalinization
of OAB, competitively inhibiting either all muscarinic receptors using oral bicarbonate resulted in a doubling of the durable tumor-
(oxybutynin) or selectively the M3 receptor (darifenacin and free rate at 5 years.20 As is the case with intravesical mitomycin, the
solifenacin).15,16 acidic urinary pH is also a limiting factor in intravesical epirubicin
(EPI) and doxorubicin (DOX) therapy.21 Another major determinant
3. Intravesical delivery for LUTS in the therapeutic success of intravesical therapy is the degree of
drug lipophilicity, which allows penetration through cell
3.1. Rationale for intravesical delivery membranes. Paclitaxel, a highly lipophilic compound, achieves
greater intraurothelial concentrations than either mitomycin or
Although a large number of therapeutic agents are available for DOX.22
treating bladder diseases, most of them are administered through
the per-oral route and hence fail to have a sufficient effect at the 3.3. Bloodeurine barrier
disease site. Systemic administration of these therapeutic agents
requires administration of large doses, but only a small fraction To elicit a therapeutic effect following intravesical delivery into
actually reaches the bladder, and the undesired biodistribution the urinary bladder, the drug has to cross a watertight barrier

Fig. 1. Schematic representation describing approaches for intravesical delivery. EMDA ¼ electromotive drug administration; GAG ¼ glycosaminoglycan.
72 J. Nirmal et al. / Urological Science 23 (2012) 70e77

between the blood and urine. The bloodeurine barrier, called the 4.2. Dimethyl sulfoxide
urothelium, is the tightest and most impermeable barrier in the
human body (Fig. 1).23 The urothelium, responsible for a multitude A 50% aqueous solution of dimethyl sulfoxide (DMSO; RIMSO-
of physiological activities, is not only effective in blocking the entry 50) is widely used to treat IC because of its analgesic, anti-
of urine contents but is also equally effective in blocking the entry inflammatory, and muscle-relaxant properties.34 Both early and
of instilled drugs.3,4 It sits at the interface between the urine and classic forms of IC respond to RIMSO-50, and it also has an influence
underlying connective tissue where it forms a barrier preventing on conduction and neurotransmission in sensory nerves.35e38
the unregulated exchange of solutes, ions, and toxic metabolites. RIMSO-50 is accepted as being moderately effective and safe for
The permeability to substances such as water, ammonia, and urea, relieving symptoms or urgency and pain in IC.39 Intravesical
which normally cross membranes relatively rapidly, is extremely instillation of DMSO in animal models demonstrated direct corre-
low in the urothelium.3,4 It has exceptionally high transepithelial lations of the drug concentration and contact time with the bladder
resistance ranging from 10,000 to >75,000 Ucm2 owing to para- with the anti-inflammatory effects without a change in the bladder
cellular resistance of tight junctions pooled with apical plasma capacity or systemic toxicity. DMSO can penetrate tissues without
membrane transcellular resistance.24,25 Urothelial tight junctions damaging them and is used to enhance the transport of chemo-
are comprised of a dense network of cytoplasmic proteins (zonula therapeutic drugs such as cisplatin, DOX, and pirarubicin into
occludens-1), cytoskeletal elements, and transmembrane proteins bladder tumors.40
(occludin and claudins) and have the highest recorded paracellular
resistance of all epithelia measured to date.26 In addition, the apical 4.3. Drug cocktails
plasma membrane of the urothelium contains specific proteins,
uroplakins, which account for the transcellular resistance. Electron Because of the multifactorial nature of IC, combination therapy
microscopic analysis of umbrella cells lying at the luminal surface of utilizing drugs with different mechanisms of action is a reasonable
the urothelium showed a hexagonal arrangement of uroplakins, approach. Parsons et al demonstrated the effect of mixing different
where six subunits of each particle are tightly joined together to ratios of heparin with lidocaine in drug cocktails for treating
form a complete hexagonal ring, with lipids contained in the patients with IC. The treatment response evaluated at different
central cavity.27 The low permeability of the urothelial barrier is time points showed a reduction in pain and urinary urgency in
believed to result from the peculiar protein array and tight junc- most patients. Relief from symptoms 2 weeks after treatment
tions between umbrella cells. The urothelial barrier restricts the suggested that the efficacy was retained beyond the duration of the
movement of drugs after intravesical administration and also local anesthetic activity of lidocaine.41
restricts the action of the active drug fraction in the urine. Hence,
many drugs fail to reach the bladder at desired therapeutic levels 4.4. Vanilloids
and ultimately lack pharmacological effects.27,28
Compounds related to capsaicin and an ultrapotent analog,
4. Intravesical therapeutics for LUTS resiniferatoxin (RTX), collectively referred to as vanilloids, interact
at a specific membrane recognition site (the vanilloid receptor)
4.1. GAG analogs expressed almost exclusively by primary sensory neurons involved
in nociception and neurogenic inflammation. Interest in using
Intravesical delivery of GAG analogs such as hyaluronic acid vanilloids for intravesical therapy in IC is based on the putative
(HA), heparin, and chondroitin sulfate (CS), restores the barrier mechanism of action, as well as on clinical and nonclinical obser-
function lost due to epithelial dysfunction in interstitial cystitis. HA vations. Suprapubic pain, the major symptom and cause of
itself is an important component of the urothelium, and sodium morbidity in patients with IC, is believed to be primarily mediated
hyaluronate is used to replenish bladder GAG when treating IC. HA by afferent type-C sensory neurons in the bladder. C fibers may also
inhibits leukocyte aggregation and migration, and adherence of become mechanosensitive, producing increased urinary frequency
immune complexes to polymorphonuclear cells.29,30 From a clinical and urgency in IC. Vanilloids are used to downregulate sensory
study involving 48 patients, it was evident that intravesical heparin nerves in the IC bladder, thereby mitigating the pain response. RTX
(104 units/10 mL sterile water, three times a week for 3 months) is an ultrapotent analog of capsaicin that may have improved
controlled symptoms of IC with continued improvement even after tolerability and enhanced efficacy compared with capsaicin. RTX
1 year of therapy.31 When used in conjugation with hydro- resulted in a more favorable ratio of desensitization to initial
distention therapy, both HA and heparin prolonged the beneficial excitation compared with capsaicin in animal models of pain
effects of hydrodistention in patients with a small functional behavior.42,43 In a rat model, intravesical RTX (0.01 mM) reversibly
bladder capacity. HA was found to be more effective compared with desensitized bladder afferents for 3 weeks, and a 0.1-mM dose
heparin. In contrast to no improvement in the control group, there resulted in sustained desensitization for at least 4 weeks. In
was a significantly higher rate of improvement at 6 and 9 months in patients with neurogenic bladder problem, intravesical RTX and
the HA group relative to the heparin group (50% vs. 20%, p < 0.05). capsaicin were comparably effective; however, RTX is better toler-
Improvements in the number of times voiding per day (e1.8  2.5, ated. A randomized, placebo-controlled study utilizing 18 patients
p < 0.01), a visual analog scale (e0.9  1.1, p < 0.01), and bladder with hypersensitive bladder disorders showed that a single intra-
capacity (16  18 mL, p < 0.01) were more significant in the HA vesical administration of RTX (0.01 mM) significantly improved
group at 9 months relative to no improvement in the heparin pain, urinary frequency, and nocturia at 1 month, and urinary
group.32 Hauser et al investigated CS binding to the bladder uro- frequency at 3 months following treatment.44
thelium in a mouse model of urothelial acid damage. CS was labeled
with Texas Red, and the efficacy of restoring the barrier function 4.5. Antimuscarinics
was determined by intravesically instilling 86Rb, a potassium ion
mimetic, through the urothelium into the bloodstream. The results Recent reports showed that intravesically administered anti-
demonstrated that CS preferentially binds to damaged urothelium cholinergic agents, apart from blocking muscarinic receptors in the
and restores the impermeability barrier. Maximum efficacy was bladder, may also act by blocking the bladder-cooling reflex
achieved with a dose of 400 mg CS per instillation.33 mediated by C fibers with an incomplete neurogenic lesion and
J. Nirmal et al. / Urological Science 23 (2012) 70e77 73

neurogenic detrusor overactivity (NDO).45 Intravesical oxybutynin NGF levels in bladder afferent pathways, and is able to normalize
(1.25 mg/5 mL, twice a day) was shown to be a relatively safe and bladder/urethral function in spinal cordeinjured rats.54 Similar
effective therapeutic option for children with neurogenic bladders clinical outcome was reported with systemic administration of
who experienced intolerable side effects or were unresponsive to monoclonal human NGF antibodies (tanezumab) in IC patients.55
oral antimuscranics.46 Intravesical delivery of oxybutynin was Although the systemic administration of antibodies was able to
proven to be suitable for patients who have an OAB and suffer from reduce pain, it encumbered patients with safety concerns such as
side effects of the metabolite, N-desethyl-oxybutynin, following paresthesia, hypoesthesia, and arthralgia. The results revealed that
per-oral administration.47,48 NGF is a viable drug target, and the fact that the intravesical route
can avoid systemic side effects prompted alternative approaches for
4.6. Neurotoxin targeting NGF. As it is likely that the major pathology of OAB is
driven by NGF, targeting the intracellular synthesis of NGF in the
The use of botulinum neurotoxin A (BoNT-A) to treat LUT bladder is a promising therapeutic alternative. The proof of the
dysfunction has expanded in recent years, and the off-license usage concept of this approach was demonstrated by suppression of
list includes PBS, NDO, idiopathic detrusor overactivity (IDO), and bladder overactivity by the local instillation of an antisense agent
LUTS resulting from bladder outflow obstruction. There are two against NGF based on the PNA backbone.52,56
commonly used preparations of BoNT-A: BOTOX (onabotuli- One mechanism by which bladder pain is induced was postu-
numtoxinA) and Dysport (abobotulinumtoxinA). BoNT-A, the most lated to involve chronic tissue inflammation that can lead to
popularly used BoNT, acts by cleaving the soluble N-ethyl- functional changes in C-fiber afferents. It was previously reported
maleimide-sensitive fusion attachment protein receptor (SNARE) that endogenous enkephalins are expressed in bladder afferent and
protein, SNAP-25, and inhibiting the release of various neuro- efferent pathways to inhibit micturition.57,58 Yokoyama et al59
transmitters at the presynaptic vesicle by binding to the synaptic examined the efficacy of targeted and localized expression of
vesicle protein, SV2, during neurotransmitter exocytosis. BoNT-A enkephalin in afferent nerves, which innervate the bladder, using
was successfully used to treat IC and OAB through a cystoscopi- herpes simplex virus (HSV) vectorebased gene transfer in bladder
cally guided injection. However, instillation of this high molecular pain and hyperactivity. A replication-defective HSV-1 vector, SHPE,
weight toxin has to be localized because any systemic absorption engineered to carry the human preproenkephalin (hPPE) transgene
can prove fatal. Pretreatment of the urothelium with protamine to the bladder and its sensory nerves was used. The results of this
sulfate (PS) to improve its permeability to BoNT-A was attempted in study indicated that HSV vectors injected into the bladder wall
rats.49,50 The cationic PS interacts with the anionic GAG layer, were transported through bladder afferent pathways and
leading to a slight increase in permeability of the urothelium.51 expressed hPPE. After SHPE bladder inoculation, the bladder
Instillation of neurotoxins into the bladder is an accepted hyperactivity induced by nociceptive stimuli (i.e., capsaicin) was
approach to achieve chemical neuromodulation of afferent neuro- reduced via naloxone-dependent opioid mechanisms. In addition
transmission underlying IC and OAB. However, neuromodulation to this, nociceptive freezing behavior induced by intravesical
by instilled neurotoxins is suboptimal, possibly due to protein capsaicin was also suppressed in association with increased bladder
degradation by proteases and proteinases in the urine, dilution by capacity after SHPE treatment. These data illustrate that enkephalin
urine, or poor uptake of the BoNT solution into the urothelium. gene therapy for bladder pain response is feasible and achieves
a physiological decrease in bladder irritative responses, and gene
4.7. Antisense oligodeoxynucleotide and gene therapeutics transfer using replication-defective HSV vectors may offer new
hope for treating refractory IC. Gene therapy strategies for OAB may
Until recently, there has been little research carried out on gene include suppression of bladder muscle activity or neural pathways
and antisense oligodeoxynucleotide (ODN) therapy for the urinary that trigger the micturition reflex. Kþ channel gene therapy for
tract. Most of the previous work focused on urinary tract malig- urinary incontinence is under development (by Christ et al).60
nancies such as bladder and prostate cancers. However, this situ- Intravesical instillation of naked pcDNA/hSlo (complementary
ation has changed radically in the past few years. Interest in gene DNA) causes overexpression of bladder Kþ channels that may
therapy for urinary tract dysfunction is due to the potential market inhibit OAB.60
size that urological diseases represent as well as the fact that the
LUT is ideally suited for minimally invasive molecular medicine 5. Nano approaches to intravesical delivery
therapy owing to a lower risk of systemic toxicity. Overexpression
of nerve growth factor (NGF) was linked to inflammation of the Because of the limited permeability of the bladder wall and
bladder and to the pathogenesis of IC. Tyagi et al52 investigated the undesired side effects of chemical enhancers, the IDD approach is
potential of intravesical instillation of antisense ODNs for localized amenable to modulating the release and absorption characteristics
reduction of NGF expression in the urinary bladder. Antisense ODNs of instilled drugs by coupling them to novel carriers such as lipo-
were replaced with antisense peptide nucleic acid (PNA) to improve somes, microspheres, nanoparticles (NPs), and so forth. Strategies
the stability, and tethering with a TAT peptide sequence was used to involving NPs and in situ gels for IDD are depicted in Fig. 1.
facilitate penetration into the bladder. Cystometrograms (CMGs)
exhibited significantly reduced bladder contraction frequency in 5.1. Vesicular systems: liposomes
the antisense PNA-treated rats compared to rats treated with the
control. NGF immunoreactivity was also reduced in the urothelium Liposomes are lipid vesicles composed of synthetic or natural
of antisense PNA-treated bladders. These findings demonstrated phospholipids that self-assemble to form bilayers surrounding an
the feasibility of using TATePNA conjugates for intravesical anti- aqueous core. They can incorporate small drug molecules, both
sense therapy. hydrophilic and hydrophobic, macromolecules, and even plas-
In our previous studies, we also showed that overexpression of mids, and show greater uptake into cells via endocytosis.61
NGF in the bladder and bladder afferent pathways is directly Intravesical liposomes, even without drugs, have therapeutic
involved in the emergence of hyperexcitability of C fibers, leading effects on IC patients, mainly due to their ability to form
to the pathology of OAB.53 Based on research findings by our group, a protective lipid film on the urothelial surface. Fraser et al62
the intrathecal application of NGF antibodies results in reduced examined the effect of intravesically administered liposomes of
74 J. Nirmal et al. / Urological Science 23 (2012) 70e77

L-a-phosphatidylcholine:cholesterol at ratio of 2:1 in a rat model confirmed the liposome-mediated transport of BoNT into the uro-
of hyperactive bladder. PS was used to mimic the interstitial thelium.67 Animal studies indicate that liposomes can restrict BoNT
cystitis state, followed by a KCl and acetic acid infusion to serve as delivery to the detrusor muscle and avoid the risk of retention and
an irritant. The cystometrographic results showed that the incomplete bladder emptying.
hyperactivity induced by PS/KCl was partially reversed by treat-
ment with liposomes/KCl, showing a significant change in the 5.2. Polymeric NPs
intercontraction interval (ICI) from 15.8  1.4 minutes with the
control KCl solution to 2.7  1.0 minutes with PS/KCl and finally an Polymer-based NPs offer a variety of compositions, all of which
increase to 4.4  1.2 minutes with liposome treatment. Similarly, provide a variable degree of drug loading and release by changing
the irritant effect induced by acetic acid was reduced by liposome the parameters of the chemical nature and cross-linking reactions
treatment, with ICI values changing from 15.3  2.2 minutes with involved. The encapsulated drug is released in a controlled manner
saline to 6.7  1.5 minutes with the liposome formulation. The by either diffusion, erosion, or a combination of both.68 Although
possible mechanism of action of liposomes is the formation of a large number of synthetic polymers can be used to form NPs, those
a lipid film on the urothelium that protects it from penetration by which are biocompatible and biodegradable are used for drug-
irritants. Additionally, liposomes can augment the barrier prop- delivery purposes. Chang et al69 studied poly(ethyl-2-
erties of the urothelium by stabilizing neuro membranes of cyanoacrylate) epirubicin nanoparticles (EPI-NPs) in bladder
damaged nerves and reduce their hyperexcitability. In addition, cancer cell lines (T24 and RT4). The NPs greatly improved the
liposomes composed of phospholipids can exert anti- penetration of EPI into the bladder wall, as commercial aqueous
inflammatory effects by initiating local lipid signaling and formulations of EPI have very low efficacy. Adhesion of the NPs to
affecting mast cell activity. Encouraged by the efficacy of empty bladder samples was evaluated using a diffusion cell. Histological
liposomes as a therapeutic agent for intravesical therapy of IC/PBS, staining showed that the formulation caused no structural damage
Chuang et al recently published information on the clinical safety to the urothelium. Tissue analyses also showed higher penetration
and efficacy of liposomes in IC/PBS patients.63 In an open-labeled and accumulation of the EPI-NP formulation in tissues compared
prospective study of 24 IC/PBS patients, the effect of intravesical with those of the free drug. A large number of polymers such as
liposomes was compared with oral pentosan polysulfate sodium. polycaprolactone, polyacrylates, polylactic acid, polyglycolides can
Statistically significant decrease in pain, urgency, and the be formulated into nanocarriers for intravesical applications.70
O’LearyeSant symptom index were observed in the liposome
group. None of the patients reported urinary incontinence, 5.3. Magnetic NPs
retention, or infection due to liposome instillation. Intravesical
instillation of liposomes was found to be safe for IC with potential NPs developed using magnetic compounds have immense
improvement after one course of therapy for up to 8 weeks. potential as both targeted drug carriers and for diagnostic
Intravesical liposomes appear to be a promising new treatment for imaging.71 Magnetic NPs can be used as contrast agents to visualize
IC/PBS, and future large-scale placebo-controlled studies are diseased regions of the bladder, by targeting specific regions of the
needed to verify results of the pilot study. bladder using a magnetic field to localize drug-loaded magnetic
A potent immunosuppressive effect of tacrolimus encouraged particles.72,73 The core of these NPs can be coated with an organic
its topical application to achieve a local anti-inflammatory effect. or inorganic shell to allow drug adsorption or for ligand attachment
However, its poor aqueous solubility presents challenges in to their surface. However, the size and properties of these particles
formulating biocompatible instillations. Liposomal tacrolimus need to be optimized for intravesical delivery. A recent study by
significantly inhibited cyclophosphamide-induced inflammatory Leakakos et al74 used magnetic targeted carriers (MTCs, with
cystitis through modulating interleukin-2, prostaglandin E2, and particle sizes of approximately 0.5e5 mm) to deliver the anticancer
prostaglandin E receptor 4(EP4) function. These findings support drug, DOX, into the bladder wall. The MTCs contained an iron
the investigation of local tacrolimus in cases of IC refractory to component to allow targeting by a magnet and an activated carbon
conventional therapy.64 A study conducted by the authors’ labo- component to adsorb the drug. These particles were evaluated in
ratory evaluated the potential of liposomes as a vehicle for the transitional cell bladder carcinomas in swine bladder, and the study
intravesical delivery of the neurotoxin, capsaicin, for treating IC. results showed greater accumulation of magnetic particles at the
Capsaicin is a water-insoluble hydrophobic drug, so its instillation region of the bladder closest to the external magnet. Thus, magnetic
requires the use of ethanolic saline to enhance urothelial penetra- guiding of such particles can provide site-specific intravesical
tion, but this can damage bladder tissues. The efficacy of liposome- delivery of drugs.
encapsulated capsaicin was evaluated by measuring the micturition
reflex in normal rats under urethane anesthesia.65 The CMG trac- 5.4. Dendrimers
ings showed that liposomes were able to deliver capsaicin with an
efficacy similar to ethanolic saline. Tissue histological and Dendrimers are core-shell macromolecules made up of highly
morphological studies, however, revealed that toxicity to the organized layers of monomers that make up branches surrounding
bladder was drastically reduced. In the context of toxins instilled in a core. Successive branches (called generations) can be added to
the bladder, fat-soluble neurotoxins such as capsaicin can be inte- increase the molecular size and number of available surface
grated into the phospholipid bilayer, and water-soluble neurotoxins groups.75 Dendrimers have a narrow size distribution, highly
such as botulinum can be protected within the aqueous compart- ordered structures, availability of a large number of functional
ment of liposomes.66 Liposomes can co-opt the native vesicular groups for attachment of targeting ligands and drug molecules, and
traffic ongoing in the bladder, necessary for periodic expansion of a high degree of control over drug-release properties.76e79
the bladder lining during urine storage phases, which may provide Recently, François et al evaluated the possibility of utilizing 5-
a favorable environment for drug delivery. Liposomes can mimic amino levulinic acid (5-ALA)eloaded dendrimers to decrease the
these vesicles and thereby aid in improving the delivery of cargo photobleaching of the fluorophore, protoporphyrin IX (PpIX), and
across the blood-urine barrier. Encapsulation of BoNT inside lipo- to improve the visualization and specificity during cystoscopy.
somes protected it from urinary degradation without compro- Intravesical administration of 5-ALA dendrimers in rats resulted in
mising the efficacy in a rat model. Immunohistochemical detection PpIX synthesis which was much more selective toward the tumor
J. Nirmal et al. / Urological Science 23 (2012) 70e77 75

as opposed to 5-ALA or hexyl derivative ALA (h-ALA). In vitro coat and protect damaged tissue surfaces or even to increase
experiments showed that there was prolonged and sustained PpIX penetration of therapeutic agents into the cell lining.83 Mucoad-
synthesis with reduced photobleaching compared to h-ALA- hesive formulations must meet three basic criteria: the carrier
induced PpIX. These are primary indicators that 5-ALA den- should rapidly adhere to the bladder wall, the carrier must not
drimers can possibly overcome drawbacks of 5-ALA/h-ALA obstruct the flow of urine or any of the normal functions of the
fluorescenceeguided cystoscopy.80 bladder, and it should be able to remain attached to the affected site
even after urine is voided.84 A number of biomolecules, such as
5.5. Nanocrystalline silver chitosan, carbomers, and cellulose derivatives, were identified as
having mucoadhesive properties, among which chitosan is widely
Boucher et al investigated the effect of nanocrystalline silver in used for permeability enhancement of drug solutions through the
experimental bladder inflammation. Nanocrystalline silver or urothelium. Chitosan has many properties advantageous for intra-
phosphate-buffered saline was introduced intravesically into rat vesical delivery, as it is biodegradable, biocompatible, and poly-
bladders for 20 minutes followed by vehicle or PS and lipopoly- cationic and has reactive amine and alcohol groups.85 Derivatives of
saccharide. Nanocrystalline silver was not effective at <1%. Intra- chitosan are used to treat IC using the sulfated form N-sulfonato-
vesical administration of nanocrystalline silver (1%) decreased N,O-carboxymethylchitosan (SNOCC) to encapsulate and transport
urine histamine, bladder tumor necrosis factor-a, and mast cell 5-aminosalicylic acid (5-ASA) into the rat bladder wall. The
activation with no toxic effects. The effects were apparent even inflammation and urinary frequency was found to be reduced using
4 days later and may be useful for IC.81 a combination of 3% SNOCC with 5-ASA.86 The SNOCC coats the wall
of the bladder while chitosan enhances permeation of the anti-
6. Mucoadhesive approaches to intravesical delivery inflammatory agent into the bladder lining. In a recent study,
chitosanethioglycolic acid (TGA) NPs were evaluated as carriers for
6.1. Polymeric hydrogels intravesical delivery. Because of its thiol groups and disulfide
bonds, chitosaneTGA NPs showed greater stability, superior
Major progress in IDD is the use of hydrogels to serve as drug mucoadhesion, and more sustained and more controlled release
depots on the bladder wall. This eliminates the need for repeated than the corresponding unmodified chitosan particles. The adhe-
infusions of drug solutions, in which the drug is mostly washed out sion of prehydrated chitosaneTGA NPs to the urinary bladder
when voiding urine. Large varieties of biocompatible, bioactive mucosa under continuous urine voiding was 14-fold higher
polymeric hydrogels are available which can remain attached to the compared with unmodified chitosan NPs. Release studies indicated
bladder wall even after urine voiding. A recent study by Tyagi et al82 more sustained trimethoprim release from covalently cross-linked
involved in vivo evaluation of a fluorescein isothiocyanate (FITC)- particles compared with unmodified chitosanetripolyphosphate
and misoprostol-loaded thermosensitive polymeric hydrogel in NPs over a period of 3 hours in artificial urine at 37  C. The study
a rat model. Thermosensitive polymers can be used to first inject revealed that the chitosaneTGA IDD system may prove to be useful
the hydrogel in its liquid form, which then forms a gel in situ inside carriers for local drug applications in the urinary bladder, which
the bladder cavity at an elevated body temperature. The triblock co- increases the residence time of the drug at the target site and
polymer, poly(ethylene glycol)epoly[lactic acid-co-glycolic acid]e enables sustainable delivery for an extended period.87
poly(ethylene glycol), was modified to make it more hydrophobic
in nature so as to enable it to withstand urine components. Results
of the study showed that the injected gel did not obstruct urine 7. Electromotive drug administration
outflow, proving that it attached itself as a thin layer onto the
bladder wall. The prolonged excretion (over 24 hours) of FITC in the The desire to increase the penetration of drugs instilled into the
urine suggested that the hydrogel did not get washed off during bladder has also tempted exploration of electromotive drug
urine voiding. Another aspect of this gel is its reversible gelling administration (EMDA). In a recent study, patients suffering from
property, which enables easy removal of the gel from the bladder urge syndrome with and without urge incontinence (25.6% OAB
by simple washing with saline at room temperature. Efficacy wet, 20.0% OAB dry, and 54.4% mixed urinary incontinence) and
studies showed that the misoprostol trapped in the hydrogel non responsive to oral anticholinergic drugs underwent EMDA
maintained its biological activity and successfully reduced incon- therapy performed once in every 4 weeks for a period of 3 months.
tinence in rats. Although there have been few studies on such EMDA significantly improved urodynamic parameters, the quality
systems, there are a large number of potential polymeric gel of life as evaluated with the King’s health questionnaire, and pad
systems that can be explored. Translating this method for use in the usage in patients with urge syndrome and therapy-resistant IDO as
human bladder involves a number of unique challenges such as (1) evident from a micturition chart over 48 hours.88
the gels need to be designed for the higher capacities of the human
urinary bladder; (2) the gels should form a uniform thin layer inside 8. Conclusions
the bladder at the affected site; (3) the gels should have optimal
contact with the urothelial surface to allow for strong adherence; Future development of novel drug-delivery systems as
and (4) mechanical properties of the gels should also allow them to a delivery platform for intravesical administration of drugs can
resist detachment by shear forces of urine flow and stretching of improve the efficacy and safety of pharmacotherapy for bladder
the bladder wall.18 diseases. The latest developments in the field of nanotechnology
bring this mode of therapy to the forefront as a new hope for
6.2. Mucoadhesive NPs disease management in the LUT.

IDD systems can be made more effective by using mucoadhesive


materials that can attach to the mucous membrane of the urothe- Conflicts of interest statement
lium. This allows the formulation to be retained at the site for
a longer duration and ensures sustained and enhanced contact with Yao-Chi Chuang, Pradeep Tyagi and Michael B. Chancellor are
the mucous layer. Mucoadhesive formulations can also be used to consultants for Lipella Pharmaceuticals Inc.
76 J. Nirmal et al. / Urological Science 23 (2012) 70e77

Source of Funding 33. Hauser PJ, Buethe DA, Califano J, Sofinowski TM, Culkin DJ, Hurst RE. Restoring
barrier function to acid damaged bladder by intravesical chondroitin sulfate.
J Urol 2009;182:2477e82.
None. 34. Parkin J, Shea C, Sant GR. Intravesical dimethyl sulfoxide (DMSO) for interstitial
cystitisda practical approach. Urology 1997;49:105e7.
35. Sant GR. Intravesical 50% dimethyl sulfoxide (RIMSO-50) in treatment of
References interstitial cystitis. Urology 1987;29:17e21.
36. Fowler Jr JE. Prospective study of intravesical dimethyl sulfoxide in treatment
1. Shen Z, Shen T, Wientjes MG, O’Donnell MA, Au JL. Intravesical treatments of of suspected early interstitial cystitis. Urology 1981;18:21e6.
bladder cancer: review. Pharm Res 2008;25:1500e10. 37. Barker SB, Matthews PN, Philip PF, Williams G. Prospective study of intravesical
2. Highley MS, van Oosterom AT, Maes RA, De Bruijn EA. Intravesical drug dimethyl sulphoxide in the treatment of chronic inflammatory bladder disease.
delivery. Pharmacokinetic and clinical considerations. Clin Pharmacokinet Br J Urol 1987;59:142e4.
1999;37:59e73. 38. Perez-Marrero R, Emerson LE, Feltis JT. A controlled study of dimethyl sulfoxide
3. Melicow MM. The urothelium: a battleground for oncogenesis. J Urol 1978;120: in interstitial cystitis. J Urol 1988;140:36e9.
43e7. 39. Sun Y, Chai TC. Effects of dimethyl sulphoxide and heparin on stretch-activated
4. Apodaca G. The uroepithelium: not just a passive barrier. Traffic 2004;5: ATP release by bladder urothelial cells from patients with interstitial cystitis.
117e28. BJU Int 2002;90:381e5.
5. Lu Z, Yeh TK, Tsai M, Au JL, Wientjes MG. Paclitaxel-loaded gelatin nano- 40. Giannantoni A, Di Stasi SM, Chancellor MB, Costantini E, Porena M. New
particles for intravesical bladder cancer therapy. Clin Cancer Res 2004;10: frontiers in intravesical therapies and drug delivery. Eur Urol 2006;50:
7677e84. 1183e93.
6. Parsons JK, Parsons CL. The historical origins of interstitial cystitis. J Urol 41. Parsons CL. Successful downregulation of bladder sensory nerves with
2004;171:20e2. combination of heparin and alkalinized lidocaine in patients with interstitial
7. Parsons CL, Greene RA, Chung M, Stanford EJ, Singh G. Abnormal urinary cystitis. Urology 2005;65:45e8.
potassium metabolism in patients with interstitial cystitis. J Urol 2005;173: 42. Chancellor MB, de Groat WC. Intravesical capsaicin and resiniferatoxin
1182e5. therapy: spicing up the ways to treat the overactive bladder. J Urol
8. Theoharides TC, Sant GR. A pilot open label study of cystoprotek in interstitial 1999;162:3e11.
cystitis. Int J Immunopathol Pharmacol 2005;18:183e8. 43. Byrne DS, Das A, Sedor J, Huang B, Rivas DA, Flood HJ, et al. Effect of intravesical
9. Smaldone MC, Vodovotz Y, Tyagi V, Barclay D, Philips BJ, Yoshimura N, et al. capsaicin and vehicle on bladder integrity control and spinal cord injured rats.
Multiplex analysis of urinary cytokine levels in rat model of J Urol 1998;159:1074e8.
cyclophosphamide-induced cystitis. Urology 2009;73:421e6. 44. Lazzeri M, Beneforti P, Spinelli M, Zanollo A, Barbagli G, Turini D. Intravesical
10. Abrams P, Cardozo L, Fall M, Griffiths D, Rosier P, Ulmsten U, et al. The resiniferatoxin for the treatment of hypersensitive disorder: a randomized
standardisation of terminology of lower urinary tract function: report from the placebo controlled study. J Urol 2000;164:676e9.
Standardisation Sub-committee of the International Continence Society. Neu- 45. Van Meel TD, De Wachter S, Wyndaele JJ. The effect of intravesical oxybutynin
rourol Urodyn 2002;21:167e78. on the ice water test and on electrical perception thresholds in patients with
11. de Groat WC. A neurologic basis for the overactive bladder. Urology 1997;50(6A neurogenic detrusor overactivity. Neurourol Urodyn 2010;29:391e4.
Suppl.):36e52. discussion 53e6. 46. Hayashi A, Saito M, Okada S, Hanada T, Watanabe T, Satoh K, et al. Treatment
12. Brading AF, Turner WH. The unstable bladder: towards a common mechanism. with modified intravesical oxybutynin chloride for neurogenic bladder in
Br J Urol 1994;73:3e8. children. J Pediatr Urol 2007;3:438e42.
13. Drake MJ, Mills IW, Gillespie JI. Model of peripheral autonomous modules and 47. Buyse G, Waldeck K, Verpoorten C, Björk H, Casaer P, Andersson KE. Intra-
a myovesical plexus in normal and overactive bladder function. Lancet vesical oxybutynin for neurogenic bladder dysfunction: less systemic side
2001;358:401e3. effects due to reduced first pass metabolism. J Urol 1998;160:892e6.
14. Andersson KE. Detrusor myocyte activity and afferent signaling. Neurourol 48. Abramov Y, Sand PK. Oxybutynin for treatment of urge urinary incontinence
Urodyn 2010;29:97e106. and overactive bladder: an updated review. Expert Opin Pharmacother
15. Chuang YC, Thomas CA, Tyagi S, Yoshimura N, Tyagi P, Chancellor MB. Human 2004;5:2351e9.
urine with solifenacin intake but not tolterodine or darifenacin intake blocks 49. Vemulakonda VM, Somogyi GT, Kiss S, Salas NA, Boone TB, Smith CP. Inhibitory
detrusor overactivity. Int Urogynecol J Pelvic Floor Dysfunct 2008;19:1353e7. effect of intravesically applied botulinum toxin A in chronic bladder inflam-
16. Henderson E, Drake M. Overactive bladder. Maturitas 2010;66:257e62. mation. J Urol 2005;173:621e4.
17. Kaufman J, Tyagi V, Anthony M, Chancellor MB, Tyagi P. State of the art in 50. Khera M, Somogyi GT, Salas NA, Kiss S, Boone TB, Smith CP. In vivo effects of
intravesical therapy for lower urinary tract symptoms. Rev Urol botulinum toxin A on visceral sensory function in chronic spinal cord-injured
2010;12:e181e9. rats. Urology 2005;66:208e12.
18. GuhaSarkar S, Banerjee R. Intravesical drug delivery: challenges, current status, 51. Tzan CJ, Berg JR, Lewis SA. Mammalian urinary bladder permeability is altered
opportunities and novel strategies. J Control Release 2010;148:147e59. by cationic proteins: modulation by divalent cations. Am J Physiol 1994;267:
19. Gao X, Au JL, Badalament RA, Wientjes MG. Bladder tissue uptake of mitomycin C1013e26.
C during intravesical therapy is linear with drug concentration in urine. Clin 52. Tyagi P, Banerjee R, Basu S, Yoshimura N, Chancellor M, Huang L. Intravesical
Cancer Res 1998;4:139e43. antisense therapy for cystitis using TAT-peptide nucleic acid conjugates. Mol
20. Au JL, Badalament RA, Wientjes MG, Young DC, Warner JA, Venema PL, et al. Pharm 2006;3:398e406.
Methods to improve efficacy of intravesical mitomycin C: results of 53. Yoshimura N, Bennett NE, Hayashi Y, Ogawa T, Nishizawa O, Chancellor MB,
a randomized phase III trial. J Natl Cancer Inst 2001;93:597e604. et al. Bladder overactivity and hyperexcitability of bladder afferent neurons
21. Au JL, Jang SH, Wientjes MG. Clinical aspects of drug delivery to tumors. after intrathecal delivery of nerve growth factor in rats. J Neurosci 2006;26:
J Control Release 2002;78:81e95. 10847e55.
22. Song D, Wientjes MG, Au JL. Bladder tissue pharmacokinetics of intravesical 54. Seki S, Sasaki K, Igawa Y, Nishizawa O, Chancellor MB, De Groat WC, et al.
taxol. Cancer Chemother Pharmacol 1997;40:285e92. Suppression of detrusor-sphincter dyssynergia by immunoneutralization of
23. Parsons CL, Boychuk D, Jones S, Hurst R, Callahan H. Bladder surface glycos- nerve growth factor in lumbosacral spinal cord in spinal cord injured rats. J Urol
aminoglycans: an epithelial permeability barrier. J Urol 1990;143:139e42. 2004;171:478e82.
24. Lewis SA, Diamond JM. Naþ transport by rabbit urinary bladder, a tight 55. Evans RJ, Moldwin RM, Cossons N, Darekar A, Mills IW, Scholfield D. Proof of
epithelium. J Membr Biol 1976;28:1e40. concept trial of tanezumab for the treatment of symptoms associated with
25. Negrete HO, Lavelle JP, Berg J, Lewis SA, Zeidel ML. Permeability properties of interstitial cystitis. J Urol 2011;185:1716e21.
the intact mammalian bladder epithelium. Am J Physiol 1996;271:F886e94. 56. Tyagi V, Sugino Y, Yoshimura N, Chancellor M, Tyagi P, Oak R. Liposomes
26. Khandelwal P, Abraham SN, Apodaca G. Cell biology and physiology of the assisted bladder uptake of NGF antisense oligonucleotides ameliorate symp-
uroepithelium. Am J Physiol Renal Physiol 2009;297:F1477e501. toms of detrusor overactivity in rat model. J Urol 2011;185(Suppl 4):e318.
27. Min G, Zhou G, Schapira M, Sun TT, Kong XP. Structural basis of urothelial 57. Glazer EJ, Basbaum AI. Leucine enkephalin: localization in and axoplasmic trans-
permeability barrier function as revealed by Cryo-EM studies of the 16 nm port by sacral parasympathetic preganglionic neurons. Science 1980;208:1479e81.
uroplakin particle. J Cell Sci 2003;116:4087e94. 58. de Groat WC. Neuropeptides in pelvic afferent pathways. Experientia
28. Tammela T, Wein AJ, Monson FC, Levin RM. Urothelial permeability of the 1987;43:801e13.
isolated whole bladder. Neurourol Urodyn 1993;12:39e47. 59. Yokoyama H, Sasaki K, Franks ME, Goins WF, Goss JR, de Groat WC, et al. Gene
29. Daha LK, Riedl CR, Lazar D, Hohlbrugger G, Pflüger H. Do cystometric findings therapy for bladder overactivity and nociception with herpes simplex virus
predict the results of intravesical hyaluronic acid in women with interstitial vectors expressing preproenkephalin. Hum Gene Ther 2009;20:63e71.
cystitis? Eur Urol 2005;47:393e7. 60. Christ GJ, Day NS, Day M, Santizo C, Zhao W, Sclafani T, et al. Bladder injection
30. Parsons CL. Current strategies for managing interstitial cystitis. Expert Opin of “naked” hSlo/pcDNA3 ameliorates detrusor hyperactivity in obstructed rats
Pharmacother 2004;5:287e93. in vivo. Am J Physiol Regul Integr Comp Physiol 2001;281:R1699e709.
31. Parsons CL, Housley T, Schmidt JD, Lebow D. Treatment of interstitial cystitis 61. Cortesi R, Nastruzzi C. Liposomes, micelles and microemulsions as new delivery
with intravesical heparin. Br J Urol 1994;73:504e7. systems for cytotoxic alkaloids. Pharm Sci Technol Today 1999;2:288e98.
32. Shao Y, Shen ZJ, Rui WB, Zhou WL. Intravesical instillation of hyaluronic acid 62. Fraser MO, Chuang YC, Tyagi P, Yokoyama T, Yoshimura N, Huang L, et al.
prolonged the effect of bladder hydrodistention in patients with severe Intravesical liposome administrationda novel treatment for hyperactive
interstitial cystitis. Urology 2010;75:547e50. bladder in the rat. Urology 2003;61:656e63.
J. Nirmal et al. / Urological Science 23 (2012) 70e77 77

63. Chuang YC, Lee WC, Lee WC, Chiang PH. Intravesical liposome versus oral 76. Svenson S. Dendrimers as versatile platform in drug delivery applications. Eur
pentosan polysulfate for interstitial cystitis/painful bladder syndrome. J Urol J Pharm Biopharm 2009;71:445e62.
2009;182:1393e400. 77. Esfand R, Tomalia DA. Poly(amidoamine) (PAMAM) dendrimers: from bio-
64. Chuang YC, Tyagi P, Huang HY, Yoshimura N, Wu M, Kaufman J, et al. Intra- mimicry to drug delivery and biomedical applications. Drug Discov Today
vesical immune suppression by liposomal tacrolimus in cyclophosphamide- 2001;6:427e36.
induced inflammatory cystitis. Neurourol Urodyn 2011;30:421e7. 78. Tang MX, Redemann CT, Szoka Jr FC. In vitro gene delivery by degraded pol-
65. Tyagi P, Chancellor MB, Li Z, De Groat WC, Yoshimura N, Fraser MO, et al. yamidoamine dendrimers. Bioconjug Chem 1996;7:703e14.
Urodynamic and immunohistochemical evaluation of intravesical capsaicin 79. Patri AK, Myc A, Beals J, Thomas TP, Bander NH, Baker Jr JR. Synthesis and
delivery using thermosensitive hydrogel and liposomes. J Urol 2004;171: in vitro testing of J591 antibody-dendrimer conjugates for targeted prostate
483e9. cancer therapy. Bioconjug Chem 2004;15:1174e81.
66. Chuang YC, Tyagi P, Huang CC, Yoshimura N, Wu M, Kaufman J, et al. Urody- 80. François A, Matthieu Y, Bezdetnaya L, Guillemin F, Battah S, Macrobert A et al.
namic and immunohistochemical evaluation of intravesical botulinum toxin Improvement of fluorescence diagnosis of bladder cancer with ALA den-
a delivery using liposomes. J Urol 2009;182:786e92. drimers. In: 14th Congress of the European Society for Photobiology (ESP
67. Caccin P, Rossetto O, Rigoni M, Johnson E, Schiavo G, Montecucco C. VAMP/ 2011), Sept 1-6, 2011; Geneva, Switzerland.
synaptobrevin cleavage by tetanus and botulinum neurotoxins is strongly 81. Boucher W, Stern JM, Kotsinyan V, Kempuraj D, Papaliodis D, Cohen MS, et al.
enhanced by acidic liposomes. FEBS Lett 2003;542:132e6. Intravesical nanocrystalline silver decreases experimental bladder inflamma-
68. Alexis F, Rhee JW, Richie JP, Radovic-Moreno AF, Langer R, Farokhzad OC. New tion. J Urol 2008;179:1598e602.
frontiers in nanotechnology for cancer treatment. Urol Oncol 2008;26:74e85. 82. Tyagi P, Li Z, Chancellor M, De Groat WC, Yoshimura N, Huang L. Sustained
69. Chang LC, Wu SC, Tsai JW, Yu TJ, Tsai TR. Optimization of epirubicin nano- intravesical drug delivery using thermo hydrogel. Pharm Res 2004;21:832e7.
particles using experimental design for enhanced intravesical drug delivery. Int 83. Smart JD. The basics and underlying mechanisms of mucoadhesion. Adv Drug
J Pharm 2009;376:195e203. Deliv Rev 2005;57:1556e68.
70. Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric nanoparticles based 84. Tyagi P, Wu PC, Chancellor M, Yoshimura N, Huang L. Recent advances in
drug delivery systems. Colloids Surf B Biointerfaces 2010;75:1e18. intravesical drug/gene delivery. Mol Pharm 2006;3:369e79.
71. Gommersall L, Shergill IS, Ahmed HU, Hayne D, Arya M, Patel HR, et al. 85. Eroglu M, Irmak S, Acar A, Denkbaş EB. Design and evaluation of a mucoad-
Nanotechnology and its relevance to the urologist. Eur Urol 2007;52:368e75. hesive therapeutic agent delivery system for postoperative chemotherapy in
72. Arruebo M, Fernández-Pacheco R, Ibarra MR, Santamaría J. Magnetic nano- superficial bladder cancer. Int J Pharm 2002;235:51e9.
particles for drug delivery. Nano Today 2007;2:22e32. 86. Jordan JL, Henderson S, Elson CM, Zhou J, Kydonieus A, Downie J, et al. Use of
73. Chertok B, Moffat BA, David AE, Yu F, Bergemann C, Ross BD, et al. Iron oxide a sulfated chitosan derivative to reduce bladder inflammation in the rat.
nanoparticles as a drug delivery vehicle for MRI monitored magnetic targeting Urology 2007;70:1014e8.
of brain tumors. Biomaterials 2008;29:487e96. 87. Barthelmes J, Perera G, Hombach J, Dunnhaupt S, Bernkop-Schnürch A.
74. Leakakos T, Ji C, Lawson G, Peterson C, Goodwin S. Intravesical administration Development of a mucoadhesive nanoparticulate drug delivery system for
of doxorubicin to swine bladder using magnetically targeted carriers. Cancer a targeted drug release in the bladder. Int J Pharm 2011;416:339e45.
Chemother Pharmacol 2003;51:445e50. 88. Bach P, Wormland RT, Möhring C, Goepel M. Electromotive drug-
75. Duncan R, Izzo L. Dendrimer biocompatibility and toxicity. Adv Drug Deliv Rev administration: a pilot study for minimal-invasive treatment of therapy-
2005;57:2215e37. resistant idiopathic detrusor overactivity. Neurourol Urodyn 2009;28:209e13.

You might also like