Title of Invention

THE COMPOSITIONS FOR INHIBITING PAIN/INFLAMATION AND SPASM COMPRISING KETOPROFEN AND NIFEDIPINE

Abstract Compositions of a cyclooxygenase inhibitor and a calcium channel antagonist in a liquid carrier. The composition may be administered the the urinary tract during urological diagnostic, interventional, surgical and other medical procedures. One disclosed composition comprises ketoprofen and nifedipine in a liquid irrigation carrier, and includes a solubilizing agent, stabilizing agents and a buffering agent.
Full Text CYCLOOXYGENASE INHIBITOR AND CALCIUM CHANNEL ANTAGONIST
COMPOSITIONS AND METHODS FOR USE IN UROLOGICAL PROCEDURES
CROSS REFERENCE TO RELATED APPLICATIONS
The present application claims the benefit of U.S. Provisional Application
No. 60/683,488, filed May 20, 2005.
FIELD OF THE INVENTION
The present invention relates to pharmaceutical compositions for administration to
the urinary tract during urological diagnostic, interventional, surgical and other medical
procedures and for therapeutic treatment of urologic structures.
BACKGROUND OF THE INVENTION
Many urological procedures are now performed using minimally invasive
endoscopic (e.g., cystoscopic or uteroscopic) techniques. These include examination of
the urethra, bladder and ureters, therapeutic treatments for benign prostatic hypertrophy,
removal or fragmentation of kidney and bladder stones, the placement of urethral or
ureteral stents to facilitate the passage of stones, the performance of biopsies and the
excision of tumors. While less invasive than open surgery, these techniques involve
procedural irritation and trauma to the urinary tract leading to pain, inflammation and
smooth muscle spasm. Postoperative lower urinary tract symptoms (LUTS) following
urological procedures often include pain, hyperreflexia (unstable bladder contractions),
urinary frequency, nocturia and urgency, and in some cases urinary retention requiring
prolonged catheterization.
For some surgical procedures, such as transurethral resection of the prostate
(TURP), frequent urination and other symptoms resulting, from the procedural irritation
and inflammation may continue for a prolonged period, gradually resolving during the
first six postoperative weeks. For urologic procedures employing a laser, postoperative
complications such as inflammation and muscle spasm may continue for several weeks.
Patients are frequently prescribed oral anticholinergic medication to inhibit postoperative
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM 1

spasm and reduce the severity of unstable contractions. However, not all patients respond
adequately to these drugs, and side effects may lead to discontinuation of these
medications.
Urological procedures are often performed with concurrent irrigation of the
urinary tract, to remove blood and tissue debris so that a clear endoscopic field of view is
maintained. Conventional irrigation solutions include saline, lactated Ringer's, glycine,
sorbitol, manitol and sorbitol/manitol. These conventional irrigation solutions do not
contain active pharmaceutical agents.
US Patent 5,858,017 to Demopulos, et al, the disclosure of which is hereby
incorporated by reference, discloses surgical irrigation solutions and methods for the
inhibition of pain, inflammation and/or spasm. The use of irrigation solutions containing
pain/inflamation inhibitors and anti-spasm agents during urological procedures in general
and during TURP specifically is disclosed, including five-drug and nine-drug
combinations. This reference does not teach optimized pairings of a pain/inflammation
inhibitory agent with an anti-spasm agent for given urological procedures.
SUMMARY OF THE INVENTION
The present invention provides a locally deliverable composition for inhibiting
pain/inflammation and spasm, comprising a combination of ketoprofen and a calcium
channel antagonist in a carrier. Ketoprofen and the calcium channel antagonist are each
included in a therapeuticaliy effective amount such that the combination inhibits
pain/inflammation and spasm at a site of local delivery.
In a further aspect of the present invention, a locally deliverable composition for
inhibiting pain/inflammation and spasm comprises a combination of a cyclooxygenase
inhibitor and a calcium channel antagonist, propyl gallate as a stabilizing agent and a
liquid carrier. Each active agent is included in a therapeuticaliy effective amount such
that the combination inhibits pain/inflammation and spasm at a site of local delivery.
In a further aspect of the present invention, a locally deliverable composition for
inhibiting pain/inflammation and spasm comprises a combination of a cyclooxygenase
inhibitor and a calcium channel antagonist an aqueous liquid carrier, a cosolvent, at least
one stabilizing agent and a buffer. Each active agent is included in a therapeuticaliy
effective amount such that the combination inhibits pain/inflammation and spasm at a site
of local delivery.
A further aspect of the present invention provides a method of inhibiting
pain/inflammation and spasm in the urinary tract, comprising delivering to the urinary
tract a composition including a combination of ketoprofen and a calcium channel
antagonist in a carrier. Ketoprofen and the calcium channel antagonist are each included
-2-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM


in a therapeutically effective amount such that the combination inhibits pain/inflammation
and spasm in the urinary tract.
A still further aspect of the present invention provides a method of inhibiting
pain/inflammation and spasm in the urinary tract during a diagnostic, interventional,
surgical or other medical urological procedure, comprising periprocedurally delivering to
the urinary tract during a urological procedure a composition including a combination of
ketoprofen and nifedipine in a carrier. Ketoprofen and nifedipine are each included in a
therapeutically effective amount such that the combination inhibits pain/inflammation and
spasm in the urinary tract.
A still further aspect of the present invention provides a method of inhibiting
pain/inflammation and spasm in the urinary tract during a urological procedure,
comprising periprocedurally delivering to the urinary tract during a ureteroscopic
procedure a composition including a combination of a cyclooxygenase inhibitor and a
calcium channel antagonist in a carrier. The cyclooxygenase inhibitor and the calcium
channel antagonist are each included in a therapeuticaily effective amount such that the
combination inhibits pain/inflammation and spasm in the urinary tract.
A still further aspect of the present invention provides a method of inhibiting
pain/inflammation and spasm in the urinary tract during a urological procedure,
comprising periprocedurally delivering to the urinary tract during a procedure to remove,
fragment or dislodge a kidney or bladder stone a composition including a combination of
a cyclooxygenase inhibitor and a calcium channel antagonist in a carrier. The
cyclooxygenase inhibitor and the calcium channel antagonist are each, included in a
therapeutically effective amount such that the combination inhibits pain/inflammation and
spasm in the urinary tract.
A still further aspect of the present invention provides a method of inhibiting
pain/inflammation and spasm in the urinary tract during a urological procedure,
comprising periprocedurally delivering to a urologic structure during a procedure that
causes thermal injury to urinary tract tissue a composition including a combination of a
cyclooxygenase inhibitor and a calcium channel antagonist in a carrier. The
cyclooxygenase inhibitor and the calcium channel antagonist are each included in a
therapeutically effective amount such that the combination inhibits pain/inflammation and
spasm in the urinary tract.
A still further aspect of the present invention provides a method of inhibiting pain,
inflammation and/or spasm in the urinary tract during a urological procedure, comprising
periprocedurally delivering to a urologic structure during a ureteroscopic procedure a
composition including a combination of a plurality of agents that inhibit
pain/inflammation and/or spasm in a carrier. Each agent is included in a therapeutically
-3-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

effective amount such that the combination inhibits pain/inflammation and/or spasm in
the urinary tract.
A still further aspect of the present invention provides a method of inhibiting pain,
inflammation and/or spasm in the urinary tract during a urological procedure, comprising
periprocedurally delivering to a urologic structure during a procedure to remove, fragment
or dislodge a kidney or bladder stone a composition including a combination of a plurality
of agents that inhibit pain/inflammation and/or spasm in a carrier. Each agent is included
in a therapeutically effective amount such that the combination inhibits pain/inflammation
and/or spasm in the urinary tract.
A still further aspect of the present invention provides a method of inhibiting pain,
inflammation and/or spasm in the urinary tract during a urological procedure, comprising
periprocedurally delivering to a urologic structure during a procedure that causes thermal
injury to urinary tract tissue a composition including a combination of a plurality of
agents that inhibit pain/inflammation and/or spasm in a carrier. Each agent is included in
a therapeutically effective amount such that the combination inhibits pain/inflammation
and/or spasm in the urinary tract.
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention will now be described in greater detail, by way of example,
with reference to the accompanying drawings in which:
FIGURE 1 provides a model for action of prostaglandin activity.
FIGURE 2 illustrates the bradykinin and substance P cumulative concentration-
response curves obtained from normal animals in Example I.
FIGURE 3A illustrates bradykinin concentration-response curves produced in the
presence of 0.25, 1.0, 2.5 and 10 µM ketoprofen from Example I; FIGURE 3B illustrates
the Schild plot for pA2 analysis of ketoprofen from Example I.
FIGURE 4A demonstrates that bradykinin rapidly induces the formation of PGE2
in rat bladder tissue strips tested in Example I within the first minutes of stimulation and
reaches a maximum within 30 minutes, with a t1/2 for formation of about 7.5 minutes.
FIGURE 4B illustrates the rapid kinetics of PGE2 formation detected within minutes in
Example I.
FIGURE 5A illustrates that intravenous aspirin (10 mg/kg) produced a gradual
time-dependent inhibition of the acetic acid induced reduction in the intercontraction
interval (ICI), and FIGURE 5B illustrates the parallel changes in bladder capacity, from
Example I.
FIGURE 6 shows the effect of increasing concentrations of nifedipine on
contractility of rat bladder strips from Example II.
-4-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

FIGURE 7 shows the combined effect of nifedipine (0.1 µM) and ketoprofen (0.3-
3.0 µM) on bradykinin-stimulated contractility of rat bladder strips from Example III.
FIGURE 8 shows the combined effect of nifedipine (0.3 µM) and ketoprofen (0.3-
3.0 µM) on bradykinin-stimulated contractility of rat bladder strips from Example III.
FIGURE 9 shows the combined effect of nifedipine (1.0 µM) and ketoprofen (0.3-
3.0 µM) on bradykinin-stimulated contractility of rat bladder strips from Example III.
FIGURE 10 illustrates the concentration-response surface (reduced model) of
individual tension values from dose response curves corresponding to 30 µM bradykinin-
induced tension in rat bladder strips from Example III.
FIGURE 11 shows the effect of ketoprofen (10 µM) and nifedipine (1 µM),
individually, on multiple agonist-stimulated tension in rat bladder tissue strips from
Example IV.
FIGURE 12 shows the effect of ketoprofen (10 µM) and nifedipine (1 µM),
individually, on bradykinin-stimulated PGE2 release from rat bladder tissue strips from
Example IV.
FIGURE 13 shows a rat bladder cystometry tracing demonstrating the effect of
acetic acid perfused as described in Example V.
FIGURE 14 demonstrates the effect of ketoprofen pretreatment on acetic acid-
induced bladder hyperactivity from Example V.
FIGURE 15 demonstrates the effect of nifedipine pretreatment on acetic acid-
induced bladder hyperactivity from Example V.
FIGURE 16 illustrates mean ketoprofen plasma levels for rats treated with
ketoprofen or a combination of ketoprofen and nifedipine in the pharmacokinetic study of
Example VI.
FIGURE 17 illustrates mean nifedipine plasma levels for rats treated with
nifedipine or a combination of ketoprofen and nifedipine in the pharmacokinetic study of
Example VI.
FIGURE 18 illustrates the effects of nifedipine, ketoprofen and a combination of
nifedipine and ketoprofen on PGE2 in rat bladders from the pharmacokinetic study of
Example VI.
FIGURE 19 shows a chromatogram of a nifedipine and ketoprofen formulation F1
in accordance with Example VIII after having been stressed at 60°C for 1 month.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention provides methods and compositions for inhibiting pain,
inflammation and/or spasm during urological procedures by locally delivering such
compositions to structures of the urological tract during the procedure. The compositions
include at least one agent that is a pain/inflammation inhibitory agent or a spasm
-5-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

inhibitory agent, or that acts to inhibit both pain/inflammation and spasm. Preferably, the
compositions and methods of the present invention include two or more
pain/inflammation inhibitory or spasm inhibitory agents that act on different molecular
targets (i.e., enzymes, receptors or ion channels) or that act through different mechanisms
of action. More preferably, the compositions of the present invention include at least one
pain/inflammation inhibitory agent and at least one spasm inhibitory agent.
As used herein, the term "pain/inflammation inhibitory agent" includes analgesic
agents (i.e., antinociceptive agents), non-steroidal agents that inhibit inflammation
[including both "non-steroidal anti-inflammatory drugs" (i.e., NSAIDS or cyclooxygenase
inhibitors) and other agents that are not steroidal that act to inhibit inflammation],
corticosteroids and local anesthetics.
As used herein, the term "spasm inhibitory agent" includes agents that inhibit
spasm or contraction of smooth muscle tissue and agents that inhibit spasm or contraction
of other muscle tissue associated with the urinary tract (e.g., prostatic muscle tissue).
Another aspect of the present invention is directed to the periprocedural delivery
to the urinary tract of a cyclooxygenase (COX) inhibitor, suitably a non-selective COX-
1/C0X-2 inhibitor, preferably a non-selective COX-1/COX-2 inhibitor that is a propionic
acid derivative, more preferably ketoprofen, alone or with at least one additional agent
that inhibits pain/inflammation and/or that inhibits spasm, such as a calcium channel
antagonist.
Another aspect of the present invention is directed to the periprocedural delivery
to the urinary tract of a calcium channel antagonist (i.e., a calcium channel blocker),
suitably an L-type calcium antagonist, preferably a dihydropyridine calcium channel
antagonist, more preferably nifedipine, alone or with at least one additional agent that
inhibits pain/inflammation and/or that inhibits spasm, such as a COX inhibitor.
Another aspect of the present invention is directed to the periprocedural delivery
to the urinary tract of a combination of a COX inhibitor and a calcium channel antagonist,
preferably a non-selective COX-1/COX-2 inhibitor in combination with an L-type
calcium antagonist, more preferably ketoprofen in combination with nifedipine.
Ketoprofen and nifedipine have been found by the present inventors to provide greater
than additive or synergistic results in the inhibition of bladder spasm, as described in the
examples below.
One aspect of the present invention entails the local delivery of the compositions
of the present invention to the bladder, ureter, urethra, or other urinary tract structures to
inhibit pain, inflammation and/or smooth muscle spasm during urological therapeutic,
diagnostic, interventional, surgical and other medical procedures.
-6-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

As used herein, the terms "urinary tract" and "urinary sytem" refer to the kidneys,
ureters, bladder, urethra and associated nerves, blood vessels and muscles. The term
"lower urinary tract" refers to the bladder and urethra and associated nerves, blood vessels
and muscles.
A further aspect of the present invention entails the local delivery of the
compositions of the present invention to urinary tract structures to reduce postoperative
irritative voiding symptoms (e.g., void frequency, nocturia, urgency), pain and/or other
lower urinary tract symptoms following such urological procedures.
A further aspect of the present invention entails the local delivery of the
compositions of the present invention to urinary tract structures to improve postoperative
urinary function (e.g., decrease undesirable urinary retention) following such urological
procedures.
The compositions of the present invention are suitably delivered to the urinary
tract before, during and/or after urological procedures, i.e., before (pre-) procedurally,
during (intra-) procedurally, after (post-) procedurally, pre- and intraprocedurally, pre-
and postprocedurally, intra- and postprocedurally or pre-, intra- and postprocedurally.
Preferably, the compositions of the present invention are locally delivered to the
urinary tract "periprocedurally", which as used herein means intraprocedurally, pre- and
intraprocedurally, intra- and postprocedurally or pre-, intra- and postprocedurally.
Periprocedural delivery may be either continuous or intermittent during the procedure.
Preferably, the compositions of the present invention are delivered "continuously" during
the procedure, which as used herein means delivery so as to maintain an approximately
constant concentration of active agent(s) at the local delivery site. When delivered
periprocedurally during a surgical procedure, the term "perioperatively" may be used
interchangeably with periprocedurally herein. Preferably, the compositions of the present
invention are delivered periprocedurely during the period of time when surgical or other
procedural trauma and irritation is being incurred by urinary tract tissue.
"Local" delivery of the compositions of the present invention to the urinary tract
as used herein refers to deliver}' of the compositions directly to one or more structures of
the urinary tract. The therapeutic agent(s) contained in the locally delivered compositions
are not subject to first and/or second pass metabolism before reaching the local site of
intended therapeutic (e.g., inhibitory) effect, in contrast to systemically delivered drugs.
Pathophysiologic Effects of Urological Procedures
The trauma of urological procedures results in an acute, localized inflammatory
response in the associated urological structures. Inflammation is associated with a
complex pattern of biochemical and cellular processes occurring at the local site,
involving positive-feedback interactions between the peripheral nervous system, immune
-7-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

cells, the local vasculature and the central nervous system. The inflammatory response to
procedural trauma in the urinary tract includes cytokine release, inflammatory cell
migration, edema, pain and hyperalgesia.
In response to tissue injury, numerous local mediators are rapidly released, which
result in nociceptive stimulation of sensory C-fibers. The inflammatory response
triggered by peripheral injury shows that, in addition to cytokines, small G-protein
receptor-linked inflammatory mediators also modulate the rapid pathophysiological
response of the bladder and urethra. In models of urinary bladder inflammation,
bradykinin, histamine, substance P (SP), leukotrienes and prostaglandins have been found
to be released from the bladder. Lecci, A., et al., Pharmacological Analysis of the Local
and Reflex Responses to Bradykinin on Rat Urinary Bladder Motility in Vivo, Br. J.
Pharmacol., 114:708-14 (1995); Lecci, A., et al., Capsaicin Pretreatment Does Not Alter
Rat Urinary Bladder Motor Responses Induced by a Kinin B1 Receptor Agonist After
Endotoxin Treatment, Neurosci. Lett. 262:73-76 (1999); Vasko, M., et al., Prostaglandin
E2 Enhances Bradykinin-Stimulated Release of Neuropeptides from Rat Sensory Neurons
in Culture, J Neurosci. 14:4987-97 (1994). Certain inflammatory mediators, such as
prostaglandins and kinins, activate and sensitize C-fibers through interaction with specific
receptors on nerve terminals. Other inflammatory mediators that have been described in
the lower urinary tract include tachykinins and ATP (from C-fibers) (Maggi, C, et al.,
Tachkykinin Antagonists and Capsaicin-Induced Contraction of the Rat Isolated Urinay
Bladder: Evidence for Tachykinin-Mediated Cotransmission, Br. J. Pharmacol. 103:1535-
41 (1991), CGRP (from C-fibers), serotonin (from mast cells and platelets), and
endothelin. Maggi, C, et al., Contractile Responses of the Human Urinary Bladder,
Renal Pelvis and Renal Artery to Endothelins and Sarafotoxin S6b, Gen. Pharmacol.
21:247-49 (1990). These mediators operate together in a synergistic manner to increase
postsurgical hyperalgesia, inflammation and muscle spasm. The number of mediators
involved in the response underscores the multifactorial origin of the pain and
inflammation process.
The immediate activation of the sensory nerves (primary hyperalgesia) triggers a
cascade of processes that involves alterations in the local vasculature, and influences
muscle contractility. Capsaicin-sensitive afferent fiber stimulation elicits a local efferent
response, which is characterized by release of neuropeptides (tachykinins and CGRP)
from nerve endings. This release produces a number of local responses, which are part of
the pathophysiological effects in the lower urinary tract. These include: (1) direct effects
of released neurotransmitters on smooth muscle contraction; (2) changes in microvascular
permeability resulting in plasma extravasation and edema of the bladder, urethra and
prostate; (3) infiltration of immune cells; and (4) sensitization of nociceptors (secondary
-8-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

hyperalgesia) resulting in increased pain. The consequences of these processes can affect
normal bladder capacity and frequency of micturition, and often result in hypersensitivity,
pain and smooth muscle spasm.
The pathophysiologic response to procedural trauma of the urinary tract involves a
complex cascade of molecular signaling and biochemical changes resulting in
inflammation, pain, spasm and lower urinary tract symptoms. These are preferably
addressed in accordance with the methods and compositions of the present invention by
locally and periprocedurally delivering a combination of pharmacologic agents acting on
multiple molecular targets to inhibit pain, inflammation and/or spasm. Preferred agents
include cyclooxygenase inhibitors and calcium channel antagonists, more preferably in
combination.
Cyclooxygenase Inhibitors
Prostaglandins are produced throughout the lower urinary tract and play a role in
neurotransmission, bladder contractility and inflammatory responses. Human bladder
mucosa has been found to contain several types of prostaglandins, which have been
shown to contract the human detrusor. Prostaglandin E2 (PGE2) is a potent mediator of
pain and edema, and the exogenous administration of PGE2 induces contractile responses
in inflamed bladders. Intravesical PGE2 produces both urgency and involuntary bladder
contractions. Lepor, H., The Pathophysiology of Lower Urinary Tract Symptoms in the
Ageing Male Population, Br. J Urol., 81 Suppl 1:29-33 (1998); Maggi, C, et al.,
Prostanoids Modulate Reflex Micturition by Acting Through Capsaicin-Sensitive
Afferents, Eur. J. Pharmacol. 145: 105-12 (1988). PGE2 given intravesically may
stimulate micturition by releasing tachykinins from nerves in and/or immediately below
the urothelium. Ishizuka, O., et al., Prostaglandin E2-Induced Bladder Hyperactivity in
Normal, Conscious Rats: Involvement of Tachykinins?, J Urol. 153:2034-38 (1995).
Prostanoids may, via release of tachykinins, contribute to both urge and bladder
hyperactivity seen in inflammatory conditions of the lower urinary tract. While not
wishing to be limited by theory, these actions are most likely mediated through activation
of specific prostanoid receptor subtypes (EP1R) located on C-fibers and on bladder
smooth muscle (FIGURE 1).
In the inflamed bladder, the basal production of PGE2 is significantly higher than
in control conditions. A number of inflammatory mediators acting through GPCR
pathways that are linked to the production of arachidonic acid may up-regulate
prostaglandin levels in the mucosa and vascular endothelium. Bradykinin is a well-
established mediator of inflammation, and bradykinin receptor agonists stimulate greater
PGE2 production in inflamed bladders than in control bladders. Topical application of
bradykinin activates bladder sensory nerves. Lecci, A., et al., Kinin Bl Receptor-
-9-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Mediated Motor Responses in Normal or Inflamed Rat Urinary Bladder in Vivo, Regul.
Pept. 80:41-47 (1999); Maggi, C, et al., Multiple Mechanisms in the Motor Responses of
the Guinea-Pig Isolated Urinary Bladder to Bradykinin, Br. J. Pharmacol. 98:619-29
(1989). Contractile responses elicited by the selective B1 and B2 receptor agonists tested
in isolated rat urinary bladder strips showed that the contractile responses to a selective
Bl agonist were also potentiated in inflamed bladders. The role of bradykinin in reflex
voiding has also been investigated in normal rats using continuous infusion cystometry.
Infusion of bradykinin produced a significant decrease in the intercontraction interval
(ICI) between voiding events and an increase in bladder contraction amplitude that is
completely blocked by a B2 receptor antagonist.
Microvascular leakage induced by administration of substance P acting through
the NK1 receptor also involves the release of cyclooxygenase metabolites of arachidonic
acid. Abelli, L., et al., Microvascular Leakage Induced by Substance P in Rat Urinary
Bladder: Involvement of Cyclo-oxygenase Metabolites of Arachidonic Acid, J. Auton.
Pharmacol. 12:269-76 (1992). These findings demonstrate that distinct inflammatory
mediators act through independent receptor mechanisms to trigger the production of
prostaglandins. NSAIDs that act at a common target downstream of multiple GPCRs to
inhibit COX-1/COX-2 have the capacity to block the formation of prostaglandins derived
from multiple pro inflammatory mediators.
A number of studies have shown that both COX-1 and COX-2 are involved in the
production of PGE2 during tissue trauma and the acute inflammatory response. Martinez,
R., et al., Involvement of Peripheral Cyclooxygenase-1 and Cyclooxygenase-2 in
Inflammatory Pain, J Pharm Pharmacol. 54:405-412 (2002); Mazario, J, et al.,
Cyclooxygenase-1 vs. Cyclooxygenase-2 Inhibitors in the Induction of Antinociception in
Rodent Withdrawal Reflexes, Neuropharmacology. 40:937-946 (2001); Torres-Lopez, J.,
et al., Comparison of the Antinociceptive Effect of Celecoxib, Diclofenac and Resveratrol
in the Formalin Test, Life Sci. 70:1669-1676 (2002). In normal bladders, activation of
B2 receptors evokes bladder contraction mediated by COX-1 activity, whereas COX-2
activity is involved in production of PGE2 driven through stimulation of Bl receptors
only. COX-2 is the major isoform that is rapidly expressed and dramatically up-regulated
during bladder inflammation. It is believed to be responsible for the high levels of
prostanoids released during acute and chronic inflammation of the bladder. COX-2 is up-
regulated in response to proinflammatory cytokines and bladder treatment with either
endotoxin or cyclophosphamide. Both COX isozymes are therefore suitable molecular
targets for the drug compositions of the present invention.
An aspect of the present invention is directed to therapeutic compositions
including a cyclooxygenase inhibitor in a carrier suitable for local delivery to urologic
-10-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

structures in the urinary tract. To achieve maximal inhibition of prostaglandin synthesis
at sites of acute inflammation, it is believed desirable to inhibit both COX isoenzymes.
The COX inhibitor is therefore preferably non-selective with respect to activity at
COX-1 and COX-2, which for purposes of the present invention may be defined as an
agent for which the ratio of (a) the concentration of the agent effective for the inhibition
of 50% (IC50) of the activity of COX-1 relative to (b) the IC50 for the inhibition of the
activity of COX-2 is greater than or equal to 0.1 and less than or equal to 10.0, and more
preferably is greater than or equal to 0.1 and less than or equal to 1.0. Suitable assays for
determining COX-1 and COX-2 inhibitory effect are disclosed in Riendau, D., et al.,
Comparison of the Cyclooxygenase-l Inhibitory Properties of Nonsteroidal Anti-
inflammatory Drugs (NSAIDs) and Selective COX-2 Inhibitors, Using Sensitive
Microsomal and Platelet Assays, Can. J. Physiol. Pharmacol. 75:1088-1095 (1997).
Suitable non-selective COX-1/COX-2 inhibitors include, for purposes of
illustration, salicylic acid derivatives including aspirin, sodium salicylate, choline
magnesium trisalicylate, salsalate, diflunisal, sulfasalazine and olsalazine, para-
aminophenol derivatives such as acetaminophen, indole and indene acetic acids such as
indomethacin and sulindac, heteroaryl acetic acids including tolmetin, diclofenac and
keterolac, arylpropionic acids including ibuprofen, naproxen, flurbiprofen, ketoprofen,
fenoprofen and oxaprozin, anthranilic acids (fenamates) including mefanamic acid and
meclofenamic acid, enolic acids including oxicams such as piroxicam and meloxicam and
alkanones such as nabumetone, as well as pharmaceutically effective esters, salts,
isomers, conjugates and prodrugs thereof.
Still more preferably, the non-selective COX-1/COX-2 inhibitor is an
arylpropionic acid, i.e., a propionic acid derivative, such as ketoprofen, dexketoprofen,
ibuprofen, naproxen, flurbiprofen, fenoprofen and oxaprozin. Most preferably, the agent
is ketoprofen.
In another aspect of the invention, the non-selective COX-1/COX-2 inhibitor used
in the compositions and methods of the present invention is selected as having an IC50
for the inhibition of bradykinin-induced bladder smooth-muscle strip contractility (as
determined by the bladder contractility model described herein below) of less than or
equal to 100 µM, preferably less than or equal to 25 µM, more preferably less than or
equal to 5 µM, still more preferably less than 2 µM.
In a further aspect of the invention, the non-selective COX-1/COX-2 inhibitor
used in the compositions and methods of the present invention is selected as having an
IC50 for the inhibition of bradykinin-induced prostaglandin E2 (PGE2) (as determined by
the PGE2 bladder tissue analysis model described herein below) of less than or equal to
-11-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

100 µM, preferably less than or equal to 25 µM, more preferably less than or equal to 5
µM, still more preferably less than 2 µM.
In a still further aspect of the invention, the non-selective COX-1/COX-2 inhibitor
used in the compositions and methods of the present invention is selected as having (a) an
IC50 for the inhibition of bradykinin-induced bladder smooth-muscle strip contractility
(as determined by the bladder contractility model described herein below) of less than or
equal to 100 µM , preferably less than or equal to 25 µM, more preferably less than or
equal to 5 µM, still more preferably less than 2 µM, and (b) an IC50 for the inhibition of
bradykinin-induced PGE2 (as determined by the PGE2 bladder tissue analysis model
described herein below) of less than or equal to 100 µM , preferably less than or equal to
25 µM, more preferably less that or equal to 5 µM, still more preferably less than 2 µM.
The above noted IC50 concentrations are not to be interpreted as limitations on
drug concentrations in the compositions of the present invention, which may suitably be
determined by the concentrations needed to approach maximal effectiveness and thus may
be higher than the IC50 levels.
In a still further aspect of the invention, the non-selective COX-1/COX-2 inhibitor
used in the compositions and methods of the present invention is selected as having a pA2
(antagonist potency) of greater than or equal to 7, wherein pA2 is the negative logarithm
of the concentration of antagonist that would produce a 2-fold shift in the concentration
response curve for an agonist, and is a logarithmic measure of the potency of an
antagonist. This potency corresponds to an equilibrium dissociation constant KD of less
than or equal to 100 nM.
In a still further aspect of the invention, the non-selective COX-1/COX-2 inhibitor
used in the compositions and methods of the present invention exhibits 50% of maximal
inhibitory response in less than or equal to 10 minutes in a kinetic study of bradykinin-
stimulated PGE2 response in the PGE2 bladder tissue analysis model described herein
below.
Ketoprofen
Unless used in a context also referring to its isomer, references herein to the use of
ketoprofen (i.e., m-benzoylhydratropic acid or 3-benzoyl-α-methylbenzeneacetic acid) in
the present invention are to be understood to also include pharmaceutically acceptable
isomers thereof, including its racemic S-(+)-enantiomer, dexketoprofen, pharmaceutically
acceptable salts or esters thereof, and pharmaceutically acceptable prodrugs or conjugates
thereof. Ketoprofen is a preferred COX inhibitor for use in the present invention.
Ketoprofen exhibits potent anti-inflammatory, analgesic, and antipyretic actions
that are associated with the inhibition of prostaglandin synthesis and antagonism of the
effects of bradykinin. Ketoprofen non-selectively inhibits the activity of COX-1 and
-12-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

COX-2, which results in the blockade of prostaglandin production, particularly that of
PGE2, preventing the development of hyperalgesia. Ketoprofen has an IC50 value of
4 - 8 nM in a non-selective COX assay, being functionally 6-12 times more potent than
other NSAIDs evaluated (e.g., naproxen or indomethacin). Kantor, T., Ketoprofen: A
review of its Pharmacologic and Clinical Properties, Pharmacotherapy 6:93-103 (1986).
Ketoprofen also has functional bradykinin antagonist activity, its effects being eight times
greater than those seen with the classical NSAID, indomethacin. Julou, L., et al.,
Ketoprofen (19.583 R.P.) (2-(3-Benzoylphenyl)-propionic acid). Main Pharmacological
Properties - Outline of Toxicological and Pharmacokinetic Data, Scand J Rheumatol
Suppl. 0:33-44 (1976).
In addition to inhibiting cyclooxygenase, ketoprofen is believed to offer the
additional anti-inflammatory benefit of inhibiting lipoxygenase. Ketoprofen has also
been found to synergise with nifedipine in the inhibition of bladder spasm, as discussed in
greater detail in the examples below.
Calcium Channel Antagonists
Multiple inflammatory mediators, including bradykinin, are released into the
bladder in response to tissue injury, which can trigger smooth muscle contraction and
spasm. The tone of the urinary bladder smooth muscle is regulated by numerous
contraction-promoting receptor systems. They include well established systems such as
muscarinic, purinergic and tachykinin receptors [Anderson, K., et al., Pharmacolgy of the
Lower Urinary Tract: Basis for Current and Future Treatments for Urinary Incontenance
Pharmacol Rev. 56:581-631 (2004)], and also include endothelin receptors [Afiatpour, P.,
et al., Development Changes in the Functional, Biochemical and Molecular Properties of
Rat Bladder Endothelin Receptors, Naunyn Schmiedebergs Arch. Pharmacol. 367:462-72
(2003)], protease-activated receptors and bradykinin receptors [Kubota, Y., et al., Role of
Mitochondria in the Generation of Spontaneous Activity in Detrusor Smooth Muscles of
the Guinea Pig Bladder, J. Urol. 170:628-33 (2003); Trevisani, M., et al., Evidence for In
Vitro Expression of Bl Receptor in the Mouse Trachea and Urinary Bladder, Br. J.
Pharmacol. 126:1293-1300 (1999)]. Because many of these receptors are prototypically
coupled via Gq proteins to the activation of a phospholipase C (PLC), it is likely that
bladder contraction elicited by such receptors is partly mediated by PLC-linked
mobilization of Ca2+ from intraceliular stores [Ouslander, J. G., Management of
Overactive Bladder, N. Engl. J. Med., 350:786-99 (2004)].
Neurally mediated contractions of the bladder and urethral smooth muscle require
mobilization of intraceliular Ca2+ as well as an influx of extracellular Ca2+. Ca2+ entry
through L-type calcium channels can contribute to muscle contractions by triggering the
intraceliular release of Ca2+, which opens ryanodine-sensitive Ca2+ release channels in the
-13-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

sarcoplasmic reticulum. Opening of L-type calcium channels in bladder muscle also
serves to replace intracellular Ca2+ stores after contraction. Recent studies conclude that
muscarinic receptor subtype signaling mediated via carbachol-induced contraction of rat
bladders largely depends on Ca2+ entry through L-type calcium channels and, perhaps,
PLD, PLA2 and store-operated Ca"+ channels. Schneider, T., et al., Signal Transduction
Underlying Carbachol-induced Contraction of Rat Urinary Bladder: I. Phospholipases
and Ca2+ sources, J Pharmacol Exp Ther (2003). Thus, blockade of L-type Ca2+
channels has the potential to depress neural, urothelial and smooth muscle evoked
contractions of bladder strips mediated by a multiplicity of endogenous GPCR agonists.
The L-type calcium channel represents a point of integration for the convergence of
multiple inflammatory mediators that can lead to hyperactive smooth muscle contractility.
Ca 2+ channels located in afferent and efferent nerve terminals in the lower urinary
tract are also important for regulation of neurotransmitter release, de Groat, W., et al.,
Pharmacology of the Lower Urinary Tract, Annu. Rev. Pharmacol Toxicol. 41:691-721
(2001). A number of active agents produce Ca + influx and transmitter release from the
peripheral nerve endings of capsaicin-sensitive afferent neurons through voltage-sensitive
Ca2+ channels. Under certain conditions, L-type Ca2+ channels can also contribute to
transmitter release.
The significant role of the L-type Ca2+ channel in the initiation of smooth muscle
contraction makes this channel a potential therapeutic target for the treatment of lower
urinary tract problems that involve hyperactivity or spasm of smooth muscle tissues. In
the presence of inflammatory mediators, signaling through these same channels may
mediate bladder hyperactivity and spasm.
An aspect of the present invention is thus directed to therapeutic compositions
including a calcium channel antagonist in a carrier suitable for delivery to urologic
structures in the urinary tract. The calcium channel antagonist is preferably an L-type
calcium channel antagonist, such as verapamil, diltiazem, bepridil, mibefradil, nifedipine,
nicardipine, isradipine, amlodipine, felodipine, nisoldipine and nimodipine, as well as
pharmaceutically effective esters, salts, isomers, conjugates and prodrugs thereof. Still
more preferably,' the calcium channel antagonist is a dihydropyridine, such as nifedipine,
nicardipine, isradipine, amlodipine, felodipine, nisoldipine and nimodipine, as well as
pharmaceutically effective esters, salts, isomers, conjugates and prodrugs thereof. Most
suitably, the agent is nifedipine.
Nifedipine
References herein to nifedipine, 1,4-dihydro-2,6-dimethyl-4-(2-nitrophenyl)-3,5-
pyridinedicarboxylic acid dimethyl ester, are to be understood to also include
pharmaceuticaliy acceptable isomers thereof, pharmaceutically acceptable salts or esters
-14-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

thereof, and pharmaceutically acceptable prodrugs or conjugates thereof. Nifedipine is a
preferred calcium channel antagonist for use in the present invention.
Nifedipine is a member of the dihydropyridine class of calcium channel
antagonists with pharmacological specificity for the L-type channel (alternatively termed
the Cavl.2 a-subunit). Nifedipine has a rapid onset of action (less than 10 minutes),
which is desirable for use in urological procedures, and as such is more preferred than
certain closely related dihydropryidine calcium channel antagonists (e.g., amlodipine) that
require longer periods for initial action. The time to response for steady-state inhibition
of muscle contraction ideally occur within 10-15 minutes of initial local drug delivery,
and nifedipine fulfills this criterion.
Carriers
The pain/inflammation and/or spasm agents of the present invention are suitably
delivered in solution or in suspension in a liquid carrier, which as used herein is intended
to encompass biocompatible solvents, suspensions, polymerizable and non-polymerizable
gels, pastes and salves. Preferably, the carrier is an aqueous irrigation solution that may
or may not include physiologic electrolytes, such as saline, distilled water, lactated
Ringer's solution, glycine .solutions, sorbitol solutions, manitol solutions or
sorbital/manitol solutions. The carrier may also include a sustained release delivery
vehicle, such as microparticles, microspheres or nanoparticles composed of proteins,
liposomes, carbohydrates, synthetic organic compounds, or inorganic compounds.
The compositions of the present invention may also be coated on ureteral and
urethral stents, catheters, radioactive seeds, seed spacers and other implantable devices
and on surgical instruments, for local delivery from such devices and instruments into the
urinary tract as further described below. Polymers that may be suitably employed to form
a drug impregnated stent or other implantable device include, by way of non-limiting
example, poly(D,L-lactic acid) (PDLLA), poly(lactide-co-glyocide) (PLGA), poly(L-
lactic acid) (PLLA), poly(glycolic acid), poly(6-hydroxycaproic acid), poly(5-
hydroxyvaleric acid), poly(4-hydroxybutyric acid), polyethylene glycol), poly(ethylene
oxide)-poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO, Pluronics™) block
copolymers, and copolymers and blends of the above.
Suitable materials for use in producing drug coated stents, catheters, other
implantable devices and instruments include biodegradable polymers and polymeric
hydrogels, such as by way of nonlimiting example, Pluronics™ triblock copolymers,
PLLAs or their copolyesters, poly(glycolic acid) or their copolyesters, poly(ethylene
oxide) - cyclodextrin (polyrotaxan) hydrogels, poly[(i?)-3-hydroxybutyrate]-poly(ethylene
oxide) - cyclodextrin hydrogels, cellulose acetate, cellulose acetate butyrate, cellulose
acetate propionate, and cellulose nitrate; polyurethane resins, including the reaction
-15-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

product of 2,4-tolylene diisocyanate, 4,4'-diphenylmethane diisocyanate,
polymethylenepolyphenyl isocyanate, or 1,5-napthylene diisocyanate with 1,2-
polypropylene glycol, polytetramethylene ether glycol, 1,4-butanediol, 1,4-butylene
glycol, 1,3-butylene glycol, poly(l,4-oxybutylene)glycol, caprolactone, adipic acid esters,
phthalic anhydride, ethylene glycol, 1,3-butylene glycol, 1,4-butylene glycol or diethylene
glycol; acrylic polymers such as ethyl and methyl acrylate and methacrylate; condensation
polymers such as those produced by sulfonoamides such as toluenesulfonamide and
aldehydes such as formaldehyde; isocyanate compounds; poly(ortho esters);
poly(anhydrides); polyamides; polycyanoacrylates, poly(amino acids), polycarbonate),
cross-linked poly(vinyl alcohol), polyacetals, polycaprolactone. In addition to these
biodegradable polymers, suitable non-biodegradable polymers include polyacrylates,
polystyrenes, polyvinyl chloride, ethylene-vinyl acetate copolymers, polyvinyl fluoride,
poly(vinyl imidazole) and chlorosulphonated polyolefms.
The pain/inflammation and/or spasm inhibitory compositions of the present
invention can also include excipients or adjuvants for enhanced uptake, release, solubility
and stability. Aspects of formulating the compositions of the present invention are
discussed below.
Additional Agents
The cyclooxygenase inhibitor, calcium channel antagonist or combination
cyclooxygcnasc inhibitor plus calcium channel antagonist compositions of the present'
invention may include alternate or additional agents that inhibit pain, inflammation and/or
spasm. Suitable agents include those disclosed in US Patent 5,858,017 to Demopulos.
In particular, suitable alternate or additional anti-inflammation/anti-pain agents
include serotonin receptor antagonists, (e.g., amitriptyline, imipramine, trazodone,
desipramine, ketanserin, tropisetron, metoclopramide, cisapride, ondansetron, yohimbine,
GR127935, methiothepin), serotonin receptor agonists (e.g., buspirone, sumatriptan,
dihydroergotamine, ergonovine), histamine receptor antagonists (e.g., promethazine,
diphenhydramine, amitriptyline, terfenadine, mepyramine (pyrilamine), tripolidine),
bradykinin receptor antagonists (e.g., [Leu8] des-Arg -BK, [des-Arg10] derivative of
HOE 140, [leu9] [des- Arg10] kalliden, [D-Phe7]-BK, NPC 349, NPC 567, HOE 140),
kallikrien inhibitors (e.g., aprotinin), tachykinin receptor antagonists, including
neurokinini receptor subtype antagonists (e.g., GR 82334, CP 96.345, RP 67580) and
neurokinm2 receptor subtype antagonists (e.g., MEN 10.627, L 659.877, (±)-SR 48968),
calcitonin gene-related peptide (CGRP) receptor antagonists [e.g., αCGRP-(8-37)],
interleukin receptor antagonists, (e.g., Lys-D-Pro-Thr), phospholipase inhibitors including
PLA2 isoform inhibitors (e.g., manoalide) and PLCγ isoform inhibitors (e.g., l-[6-((17β-
3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl]-1H-pyrrole-2,5-dione), lipooxygenase
-16-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

inhibitors, (e.g., AA 861), prostanoid receptor antagonists including eicosanoid EP-1 and
EP-4 receptor subtype antagonists and thromboxane receptor subtype antagonists, (e.g.,
SC 19220), leukotriene receptor antagonists including- leukotriene B4 receptor subtype
antagonists and leukotriene D4 receptor subtype antagonists, (e.g., SC 53228), opioid
receptor agonists, including µ-opioid, S-opioid and K-opioid receptor subtype agonists,
(e.g., DAMGO, sufentanyl, fentanyl, morphine, PL 017, DPDPE, U50,488), purinoceptor
agonists and antagonists including P2X receptor antagonists and P2Y receptor agonists,
(e.g., suramin, PPADS), adenosine triphosphate (ATP)-sensitive potassium channel
openers, (e.g., cromakalim, nicorandil, minoxidil, P 1075, KRN 2391, (-)pinacidil),
neuronal nicotinic agonists (e.g., (R)-5-(2-azetidinylmethoxy)-2-chloropyridine (ABT-
594), (S)-5-(2-azetidinyl-methoxy)-2-chloro-pyridine (S-enatiomer of ABT-594), 2-
methyl-3-(2-(S)-pyrrolidinyl-methoxy)-pyridine (ABT-089), (R)-5-(2-
Azetidinylmethoxy)-2-chloropyridine (ABT-594), (2,4)-Dimethoxy-benzylidene
anabaseine (GTS-21), - SBI-1765F, RJR-2403), 3-((1-methyl-2(S)-
pyrrolidinyl)methoxy)pyridine (A-84543), 3-(2(S)-azetidinylmethoxy)pyridine (A-
85380), (+)-anatoxin-A and (-)anatoxin-A (lR)-l-(9-Azabicyclo[4.2.2]non-2-en-2-yl)-
ethanoate fumarate, (R,S)-3-pyridyl-l-methyl-2-(3-pyridyl)-azetidine (MPA), cystisine,
lobeline, RJR-2403, SIB-1765F, GTS-21, ABT-418), α2-adrenergic receptor agonists
[e.g., clonidine, dexmedetomidine, oxymetazonline, (R)-(-)-3'-(2-amino-l-hydroxyethyl)-
4'-fluoro-methanesulfoanilide (NS-49), 2-[(5-methylbenz-1-ox-4-azin-6-
yl)imino]imidazoline (AGN-193080), AGN 191103; AGN 192172, 5-bromo-N-(4,5-
dihydro-lH-imidazol-2-yl)-6-quinoxalinamine (UK14304), 5,6,7,8-tetrahydro-6-(2-
propenyl)-4H-thiazolo[4,5-d]azepin-2-amine (BHT920), 6-ethyl-5,6,7,8-tetrahydro-4H-
oxaazolo[4,5-d]azepin-2-amine (BHT933), 5,6-dihydroxy-1,2,3,4-tetrahydro-1-naphyl-
imidazoline (A-54741)], mitogen-activated protein kinase (MAPK) inhibitors (e.g., 4-(4-
fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4- pyridyl)-1H-imidazole, [4-(3-iodo-
phenyl)-2-(4-methylsulfinylphenyl)-5-(4- pyridyl)-1H-imidazole], [4-(4-fluorophenyl)-2-
(4-hydroxyphenyl)-5-(4- pyridyl)-1H-imidazole], [4-(4-fluoro-phenyl)-2-(4-nitrophenyl)-
5-(4-pyridyl)-1H-imidazole], 2'-Amino-3'-methoxy-flavone), soluble receptors (e.g.,
tumor necrosis factor (TNF) soluble receptors, interleukin-1 (IL-1) cytokine receptors,
class I cytokine receptors, and receptor tyrosine kinases), corticosteroids. (e.g., cortisol,
cortisone, prednisone, prednisolone, flurdrocortisone, 6α-methylprednisolone,
tramcinolone, betamethasone, dexamethasone) and local anesthetics (e.g., benzocaine,
bupivacaine, chloroprocaine, cocaine, etiodocaine, lidocaine, mepivacaine, pramoxine,
prilocaine, procaine, proparacaine, ropivacaine, tetracaine, dibucaine, QX-222, ZX-314,
RAC-109, HS-37).
-17-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Suitable alternate or additional spasm inhibitory agents include serotonin receptor
antagonists (e.g., amitriptyline, imipramine, trazodone, desipramine, ketanserin,
tropisetron, metoclopramide, cisapride, ondansetron, yohimbine, GR127935,
methiothepin, oxymetazoline), tachykinin recptor antagonists including neurokinini
receptor subtype antagonists (e.g., GR 82334, CP 96.345, RP 67580) and neurokinin2
receptor subtype antagonists (e.g., MEN 10.627, L 659.877, (±)-SR 48968), adenosine
triphosphate (ATP)-sensitive potassium channel openers, (e.g., cromakalim, nicorandil,
minoxidil, P 1075, KRN 2391, (-)pinacidil), nitric-oxide donors, (e.g., nitroglycerin,
sodium nitroprusside, SIN-1, SNAP, FK 409 (NOR-3), FR 144420 (NOR-4), endothelin
receptor antagonists, (e.g., BQ 123, FR 139317, BQ 610) and anticholinergics, including
antimuscarinics (e.g., ditropan, tropicamide, cyclopentolate, scopolamine, atropine,
homatropine and oxybutynin), antinicotinics (e.g., trimethaphan, macamylamine,
pentolinium, pempidine and hexamethomium) and first generation antihistamines (e.g.,
diphenhydramine).
Methods of Use
Periprocedural Delivery
Local perioperative delivery of the compositions of the present invention are
expected to preemptively inhibit pain, inflammation and smooth muscle spasm otherwise
associated with urological procedures. The compositions of the present invention act on
molecular targets, i.e., receptors, enzymes and ion channels, that initiate pain,
inflammation and spasm pathways and mechanisms. The present invention employs local
periprocedural delivery to inhibit these pathophysiologic processes at the time they are
initiated. For example, multiple proinflammatory peptides stimulate the release of PGE2
from bladder tissue within the first five minutes of exposure, as shown in the examples
below. Solely postprocedurally administered therapeutic agents can only take effect after
these processes have commenced.
Local Delivery
Local delivery of drugs in accordance with the present invention permits the
utilization of a much lower dosage than would be needed if the same drugs were
administered systemically (e.g., orally, intravenously, intramuscularly, subcutaneously) to
achieve the same predetermined local level of inhibitory effect in the urinary tract. The
focused, local delivery of the present invention results in a significantly lower plasma
level of the drug than would result from systemic delivery of the drug to achieve the same
predetermined local level of inhibitory effect in the urinary tract, thereby reducing the
potential for undesirable systemic side effects. Local delivery permits the inclusion in the
-18-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

compositions of the present invention of drugs such as peptides that are not susceptible to
systemic delivery due to degradation during first- and second-pass metabolism.
Local delivery of drug compositions in accordance with the present invention
provides for an immediate and certain therapeutic concentration at the local urinary tract
site, which is not dependent on variations in metabolism or organ function. A constant
concentration of the drugs can be maintained during the period of delivery of the
composition during the procedure.
Urological Procedures
The compositions of the present invention can be locally delivered before, during
and/or after cystoscopy, i.e., the endoscopic examination of the urethra and bladder
through a cystoscope inserted into the lower urinary tract for purposes of examining the
urinary tract structures, preferably periprocedurally during such procedures. The
compositions of the present invention may also be used before, during and/or after
(preferably periprocedurally during) other diagnostic, interventional, medical and surgical
procedures performed in conjunction with cystoscopy, by insertion of surgical instruments
through the cystoscope, such as for the removal of tissue for biopsy, removal of growths,
removal of foreign bodies, bladder or kidney stone removal, placement, removal and
manipulation of urethral stents, transurethral resection of bladder tumors (TURBT),
treatment of tumors with electrocautery or laser or local chemotherapeutics, treatment of
bleeding in the bladder or to relieve obstructions in the urethra.
The compositions of the present invention can be locally delivered to the urinary
tract before, during and/or after ureteroscopy, i.e., the endoscopic examination of the
ureters and renal tissues through an ureteroscope inserted through the urethra and bladder
and into a ureter for purposes of examining the urinary tract structures, preferably
periperatively during such procedures. Ureteroscopy is often performed for the drawing
of urine samples from each kidney, the placement, removal and manipulation of ureteral
stents, as part of the treatment for kidney stones, or to place a catheter in the ureter for a
retrograde pyelography, and the compositions of the present invention can be delivered
before, during and/or after such procedures, preferable periprocedurally during such
procedures. A basket or other instrument employed via the ureteroscope can be used to
capture the stone, the stone may be broken up by laser or shock wave lithotripsy through
the ureteroscope, or the ureteroscope may be employed to displace a lodged stone back
into the kidney for subsequent breaking up and passage, such as by using a laser or
extracorporeal shock wave lithotripsy (ESWL).
The compositions of the present invention are suitably locally delivered to the
urinary tract before, during and/or after procedures that typically result in ureteral spasm,
-19-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

such as kidney stone removal using laser treatment, cystoscopy, ureteroscopy or
lithotripsy, and preferably periprocedurally during such stone removal procedures.
The compositions of the present invention may also be locally delivered to the
urinary tract before, during and/or after (preferably periprocedurally) urological
procedures that cause thermal trauma to tissue in and/or associated with the urinary tract.
These include laser treatment to fragment stones or ablate tissue, microwave ablation of
tissue (e.g., transurethral microwave thermotherapy (TUMT) to remove prostatic tissue),
radiofrequency ablation of tissue (e.g., transurethral needle ablation (TUNA) to remove
prostatic tissue), electrocauterization or vaporization of tissue or cryoblation of tissue.
The compositions of the present invention may also be locally delivered to the
urinary tract before, during and/or after (preferably periprocedurally) urological
procedures employing a laser for tissue resection, including Holmium: yttrium-aluminum-
garnet (Ho:YAG), neodymium:yttrium-aluminum-garnet (Nd:YAG) and potassium-
titanyl-phosphate (KTP) "green light" laser therapies. Such laser procedures may include
the treatment of benign prostatic hyperplasia (BPH) and bladder tumors, by way of non-
limiting example.
The ketoprofen composition, calcium channel antagonist and ketoprofen
combination composition and the preferred ketoprofen and nifedipine combination
composition of the present invention may also be locally delivered to the urinary tract
before, during and/or after (preferably periprocedurally) transurethral resection of the
prostate (TURP).
In addition to transurethral procedures such as those discussed above, the
compositions of the present invention may also be suitably employed for local delivery
during other minimally invasive urological procedures. These include, by way of
example, the transrectal or transperitoneal delivery of the compositions of the present
invention to the prostate and surrounding anatomic structures during implantation of
radioactive seeds and seed spacers to treat prostate cancer or prostatitis, and the
transrectal or transperitoneal delivery of the compositions of the present invention to the
prostate to treat prostatitis.
The compostions of the present invention are suitably locally delivered to the
urinary tract before, during and/or after (preferably periprocedurally) procedures that
standardly include irrigation, such as TURP, transurethral incision of the prostate (TUIP),
laser prostatectomy, cystoscopy, ureteroscopy and other procedures in which irrigation is
used to aid visualization by removing blood and tissue debris from the operative field.
The compositions of the present invention can be added to the irrigation solution
standardly used in such procedures, e.g., saline, distilled water, lactated Ringer's solution,
-20-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

glycine, sorbitol, manitol, sorbital/manitol, at dilute levels, with no change to the
urologist's standard procedure being required.
The compositions of the present invention can also be locally delivered by coating
ureteral stents, uretheral stents, catheters, radioactive seeds, seed spacers or other
implantable devices or surgical instilments, or impregnating or otherwise incorporating
the therapeutic agents into the body of stents, catheters, radioactive seeds, seed spacers or
other implantable devices or surgical instruments constructed from a polymeric material
or mesh. Techniques for coating devices with drugs and impregnating devices with drugs
are well known to those of ordinary skill in the art, and coatings or polymeric materials
may be designed to permit the drugs (e.g., a COX inhibitor and a calcium channel
antagonist) to begin releasing into the urinary tract upon implantation and continuing for a
period of time following implantation.
Formulation
One aspect of the invention is directed to a composition including a
cyclooxygenase inhibitor and a calcium channel antagonist, preferably ketoprofen and
nifedipine, which are dissolved in an aqueous solution for parenteral delivery, preferably
for intravesicular delivery. Alternately such compositions can be manufactured in a
lyophilized form and then reconstituted with an aqueous solvent prior to administration.
The cyclooxygenase inhibitor and calcium channel antagonist are suitably
included in a molar ratio (cyclooxygenase inhibitoncalcium channel antagonist) of from
10:1 to 1:10, preferably from 5:1 to 1:5, more preferably from 4:1 to 1:1, and most
preferably 3:1. Similarly, in a preferred composition ketoprofen and nifedipine are
suitably included in a molar ratio (ketoprofen:nifedipine) of from 10:1 to 1:10, preferably
from 5:1 to 1:5, more preferably from 4:1 to 1:1, and most preferably approximately (i.e.,
+/-20%)3:l.
For compositions formulated to be delivered locally in a liquid carrier, the
cyclooxygenase inhibitor such as ketoprofen is suitably included at a concentration (as
diluted for local delivery) of no more than 500,000 nanomolar, preferably no more than
300,000 nanomolar, more preferably no more than 100,000 nanomolar and most
preferably less than 50,000 nanomolar. The calcium channel antagonist such as
nifedipine is suitably included at a concentration (as diluted for local delivery) of no more
than 200,000 nanomolar, preferably no more than 100,000 nanomolar, more preferably no
more than 50,000 nanomolar and most preferably less than 25,000 nanomolar.
The compositions of the present invention may be formulated in an aqueous or
organic solvent, but preferably are formulated in an aqueous solvent. When using
aqueous solutions, an additional solvent or solvents (i.e., cosolvents or solubilizing
agents) may suitably be included to aid in dissolution of the drugs. Examples of suitable
-21-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

solvents include polyethylene glycol (PEG) of various molecular weights (e.g., PEG 200,
300, 400, 540, 600, 900, 1000, 1450, 1540, 2000, 3000, 3350, 4000, 4600, 6000, 8000,
20,000, 35,000), propylene glycol, glycerin, ethyl alcohol, oils, ethyl oleate, benzyl
benzoate, and dimethyl sulfoxide (DMSO). A preferred cosolvent for the compositions of
the present invention is PEG, most preferably PEG 400.
In a further aspect of the present invention, the composition includes ketoprofen
and nifedipine in an aqueous solution including at least one stabilizing agent. The term
stabilizing agent is used herein to refer to an agent that inhibits degradation of the active
pharmaceutical ingredients and/or extends the duration of stability of the solution when
stored under either refrigerated (e.g., 2-8°C) or ambient temperature conditions, and
includes both anti-oxidants and chelating agents. The solution may also suitably include
one or more cosolvents or buffering agents. Preferably the aqueous ketoprofen and
nifedipine solution includes one or more antioxidants as stabilizing agent(s), a cosolvent
and a buffering agent. The preferred ketoprofen and nifedipine solution formulation is
stable when stored at between 2°C and 25°C for a period of at least six months, preferably
one year, more preferably two years, most preferably longer than two years, and can be
readily diluted with standard urologic irrigation solutions for local intravesicular delivery
during urological procedures.
Examples of suitable antioxidants for use as stabilizing agents in the compositions
of the present invention include water soluble antioxidants such as sodium bisulfite,
sodium sulfite, sodium metabisulfite, sodium thiosulfate, sodium formaldehyde
sulphoxylate, ascorbic acid, acetylcysteine, cysteine, thioglycerol, thioglycollic acid,
thiolactic acid, thiourea, dithithreitol, and glutathione, or oil soluble antioxidants such as
propyl gallate, butylated hydroxyanisole, butylated hydroxytoluene, ascorbyl palmitate,
nordihydroguaiaretic acid and a-tocopherol. A preferred stabilizing agent for the present
invention is propyl gallate. When included in an aqueous compositon, a cosolvent is
included solubilizing oil soluble antioxidants such as propyl gallate. A preferred aqueous
ketoprofen and nifedipine composition of the present invention includes PEG 400 as a
cosolvent and propyl gallate as a stabilizing agent, and may more preferably also include
a second stabilizing agent such as a water soluble antioxidant, most preferably sodium
metabisulfite. A suitable range of concentrations for antioxidant(s) is typically about
0.001% to about 5%, preferably about 0.002% to about 1.0%, and more preferably about
0.01% to about 0.5%, by weight of the composition.
Because of the involvement of divalent cations in catalyzing oxidation reactions,
the inclusion of a chelating agent as a stabilizing agent may be useful in the compositions
of the present invention. Examples of suitable chelating agents for use in the
compositions of the present invention include the various salts of ethylenediarnine
-22-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

tetraacetic acid salts (EDTA), β-hydroxyethylenediaminetriacetic acid (HEDTA),
diethylenetriamine-pentaacetic acid (DTPA) and nitrilotriacetate (NTA).
The compositions of the present invention suitably include a buffering agent to
maintain pH. Examples of suitable buffering agents for inclusion in the compositions of
the present invention include acetic acid and its salts, citric acid and its salts, glutamic
acid and its salt and phosphoric acid and its salts. Citric acid also has the ability to
chelate divalent cations and can thus also prevent oxidation, thereby serving two
functions as both a buffering agent and an antioxidant stabilizing agent. A preferred
aqueous ketoprofen and nifedipine composition of the present invention includes citric
acid (such as in the form of sodium citrate) as a buffering agent and antioxidant, and in a
more preferred composition also includes PEG 400 as a cosolvent and propyl gallate and
sodium metabisulfite as stabilizing agents.
The compositions of the present invention may also include additional excipients
and adjuvants. Excipients may include a preservative to protect against microbial growth,
especially for multiple-dose containers. Suitable excipients include antimicrobial agents
such as benzyl alcohol, chlorobutanol, thimiserol, methyl paraben and propyl paraben.
Excipients may also include a surfactant to reduce surface tension and thereby facilitate
wetting for dissolution. Examples of suitable surfactants include polyoxyethylene
sorbitan monooleate and sorbitan monooleate. Excipients may also include tonicity
adjustment agents to render the solution iso-osmotic with physiologic fluids. Examples
of suitable tonicity agents include sodium chloride, sodium sulfate, mannitol, glucose,
sucrose, trehalose, and sorbitol. Additional excipients may include a colorant to impart
color, such as FD& C No. 1 blue dye, FD&C No. 4 red dye, red ferric oxide, yellow ferric
oxide, titanium dioxide, carbon black, and indigo tar pigments.
-23-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Table 1
Exemplary Ketoprofen/Nifedipine Composition for Deliveryto the Urinary Tract
("Stock Solution Concentrations Prior to Dilution)

Ingredient Function Exemplary Concentration/Amount
Ketoprofen COX inhibitor 7.63 mg/ml (30 mM)
Nifedipine CA channel antag. 3.46mg/ml(10mM)
Sodium citrate
aqueous solution' Buffered solvent 20 mM solution (pH 6.2 ± 0.5)
PEG 400 Solubilizing agent
(cosolvent) 60% PEG 400:40% Sodium citrate soln.
(v:v)
Sodium metabisulfite Antioxidant
(stabilizer) 0.02%
Propyl gallate Antioxidant
(stabilizer) 0.01%
The above concentrated solution is diluted, such as at a ratio of 1:1,000 (v:v) with
standard irrigation solution such as saline or lactated Ringer's solution. The final dilute
solution from the above exemplary formulation thus includes 0.06% PEG40, 0.00005%
sodium metabisulfite and 0.00001% propyl gallate (all by volume). The active
ingredients are present in the final dilute solution at concentrations of 0.00763 mg/ml
(30,000 nM) for ketoprofen and 0.00346 mg/ml (10,000 nM) for nifedipine.
EXAMPLES
The present invention may be illustrated by the following studies demonstrating
the effects of ketoprofen and other cyclooxygenase inhibitors, nifedipine and
combinations of these agents in urological models, and demonstrating the stability of
certain formulations of such compositions.
Example I
The Effect of COX Inhibitors on Bradykinin Induced PGE2 Production in Rat Bladders
The following studies evidence that bradykinin induces immediate
prostaglandin E2 (PGE2) production in the bladder, and demonstrate the effects of
cyclooxygenase inhibitors on this process. Bradykinin was chosen as the activating
agonist for testing in this system because its actions on the rat bladder tissue system have
been well characterized and because its role as a proinflammatory agent in acute
pathophysiology has been studied. Bradykinin is also known to stimulate contraction of
-24-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

smooth muscle of the bladder when delivered intravesically by activation of Bl and B2
receptor subtypes.
1. Introduction
Acute, localized inflammatory responses in the lower urinary tract, including
spasm, are triggered by surgical trauma. In response to tissue injury, multiple
inflammatory mediators, including bradykinin and Substance P (SP) are released into the
bladder. Exogenous application of these pro-inflammatory peptides or activation of
bladder nerves can trigger the production of prostaglandins (PGs) in the bladder. The aim
of this study was to characterize the time course of production of PGs in response to an
inflammatory mediator and evaluate the effects of COX-1/COX-2 inhibitors on bladder
tissue contractility in vitro and in vivo. The rat bladder tissue strip system represents a
well established system for characterization of the pharmacological actions on numerous
agents on smooth muscle bladder contractility [Edwards, G., et al., Comparison of the
Effects of Several Potassium-Channel Openers on Rat Bladder and Rat Portal Vein In
Vitro, Br. J. Pharmacol. 102:679-80 (1991); Birder, L., et al., β-adrenoceptor Agonists
Stimulate Endothelial Nitric Oxide Synthase in Rat Urinary Bladder Urothelial Cells, J.
Neurosci. 22:8063-70 (2002)].
2. Bladder Strip Contractility
Method
Isolated bladder smooth muscle strips of 1x2x15 mm dimension were obtained
from Wistar derived male or female rats weighing 275 + 25 g that were sacrificed by CO2
overexposure. Each strip was placed under 1 g tension in a 10 ml bath containing Krebs
solution withl µM enalaprilic acid (MK-422), composition (g/1): NaCl 6.9, KC1 0.35,
KH2PO4 0.16, NaHCO3 2.1, CaCl2 0.28, MgSO 4.7, H2O 0.29, (+)Glucose 1.8, pH 7.4
bubbled with 95% O2/5% CO2 at 32 C. Each strip was connected to an isometric
transducer (Harvard, # 50-7293) and two-pen recorder and allowed to equilibrate for 60
minutes. Before starting the experiment, mounted tissues were validated for acceptance
by challenge with 100 µM of methoxamine to obtain a minimum of 1 g tension, which
was considered as 100%. Qualified tissues were washed repeatedly every 15 minutes for
60 minutes. A cumulative contraction-response curve to bradykinin was then generated
through application of 3 concentrations of bradykinin (0.01 µM, 0.1 µM and 1 µM) at 1
minute intervals for a total of 3 minutes. The tissue was subsequently washed
periodically until tension returned to baseline value. Two hours later, the ability to inhibit
the bradykinin cumulative dose response (0.01 µM, 0.1 µM and 1 µM) after a 10 minute
pretreatment with ketoprofen was determined. Each concentration of test substance was
tested in four separate preparations.
-25-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Results
FIGURE 2 illustrates the cumulative concentration-response curves of normal
animals to the agonists bradykinin and SP. The EC50 for bradykinin was 8.5 nM and for
SP was 6.5 nM. This provided a validated system for testing the effects of the inhibitory
activity of NSAIDs (COX inhibitors).
FIGURE 3A illustrates bradykinin concentration-response curves produced in the
presence of 0.25, 1.0, 2.5' and 10 µM ketoprofen. The maximal agonist response could
not be determined experimentally for all concentrations of ketoprofen, although curve-
fitting using a standard Hill equation revealed no change in maximal response at
saturating agonist concentrations. Schild analysis was used to calculate the pA2 value of
7.26 for ketoprofen, equivalent to a KD for ketoprofen at its site of action of 5.52 x 10-8 M
(see FIGURE 3B). This finding demonstrates that the potency for inhibition in this tissue
assay system is quite comparable to values obtained from direct enzyme inhibition assays.
3. PGE2 Determination:
Methods
The release of PGE2 from urinary bladder strips into 10 ml of tissue bath was
measured using a specific enzyme immunoassay (EIA) according to the manufacturer's
instructions (Amersham Pharmacia Biotech) for the basal, bradykinin-induced and COX
inhibitor treatment plus bradykinin-induced samples. The COX inhibitors tested were
ketoprofen, flurbiprofen, 5-bromo-2-(4-fluorophenyl)-3-(4-methylsulfonyl) thiophene
(i.e., DUP-697) and 1-[(4-methysufonyl)phenyl]-3-tri-fluoromethyl-5-(4-
fluorophenyl)pyrazole (i.e., SC-58125). One mL of fluid was collected from the 10 mL
tissue bath after 10 minutes of bradykinin challenge for PGE2 determination. Samples
were frozen immediately and stored at -4°C until assay. The bladder strips were dried
gently by blotting and were then weighed. Results are expressed as picograms of PGE2
released per milligram tissue.
Results
FIGURE 4A illustrates that bradykinin rapidly induces the formation of PGE2 in
rat bladder tissue strips within the first minutes of stimulation and reaches a maximum
within 30 minutes. The t1/2 for formation was about 7.5 minutes. FIGURE 4B illustrates
the rapid kinetics of PGE2 formation detected within the first ten minutes.
Ketoprofen inhibition of bradykinin-induced bladder strip contraction was closely
correlated with inhibition of PGE2 formation, as shown in Table 2. Non-selective COX-
l/COX-2 inhibitors were found to be effective in blocking bradykinin-stimulated PGE2,
-26-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

while COX-2 selective agents were not effective. This corresponds to a lack of COX-2
inhibitor activity under bradykinin-induced normal cystometry parameters.
Table 2
Inhibition of Bradykinin (BK)-induced Contraction with COX Inhibitors

Drug BK-induced Contraction
IC50 (µM) BK-induced PGE2
IC50 (µM)
Ketoprofen 0.97 0.58
Flurbiprofen 24.8 1.65
DUP-697 >25 >25
SC-58125 >25 >25

FIGURE 1 (described previously) provides a model for action of prostaglandin
activity. Activation of bradykinin receptors on urothelial cells may produce PGs in the
urothelium, which in turn may activate bladder nerves (C-fiber and A5 fibers) to affect
bladder contractility and control micturition reflexes. Ketoprofen inhibits formation of
PGE2.
4. In Vivo Rat Cystometry Model
Methods
The rats were anesthetized with urethane at 1.2 g/kg i.p. in 5 ml/kg. A
polyethylene catheter (PE50) was implanted into the bladder for saline or acetic acid
infusion through a 3-way stopcock. A pressure transducer was connected for
measurements of intravesical pressure. Warm (37°C) saline was infused into the bladder
at a constant rate of 16.7 ml/min (1 ml/hour) until cystometry became stable (no less than
60 minutes). Thereafter, 0.2% acetic acid was infused into the urinary bladder. Aspirin
(10 mg/kg i.v.) and vehicle were administered intravenously via a PE-10 catheter in the
femoral vein at 5 minutes after infusion of acetic acid was started and at the end of first
micturition cycle. Dunnett's test was applied for comparison between the time before and
after test substance or vehicle treatment. To ascertain differences between the test
substance and the vehicle control group, an unpaired Student's t test was used.
Differences are considered significant at p Results
FIGURE 5A illustrates that intravenous aspirin (10 mg/kg) produced a gradual
time-dependent inhibition of the acetic acid induced reduction in the intercontraction
interval (ICI), and FIGURE 5B illustrates the parallel changes in bladder capacity.
-27-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Threshold pressure and micturition pressure were not affected by aspirin treatment (data
not shown).
6. Discussion
These studies demonstrate that PGE2 is rapidly produced in rat bladder tissue
following stimulation with bradykinin and that its formation is inhibited by a 10 minute
pre-incubation with ketoprofen. Non-selective COX-1/COX-2 inhibitors were
demonstrated simultaneously to have blocked the rapid production of PGs in bladder
tissue and tissue contractility. Aspirin and other non-selective COX-1/COX-2 inhibitors
effectively inhibited cystometric changes induced by intravesical acetic acid stimulation.
These studies suggest that delivery of ketoprofen to the urinary tract may be
therapeutically beneficial for periprocedural bladder hyperactivity.
Example II
Effects of Ketoprofen and Nifedipine Individually on
Bradykinin Induced Contractility in Rat Bladder Tissue Strips
The purpose of this study was to characterize the effects of ketoprofen, a non-
selective COX-1/COX-2 inhibitor, and nifedipine, an L-type Ca2+ channel antagonist, on
agonist-stimulated rat bladder contractility using bradykinin as a stimulating agonist.
1. Methods
Ketoprofen USP and nifedipine USP were dissolved in DMSO prior to dilution to
the final concentration. Bladder tissue strips from Wistar derived rats were prepared,
transduced and equilibrated using the bladder strip contractility method described in
Example I above. Assayed tissue was incubated with the test drugs for 10 minutes before
activities were determined.
A cumulative contraction-response curve to bradykinin was generated through
application of 7 bradykinin concentrations in 3-fold increments ranging from 0.001 µM to
1 µM at 1 minute intervals for a total of 7 minutes to establish the maximal 100% control
response. The tissue was subsequently washed periodically until tension returned to
baseline value. In 24 separate tissues, similar bradykinin concentration-responses were
carried out in the presence of each respective test compound (ketoprofen: 0.25 µM, 1
µM, 2.5 µM and 10 µM; nifedipine: 0.125 µM, 0.5 µM, 1.25 µM and 5 µM) following a
10 min incubation period. Tissue strips were always used in pairs for the study of the
action of the antagonist (bradykinin) alone and in the presence of a concentration of
antagonist (ketoprofen or nifedipine). Schild plots were obtained using computer
software (Pharmacology Cumulative System, Version 4) and pA2 values were determined.
-28-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

2. Results
Nifedipine was found to exhibit a noncompetitive type of antagonism upon
bradykinin-induced contractile responses in the in vitro rat bladder preparation. This was
shown by a depression of the maximum agonist response and a small non-parallel
rightward shift of the agonist concentration response curves (FIGURE 6). In contrast, as
previously described in Example I, increasing concentrations of ketoprofen (0.25-10 µM)
produced a series of concentration-response curves (see FIGURE 3A) in which the EC50
agonist response moved progressively to higher concentrations of bradykinin (shift to the
right of over 2 orders of magnitude) with no apparent effect on maximal tension. This
pattern of inhibition is consistent with a competitive mechanism for ketoprofen and was
further analyzed by Schild regression analysis.
For nifedipine, the criteria for application of the Schild regression analysis were
not met due to the noncompetitive pattern of inhibition. Even the lowest concentration of
nifedipine (0.125 µM) resulted in a large reduction in the agonist response (to about 50%
of maximum). These studies of ketoprofen and nifedipine reveal two very different
patterns of inhibition of bradykinin-stimulated contractile tension.
Example III
Effects of Ketoprofen and Nifedipine Combination on
Bradykinin Induced Contractility in Rat Bladder Tissue Strips
The present study evaluated the effects of nifedipine and ketoprofen administered
in combination on the contractile tension response in a rat bladder tissue strip model.
1. Methods
Bladder tissue strips from Wistar derived rats were prepared, transduced and
equilibrated using the bladder strip contractility method described in Example I above
with transduced strips being allowed to equilibrate for 45 minutes. In order to avoid
effects of bradykinin receptor desensitization from the cumulative dosing protocol, two
tissue strips were collected from each animal. The control group consisted of 12 strips and
54 strips were used for the treatment groups.
Before starting the experiment, each pair of tissue strips was qualified by treating
with 0.03 µM bradykinin to determine if the initial difference in maximal contraction
between strips was within +/-15%. Following this procedure, qualified tissues were
washed repeatedly every 15 minutes for 60 minutes. Cumulative concentration-response
curves were generated by application of bradykinin to establish maximal response. For
the control group (n = 12), a cumulative concentration-response curve to bradykinin was
then generated through application of nine concentrations from 0.1 nM to 1.0 µM in 3-
-29-

fold steps at one minute intervals, for a total of nine minutes to establish the maximal
100% control response. Response curves for the treatment groups involved pre-
incubation of the bladder tissue for a period of ten minutes (n = 6), followed by
generation of bradykinin cumulative dose-response curves by application of 12
concentrations of bradykinin (0.1 nM - 30 µM).
The concentration range that was chosen for each of the active agents was based
upon results from prior in vitro pharmacological studies of each single agent described in
Examples I and II above. Those studies showed that ketoprofen in the 0.3-3 µM range
had measurable effects on the EC50 for bradykinin activation. Ketoprofen at 3 µM was
near maximal in its ability to shift the EC50 of the bradykinin activated response curves on
muscle contractility. Similarly, prior testing of nifedipine identified a range of
concentrations (0.05-5 µM) effective at inhibiting bradykinin induced tension. A factorial
design characterized the effects of nine different two-drug combinations of ketoprofen
and nifedipine at the following concentrations of (i) ketoprofen: 0.3, 1.0, or 3.0 µM; and
(ii) nifedipine: 0.1, 0.3 or 1.0 µM. The treatment groups (groups 2-10) tested are
summarized in Table 3 below:
Table 3
Ketoprofen-Nifedipine Combinations Tested

Group Ketoprofen Cone. (µM) Nifedipine Cone. (µM)
1 (Control) - -
2 0.3 0.1
3 0.3 0.3
4 0.3 1.0
5 1.0 0.1
6 1.0 0.3
7 1.0 1.0
8 3.0 0.1
9 3.0 0.3
10 3.0 1.0
The bradykinin concentration-response data was fit to a variable slope sigmoidal
equation, also known as the 3-parameter logistic response (3PL) function, to obtain the
maximal tension, EC50, and Hill slope in which the bottom of the curve was fixed at 0.
-30-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

The force of contraction in the presence of inhibitors was expressed as a percentage of the
maximum bradykinin effects observed within the same strip before addition of an
inhibitor.
2. Results
The experimental data for all curves allowed curve fitting to accurately define the
maximal tension and EC50 values. The control curve in FIGURE 7 showed that BK
concentration-dependently increased the force of contraction with a pEC50 of 8.14 or 72
nM (n = 12 strips). A moderate Hill slope of 0.65 characterized the activation curve. All
further contraction data was expressed as a percentage of the maximum bradykinin effect
obtained from a set of 12 tissue strips without any tension and without any antagonist
present.
The results for nine distinct combinations of nifedipine and ketoprofen used to
inhibit bradykinin-induced bladder contraction are shown in the following three tables
and three figures. At the lowest concentration of nifedipine and ketoprofen tested, 0.1
µM and 0.3 µM respectively, 38% reduction of the maximal control tension was observed
(Table 4). Increasing concentrations of ketoprofen (1.0 and 3.0 µM) in the presence of
the same concentration of nifedipine further decreased the maximal contractile tension
such that only 30 and 23.4% of the control tension remained, respectively. All
concentration-response curves for bradykinin shifted to the right in the presence of
nifedipine and ketoprofen (0.3-3.0 µM), with the greatest effect seen at the highest
ketoprofen concentration. This combination was accompanied by a 1.0 log unit shift in
the pEC50 versus control. The changes in the EC50 parameter did not appear correlated
with changes in maximal tension. The results are presented graphically in FIGURE 7,
which compares the control group and the group having a constant concentration of 0.1
µM nifedipine with a range of concentrations of ketoprofen. The percent of contraction
for each drug combination is expressed as the percent of the maximal response for the
bradykinin control. The overall pattern of inhibition predominantly reflects a substantial
decrease in maximal tension, demonstrating that the combination of nifedipine and
ketoprofen act together in combination mechanistically in a non-competitive antagonist
manner towards bradykinin-induced contractions.
-31-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Table 4
Concentration-Response Curve Fitted Parameters
for 0.1 µM Nifedipine (NIF) plus 0.3-3.0 µM Ketoprofen (KET)

SEM = Standard error of the mean
Tmax = Maximal tension determined by curve fitting
In the presence of 0.3 µM nifedipine, increasing concentrations of ketoprofen
present in the combination treatment resulted in a progressive decrease in the maximal
tension, from 36.4 to 16.0%. Combinations utilizing the higher concentration of
nifedipine (0.3 µM) resulted in a greater reduction in the maximal tension relative to the
corresponding concentrations of ketoprofen in the presence of 0.1 µM nifedipine. The
maximal tension levels for 0.3 µM nifedipine combinations were determined for three
combinations, in concentration ratios of nifedipine:ketoprofen of 1:1, 1:3.3 and 1:10. The
curve fitted parameters data obtained are presented in Table 5.
Comparison to data corresponding to ketoprofen concentrations in Table 4 shows
that in all cases, greater reductions in the maximal tension were associated with the
greater nifedipine concentration. The greatest change was evident at the lowest
ketoprofen concentration, 0.3 µM, which decreased from 62.11 to 36.41%. The higher
concentrations of ketoprofen resulted in an even greater reduction in tension, such that
only 16% remained at 3.0 µM. Associated with these changes in tension, a similar shift
in the EC50 relative to the control of 0.5 log units was evident for all ketoprofen
concentrations at this nifedipine concentration, as can be seen in FIGURE 8. As in the
case of 0.1 µM nifedipine, no apparent differences between the EC50 values for this
concentration of nifedipine were evident. Small differences in the Hill slopes for
bradykinin agonist responses over the range of inhibitor concentrations were not
significant. The effect of increasing ketoprofen concentrations in the combination
treatment on the concentration-response curves is similar to the graph of the data at
0.1 µM nifedipine and various ketoprofen concentrations. These graphical data also show

the non-competitive nature of the antagonism of the BK-response, which is seen for the
combination at this higher concentration of nifedipine.
Table 5
Concentration-Response Curve Fitted Parameters
for 0.3 µM Nifedipine (NIF) plus 0.3-3.0 µM Ketoprofen (KET)

Est. = Estimated
SEM = Standard error of the mean
Tmax = Maximal tension determined by curve fitting
The overall shapes of the response curves observed in the presence of 1.0 µM
nifedipine were similar at all concentrations of ketoprofen. At 1.0 µM nifedipine, the
maximal tension levels were less than the corresponding values for 0.3 µM nifedipine
(Table 6 and FIGURE 9), and the magnitude of the additional change due to the presence
of ketoprofen is less relative to lower concentrations of nifedipine. The EC50 values were
uniformly shifted about 0.51 units for all ketoprofen concentrations and were not
correlated with maximal tension. This pattern was consistent with observations at all
other combination concentrations. A small additional increase in the inhibition of
maximal tension due to the change from 1.0 to 3.0 µM ketoprofen was observed at this
highest concentration of nifedipine. At the highest concentrations (1.0 µM nifedipine
plus 3.0 µM ketoprofen), 89% inhibition of the control tension level was achieved.
-33-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Est. = Estimated
SEM = Standard error of the mean
Tmax = Maximal tension determined by curve fitting
3. Response Surface Analysis
The concentrations of the two agents (nifedipine and ketoprofen) used in this
combination experiment represent independent variables. The maximal tension is an
effect that results from the combination and is the response variable of primary interest for
the response surface analysis. The relationship between the drug combinations and the
response variable can be represented in a three-dimensional plot in which the
concentrations are plotted as Cartesian coordinates in the x-y-plane, and the response
variable (e.g., maximal tension) is plotted as the vertical distance above the planar point.
The collection of spatial points plotted in this way provides a view that represents the
combined concentration-response relationship. The advantages of this experimental
design method include the fact that the biological response measured is not limited to a
specific response (effect) level of the system. In this way, a number of fixed-ratio
concentration combinations can be tested over a wide range of concentrations to define
the interaction efficacy of the two drugs.
As in the case of single drug concentration-biological effect relationships in which
a smooth curve (or line) may be best fit to the data according to a specific model, a
smooth surface may be fit to the data in a three-dimensional plot of a two-drug
combination concentration-response relationship. This surface represents the additivity or
interaction of the combination. The graph of this response surface becomes the reference
surface for viewing actual combination effects and allows the visualization and prediction
of effects in regions of the curve for which no data could be generated.
-34-
Table 6
Concentration-Response Curve Fitted Parameters
for 1.0 µM Nifedipine (NIF) plus 0.3-3.0 µM Ketoprofen (KET)

PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

A standard response surface analysis was performed on the estimated maximal
tension and EC50 values. The response surface model was fitted using as a response
variable the tension values at the highest agonist concentration on each individual dose
response curve, which was the tension corresponding to 30 µM BK. FIGURE 10 shows
the fitted response surface for the reduced model as a function of ketoprofen and
nifedipine concentration. The combination response curve drops steeply with increasing
concentrations of both ketoprofen and nifedipine. The surface becomes, fairly flat as the
maximal response is obtained as concentrations approach 1 µM nifedipine + 3 µM
ketoprofen. The concentration combination that results in 90% maximal inhibition of the
effect of bradykinin is 3 µM of ketoprofen + 1 µM of nifedipine.
4. Discussion
The study of this Example III evaluated the effects of nifedipine in combination
with ketoprofen using bradykinin as an agonist to stimulate smooth muscle contraction.
Bradykinin was used in the rat bladder tissue strip assay system (Examples I - III) to serve
as an endogenous mediator of contraction. The overall pattern of inhibition seen with all
combinations of nifedipine and ketoprofen concentrations was characteristic of non-
competitive antagonism. Nifedipine, which prevents the influx of calcium ions through
the cell membrane by acting on L-type voltage-dependent channels, attenuates the
bradykinin receptor activated contraction of smooth muscle without directly inhibiting the
receptor. As a single agent, nifedipine inhibition was shown above (Example II) to cause
a reduction in the maximum bradykinin responses that were not accompanied by
statistically significant changes in the agonist potency of the remaining response.
This study revealed the surprising finding that the magnitude of the inhibition is
greatly enhanced by the addition of ketoprofen at the lowest nifedipine concentration
tested and is evident at all concentrations of the combinations tested. At low
concentrations of nifedipine, this inhibition is more than additive, i.e., synergistic in
nature. In contrast, ketoprofen treatment alone at the same concentrations was observed
to not decrease maximal contractile tension, with no significant effect on the EC50 values
for the nine combinations and no concentration dependence upon ketoprofen. Thus, this
synergistic interaction on maximal tension and lack of strong effect upon the EC50 was
an unexpected result based on the study of ketoprofen action when tested as a single agent
in this test system.
Taken together, these data indicate that the effects of the proinflammatory agonist,
bradykinin, can be in part mediated by the simultaneous activation of L-type calcium
channels and the induction of arachidonic metabolites that together augment smooth
muscle contraction. While not wishing to be limited by theory, this effect may be due to a
positive feedback loop that operates at a cellular and tissue level. Prostaglandins
-35-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

generated intracellularly as a result of bractykinin receptor activation may move to the
extracellular environment, where they may interact and in turn activate prostanoid
receptors subtypes. There are at least four known prostanoid receptor subtypes, termed
EP1, EP2, EP3 and EP4. Of these subtypes, EP1 receptors are believed to be coupled
through G proteins to stimulation of phophoinositide hydrolysis and/or PLC-independent
influx of calcium. EP1 receptors have been previously identified in smooth muscle,
where they can function to mediate contractile activity. Hence, the discovery of the
combined synergistic actions of ketoprofen and nifedipine on contractile activity may be a
result of simultaneous blockade of calcium mobilization and the concurrent inhibition of
a positive-feedback loop involving PGE2 driven activation of prostanoid receptors.
In conclusion, each combination of nifedipine and ketoprofen showed a greater
inhibition of maximal bradykinin-induced contraction compared to either drug alone in
the rat bladder tissue strip assay. Furthermore, the multiple combinations of nifedipine
and ketoprofen tested allowed a response surface analysis to define optimal
concentrations. A fixed ratio combination containing 3.0 µM ketoprofen and 1.0 µM
nifedipine was identified that produced ~90% inhibition.
Example IV
Inhibition by Nifedipine and Ketoprofen of Multiple Agonist-Induced Contractile Tension
and Release of PGE2 in Rat Bladder Tissue
The objective of this study was to evaluate the effects of ketoprofen and nifedipine
on rat bladder contractility and agonist-stimulated PGE2 production using multiple
agonists. Bradykinin, substance P, histamine and ATP are endogenous mediators that can
be released as part of the acute inflammatory response and activate bradykinin receptors
(B1 and B2 subtypes), tachykinin receptors (NK1-3) and histamine receptors (all subtypes)
and purinergic P2X and P2Y receptors, respectively. Carbamylcholine is an agonist that
may activate muscle and neuronal nicotinic acetylcholine subtypes or muscarinic
acetylcholine receptors subtypes (M1-5) present in the bladder, while methoxamine is
specific for α1-adrenergic receptors. The first objective was to evaluate the effect of
ketoprofen (10 µM) and nifedipine (1 µM) individually, each at a fixed concentration, on
contractile tension induced by each of the six agonists (bradykinin, substance P,
carbamylcholine, methoxamine, histamine and ATP) in the rat bladder tissue strip model.
The second objective was to determine the amount of PGE2 released from the bladder
tissue in response to stimulation by each agonist in the presence of either ketoprofen or
nifedipine during the same test conditions employed to measure contractile smooth
muscle tension.
-36-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

1. Methods
Bladder tissue strips from Wistar derived rats were prepared, transduced and
equilibrated using the bladder strip contractility method described in Example I above.
Either 10 µM ketoprofen or 1.0 µM nifedipine was pre-incubated individually with the
tissue for a period of 10 minutes prior to stimulation with the following agonists at a
concentration equivalent to its respective ED75 for stimulation of tension: 0.03 µM
bradykinin; 0.03 µM substance P; 3.0 µM carbochol; 30 µM methoxamine; 25 µM
histamine; and 20 µM ATP. Antagonist activity for a given concentration of an
antagonist (nifedipine or ketoprofen) was determined as the ability of that concentration
of the antagonist to reduce the noted agonist-induced (e.g., 0.03 µM bradykinin-induced)
response by 50 percent or more (>50%). Each concentration of antagonist was tested in
four separate tissue preparations.
The effects of the two drugs on PGE2 release in response to multiple agonists was
compared using the same 10 min pre-incubation protocol and a subsequent 30 min
incubation period with agonist in the presence of the test compound. PGE2 produced
after 30 minutes of treatment with each agonist (e.g., 0.03 µM bradykinin) in the absence
and presence of the test compounds was determined. An initial 1.0 ml sample was taken
from the tissue bath after a 30 minute incubation with the agonist. Subsequently, the
tissue was washed using 10 ml of Krebs solution every 15 minutes for a 2 hour period.
The test compound was added and pre-incubated for a period of 10 minutes prior to re-
challenge with the same agonist. After an additional 30 minute period in the presence of
the test antagonist and agonist, 1.0 ml was removed from the bath for analysis. The
release of PGE2 from urinary bladder strips was measured using a specific enzyme
immunoassay (EIA). Samples were frozen immediately and stored at -4°C until assay.
The bladder strips were dried gently by blotting, and then weighed. Results are expressed
as picograms of PGE2 released per milligram tissue.
2. Results
All of the agonists investigated stimulated contraction of the bladder tissue strips,
independent of their mechanism of action, demonstrating that multiple mediators can
increase bladder smooth muscle contractile tension. Nifedipine (1 µM) produced a
significant inhibition (>67%) of each agonist-induced increase in contractile tension
(FIGURE 11). The contractile response to bradykinin was affected by both nifedipine and
ketoprofen (81% inhibition and 67% inhibition, respectively). In contrast, the increase in
contractile tension induced by substance P, carbamylcholine and ATP was not affected by
ketoprofen. Ketoprofen also only slightly reduced the tension for methoxamine and
histamine ( -37-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Bradykinin evoked the largest increase in PGE2 relative to the other agonists
tested. This evoked release was effectively inhibited by ketoprofen (81%) but minimally
affected by pre-treatment with nifedipine (12%) (FIGURE 12). Thus, the extent of
inhibition of smooth muscle tension by nifedipine was not linked to agonist-induced
PGE2 responses and was distinct from the effect of ketoprofen. The absolute bladder
levels of PGE2 produced in response to stimulation by other GPCR agonists were about
10-fold less than those seen with bradykinin.
In summary, the current study indicated that an increase in smooth muscle
contractile tension can be induced by a variety of GPCR agonists in bladder tissue.
Moreover, a common signaling mechanism for these agents is mediated in part through
activation of L-type Ca2+ channels in the rat urinary bladder. Nifedipine's inhibition of L-
type Ca2+ channels suggests an effective mechanism for inhibition of numerous
pathophysiological mediators that can lead to increased smooth muscle bladder tension
associated with spasm or hyperactivity. Ketoprofen inhibited bradykinin-stimulated PGE2
production and release from the bladder while nifedipine did not exhibit an effect on this
response. Thus, nifedipine and ketoprofen act through distinct mechanisms to inhibit
smooth muscle contractile tension and release of pro-inflammatory prostaglandins in
bladder tissue.
Example V
Effect of Ketoprofen and Nifedipine on Rat Bladder Function in an
Acetic-Acid Overactive Bladder Model
The primary objective of this study was to measure the effect of ketoprofen and
nifedipine during intravesical, local delivery to female rats with overactive bladder
function caused by perfusion with saline containing 0.2% acetic acid (acidified saline).
Perfusion of 0.2% acetic acid through the bladder is known to rapidly induce an acute
inflammatory state that is reflected in functional changes in bladder cystometry.
1. Methods
The method used in the current study represents an adaptation of a widely used
acetic acid-triggered rat model of hyperactive bladder. In this model, acute inflammation
of the bladder is produced by using 0.2% acetic acid in saline as the bladder perfusion
fluid and cystometry under anesthesia is performed after a recovery period from the
surgical procedure. A regular interval of voiding cycles can be seen for several hours
after the initial stabilization period occurs. A bladder catheter connected to an infusion
pump was used to deliver the drug solutions directly to the bladder at a constant, defined
rate.
-38-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

The animals were anesthesized and bladder catheters were surgically implanted to
allow irrigation of the test agents. The following cystometry parameters were monitored:
mtercontraction interval (ICI), trigger pressure (TP), micturition pressure (MP) and
micturition volume (MV) using a Med Associates Cystometry Station and software
program. Only rats that displayed normal and stable cystometry profiles during the
preliminary saline-infusion stage (not less than 15 min of baseline stabilization followed
by 7 regular representative ICI intervals) were included in the study. Following the saline
period, the rat bladder was infused with test agent in saline containing 0.2% acetic acid
for 20 min followed by the collection of 7 representative ICI intervals for analysis. Due
to the fixed concentrations of the irrigation solutions employed in the study and the use of
constant perfusion rates for fixed constant times, a fixed, uniform dose of each agent was
delivered to all animals.
Groups of female rats were administered ketoprofen at selected concentrations
(0.01-25 µM) alone or nifedipine at selected concentrations (0.1-10 µM) alone. Five to
seven animals were normally tested in each group. Acidified saline served as the control.
All infusion solutions were prepared fresh on the day of the experiment before use. For
each of the test agents, three distinct bladder irrigation periods were employed: 1)
baseline (saline only) for 1 hour; 2) drug in saline only for 15 minutes; and 3) drug in
0.2% acidified saline for 1 hour.
2. Results
In the control animals, baseline levels of bladder contractions in response to a
constant irrigation rate of 0.1 µl/min saline were established during the first hour
following surgery. The time between contractions (ICI, seconds) and peak micturition
pressure (MP, mm Hg) appeared to vary somewhat between animals but was fairly
constant within animals following stabilization. Following the addition of the 0.2%
acetic acid to the perfusion buffer, rapid contractions appeared, resulting in a significant
decrease in the ICI. Increases in contractile pressure accompanied the shortening of time
between the bladder contractions in many cases as well. These changes in functional
bladder responses could be routinely measured following perfusion of the bladder with
acidified saline, as shown in FIGURE 13. A 40-50% shortening of the ICI was typically
seen in the control group in response to the 0.2% acetic acid irrigation (mean % ICI =
58.4%±6.8%,n=8).
Inclusion of ketoprofen in the irrigation buffer leads to a concentration-dependent
inhibition of the shortening of the ICI (FIGURE 14). Complete inhibition was seen at
approximately 3 µM ketoprofen and higher concentrations tended to go above 100% (data
not shown).
-39-

Inclusion of nifedipine in the irrigation buffer also leads to a concentration-
dependent inhibition of the shortening of the ICI (FIGURE 15). Complete inhibition was
not seen but maximal effects appeared to be at 1 µM nifedipine, and higher
concentrations tended to plateau at approximately 75% of baseline.
Example VI
Pharmacokinetics of Absorption of a Nifedipine and Ketoprofen
Combination in a Rat Bladder Saline Model
The primary objective of this study was to measure systemic -plasma levels of
ketoprofen and nifedipine during and after the intravesical, local delivery of a
combination of these drugs to rats. A secondary objective of this study was to determine
the rate of appearance of ketoprofen and nifedipine when administered individually or in
combination. Finally, a third objective of this study was to evaluate the effects of local
drug delivery on the rat bladder tissue content of the pro-inflammatory mediator, PGE2,
following surgical trauma to the bladder and subsequent intravesical perfusion of each
agent or the combination.
/. Methods
The study included three main treatment groups of animals: a combination of both
ketoprofen (10 µM) and nifedipine (10 µM); ketoprofen (10 µM) alone; and nifedipine
(10 µM) alone. A bladder catheter connected to an infusion pump was used to deliver the
drug solutions directly to the bladder at a constant, defined rate.
For each of the three drug treatment groups, three distinct bladder irrigation
periods were employed that were defined by the bladder perfusion solution for each
period: 1) baseline (saline only) for 1 hour; 2) drug in saline only for 1 hour; and 3) post-
drug saline period for 30 minutes (min). The animals were anesthetized and the dome of
the bladders were surgically implanted with a catheter to allow perfusion of the test agents
with an infusion pump at a constant flow rate of 100 ul/min. During period 1, saline was
the perfusion fluid used and no plasma samples were collected. Starting at period 2,
plasma samples were collected at time points of 0, 15, 30, 45 and 60 min following
perfusion of test agents. Subsequent to 60 min of perfusion with test agents, only saline
was perfused for an additional 30 min and two additional time points at t = 75 and 90 min
were collected to determine the acute post-perfusion phase of test agents. Due to the
fixed concentrations of the irrigation solutions employed in the study and the use of
constant perfusion rates for fixed constant times, a fixed, uniform dose of each agent was
delivered to all animals.
Whole blood samples were collected into K2 EDTA tubes at the specified
collection times. The volume of whole blood collected was approximately 0.2 mL per
-40-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

sample. The blood was spun in a centrifuge and the plasma transferred into
polypropylene tubes. Plasma samples were stored frozen at -80°C until shipment for
analysis. Rats were euthanized by CO2 inhalation and the bladder was rapidly dissected
and frozen in liquid nitrogen and stored frozen at -80°C until assayed for tissue PGE2
content.
The combination of ketoprofen and nifedipine was formulated in accordance with
an aspect of the invention to include ketoprofen (10 mM), and nifedipine (10 mM) in a
60% polyethylene glycol 400 (PEG 400):40% water solvent base, including 50 mM
sodium citrate buffer for a pH 7.5 solution in a 5 mL glass vial. Immediately prior to use,
the combination solution was diluted in the standard irrigation fluid at a ratio of 1:1000
such that the final concentrations of the active drugs delivered directly to the bladder were
each 10 µM. For these experiments, a fixed concentration ratio of 1:1
nifedipine:ketoprofen was chosen, and final concentrations of 10 µM for each agent were
maintained in the irrigation buffer.
2. Results
The study demonstrated a very low level of systemic absorption of ketoprofen
following perfusion of the bladder with saline containing 10 µM ketoprofen for 60 min.
In four out of six rats, a narrow range of Cmax between 4.3-5.8 ng/ml was seen at 60 min.
At the 60 min time point, the perfusion with 10 µM ketoprofen was stopped and normal
saline irrigation was continued for an additional 30 min period. For the group of four out
of six rats which showed peak plasma levels of about 5 ng/ml, plasma levels decreased at
75 and 90 minutes following cessation of ketoprofen perfusion. Delayed absorption
during the 75-90 minute interval was observed in the other two animals in the ketoprofen-
only group.
For comparison, the ketoprofen levels were also determined for the combination
of ketoprofen and nifedipine. The increase in systemic plasma levels was approximately
linear over time during the initial 60 minute drug perfusion phase and the absolute mean
plasma levels of 9.3 ng/ml (n = 6) at 60 minutes were well below the acceptable
therapeutic daily dose of ketoprofen. As in the case of the ketoprofen only group, the
perfusion with the combination was stopped and normal saline irrigation was continued
for an additional 30 minute period. The mean ketoprofen values for all animals (n = 6)
were not significantly different at 60, 75 and 90 minutes.
A comparison of the mean plasma ketoprofen results are presented graphically in
FIGURE 16 for the ketoprofen-only group and the combination group. The mean values
(and standard error of the means, SEMs) clearly show the constant plasma levels for the
combination after 60 minutes. Although small differences are apparent in the earliest
phase of the time-course, no significant differences were observed either in the peak
-41-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

levels or in the absorption kinetics for the ketoprofen plasma levels in the combination
group versus the ketoprofen alone group after 30 minutes, or in the peak levels, indicating
that no apparent ketoproferi-nifedipine drug interactions were present.
The overall kinetic profile observed for nifedipine was similar to that observed for
ketoprofen. In the nifedipine-only plasma group, nifedipine plasma levels increased
linearly in 5/6 animals and some delayed absorption was observed in only 1/6 animals.
The Cmax plasma level in the nifedipine group was in the range of 10.6-16.0 ng/ml at 60
minutes for 5/6 animals. The mean peak plasma levels observed at 60 minutes were
below the acceptable mean peak levels of 79 + 44 ng/ml that are obtained in man as a
result of an oral therapeutic daily dose of nifedipine.
The increase in nifedipine systemic plasma levels from the combination of
ketoprofen and nifedipine also exhibited a linear increase with increasing time for the
initial 60 minute drug perfusion period. The Cmax plasma levels in the combination group
had a mean value of 18.2 ng/ml and values ranged from 8.2 - 34.6 ng/ml at 60 minutes for
all six animals. The mean peak plasma levels observed at 60 minutes are about one fourth
the mean peak levels that are obtained as a result of oral therapeutic daily dose of
nifedipine.
As shown in FIGURE 17, a comparison of the mean peak plasma concentrations
of nifedipine (and plotted SEMs) shows the similar linear increase that occurs during the
initial perfusion phase of intravesical delivery. No significant differences in nifedipine
plasma levels were seen in the nifedipine only group when compared with the nifedipine
and ketoprofen combination drug product group.
At the end of the 90 min bladder perfusion period, bladders were harvested from
the animals and subsequently the entire bladder was analyzed for PGE2 content using an
enzyme immunoassay system. Data shown in FIGURE 18 are expressed as the mean of
PGE2 using units of pg/mg protein + the standard error of the mean from six animals per
treatment group. When animals were treated with nifedipine, bladder tissue PGE2 levels
of 421 + 97 pg/mg protein (n = 6) were observed compared to statistically significantly
(p the combination treatment group, 83 + 22 (n = 6) and 115 + 63 pg/mg (n = 5),
respectively. No statistically significant differences were seen between the ketoprofen
treatment or the combination treatment groups. In summary, ketoprofen treatment alone
or treatment with the combination during bladder perfusion significantly inhibited PGE2
formed in the whole bladder relative to the nifedipine treatment group.
3. Discussion
Using a method of intravesical perfusion for local drug delivery, the drugs tested
in this study were directly in contact with the absorptive site within the bladder. The
-42-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

continuous perfusion maintained constant drug concentrations of either ketoprofen,
nifedipine or the combination within the bladder during the period of drug delivery.
Under these conditions, minimal systemic exposure to the drugs occurred in female rats
during a 1 hour intravesical perfusion. Low levels of each drug were detectable within
the first 15 min interval measured, and absorption progressed gradually as an
approximately linear function over time of drug perfusion for each agent.
The locally delivered drugs and drug combination were exposed to the structures
of the bladder, including the uroepithelium, C-fiber afferents, efferents and smooth
muscle. The data obtained in the study show that this action is local and cannot be
ascribed to systemic effect that could be mediated through central nervous system
mechanisms because the initial levels in the plasma for both drags tested are so low.
Comparison of the plasma levels for each agent tested to known human levels
associated with normal oral dosing reveals the magnitude of the difference observed. In
the combination treatment group, the maximal levels for ketoprofen were about 400-fold
less than the peak plasma levels (Cmax) in humans that are associated with the acceptable
therapeutic daily dose of ketoprofen (rat mean ketoprofen plasma level of 9.29 + 2.13
ng/ml at 60 min). For comparison, the accepted daily mean peak Cmax for a single 200 mg
ketoprofen tablet (a single oral dose) is 3900 ng/ml. Similarly, peak levels observed for
nifedipine were approximately 15 ng/ml to 25 ng/ml. The maximal levels (Cmax) typically
occurred at the end of 60 min drug perfusion period or within the following 30 min
sampling period. For comparison with known plasma levels from conventional oral
dosing, the accepted daily Cmax for a single 10 mg immediate release nifedipine tablet is
reported to be 79 + 44 ng/ml. Systemic exposure was comparable for ketoprofen plasma
levels whether administered alone or with nifedipine. Similarly, nifedipine plasma levels
were comparable whether administered alone or with ketoprofen.
This study also determined the PGE2 content of the bladder for each of the
treatment conditions in the study. An additional finding of significance is the long-lasting
effect of ketoprofen that was measured in the assay of whole bladder PGE2 levels. The
low concentrations of the ketoprofen treatment alone or the combination treatment during
bladder perfusion significantly inhibited PGE2 formed in bladder tissue relative to the
nifedipine treatment group. The PGE2 bladder tissue levels in the presence of ketoprofen
were not significantly different from those following treatment with the combination.
Because delivery of the drug was stopped at 60 minutes in this study, and then saline was
used to irrigate for an additional 30 minutes, this showed that PGE2 inhibition remained
active in the post-drug delivery period. Thus, ketoprofen demonstrated an extended
period of anti-inflammatory activity in this model of local, intravesical drug delivery.
-43-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Example VII
The purpose of this study was to evaluate the solubility of ketoprofen and
nifedipine in aqueous liquid solution formulations.
1. Methods
Three ketoprofen and nifedipine combo liquid formulations, identified as F3/1,
F10/3, and F30/10, were prepared according to the composition shown in Table 8 below.
In all three test formulations, 50 mM sodium citrate aqueous buffer was used. The target
solubility of ketoprofen /nifidipine for F3/1, F10/3, and F30/10 were 3 mM /I mM, 10
mM /3 mM, and 30 mM /10 mM respectively.
Table 8
Solubility Results for Three Nifedipine and Ketoprofen Combination Formulations

2. Results
In order to achieve complete dissolution of both actives, ketoprofen and
nifedipine, in the formulatins, different percentages of PEG 400, 35% v/v PEG 400
(F3/1), 50% v/v PEG 400 (F10/3), 60% v/v PEG 400 (F30/10), were used as a cosolvent.
With the assistance of PEG 400 as a solubilizing agent, the approximate saturation
solubility of ketoprofen and nifedipine in all three formulations was approximately 1.5x
of their respective target solubility. The solubility results in Table 8 clearly indicate that
PEG 400 is a suitable solubility enhancing agent for both drugs when it is desired to
prepare highly concentrated combination solution formulations.
-44-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

Example VIII
The purpose of this study was to evaluate the stability of exemplary combination
ketoprofen and nifedipine aqueous liquid solution formulations.
1. Methods
Four exemplary ketoprofen and nifedipine combination solution formulations,
identified as F1 to F4, were prepared according to the composition shown in Table 9. In
all four formulations, the concentrations of the active drugs were 3 mM for Ketoprofen
and 1 mM for Nifedipine. All four formulations employed sodium citrate buffer (pH 5.5)
with a 35% v/v of PEG 400. The ionic strength of the buffer used was 50 mM for F1 and
F2, and 20 mM for F3 and F4. No antioxidant was added to the virgin formulation F1,
while 0.05% propyl gallate, 0.02% sodium metabisulfite, and 0.05% propyl gallate plus
0.02% sodium metabisulfite were added to the combination formulations F2, F3, and F4
respectively.
Table 9
Tested Nifedipine and Ketoprofen Combination Formulations

Formulation
ID Drug Concentration
(Ketoprofen/Nifedipine) Formulation
Vehicle Antioxidants
Added
F1 3 mM /1 mM 50 mM NaCitrate
(pH 5.5)w/35%
v/v PEG 400 None
F2 3 mM /1 mM 50 mM NaCitrate
(pH 5.5)w/35%
v/v PEG 400 0.05% Propyl
gallate
F3 3 mM /1 mM 20 mM NaCitrate
(pH 5.5)w/35%
v/v PEG 400 0.02% Sodium
metabisulfite
F4 3 mM /1 mM 20 mM NaCitrate
(pH 5.5)w/35%
v/v PEG 400 0.05% Propyl
gallate
& 0.02% Sodium
metabisulfite
An isocratic high performance liquid chromatograph (HPLC) method was used to
quantify ketoprofen and nifedipine and their related substances in these test solution
formulations after storage for different periods of time. Formulation samples were taken
and diluted into mobile phase to obtain a final concentration of approximately 0.76
-45-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

mg/mL to 2.54 mg/mL for ketoprofen and approximately 0.35 mg/mL to about 1.15
mg/mL for nifedipine. Chromatographic conditions for the related substances assay were
as follows: (1) detection wave length: UV 241 nm; (2) column: Zorbax SB-C18, 5 µM,
4.6 x 150 mm; (3) column temperature: 30 ± 1 °C; (4) flow rate: 1.0 mL/min; (5) injection
volume: 20 µL; (6) run time: 27 minutes.
2. Results
FIGURE 19 shows an example chromatogram of the combination solution
formulation F1 after stressing by storing at 60 °C for 1 month. The two active
ingredients, ketoprofen and nifedipine, have a retention time of 24.19 minutes and 19.31
minutes respectively. There are four main related substances with relative retention times
(RRT) of 0.34, 0.58, 0.75, and 0.87 relative to the nifedipine peak. This stability data is
summarized in Table 10.
Table 10
Total related Substance (%) of Ketoprofen and Nifedipine in Tested Formulations
After Storage at Different Temperatures

The stability data in Table 10 indicates that the chemical stability of ketoprofen
and nefidipine, especially nifedipine, is significantly improved in the presence of a small
amount of either propyl gallate (0.05% w/v) or sodium metabisulfite (0.02% w/v) at
elevated temperatures such as 40°C and 60°C, with this effect being unexpectedly
pronounced for propyl gallate. When a small quantity of both propyl gallate (0.05% w/v)
and sodium metabisulfite (0.02% w/v) is added to F4, the stability of the two drugs at all
-46-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

temperatures is significantly improved when compared with the other three combination
formulations without antioxidant or with one of the two antioxidants alone, suggesting
additive or synergistic degradation inhibition effect of the two antioxidants.
While the preferred embodiment of the invention has been illustrated and
described, it will be appreciated that various changes to the disclosed solutions and
methods can be made therein without departing from the spirit and scope of the invention.
It is therefore intended that the scope of letters patent granted hereon be limited only by
the definitions of the appended claims.
-47-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

WE CLAIM:
1. A locally deliverable composition for inhibiting pain/inflammation
and spasm, comprising a combination of ketoprofen and a calcium channel antagonist
in a carrier, each of the ketoprofen and the calcium channel antagonist included in a
therapeutically effective amount such that the combination inhibits
pain/inflammation and spasm at a site of local delivery.
2. The composition of Claim 1, wherein the calcium channel antagonist
comprises an L-type calcium channel antagonist.
3. The method of Claim 2, wherein the L-type calcium channel
antagonist is selected from the group consisting of verapamil, diltiazem, bepridil,
mibefradil, nifedipine, nicardipine, isradipine, amlodipine, felodipine, nisoldipine
and nimodipine.
4. The method, of Claim 2, wherein the L-type calcium channel
antagonist comprises a dihydropyridine.
5. The composition of Claim 4, wherein the L-type calcium channel
antagonist is selected from the group consisting of nifedipine, nicardipine, isradipine,
amlodipine, felodipine, nisoldipine and nimodipine.
6. The method of Claim 5, wherein the L-type calcium channel
antagonist comprises nifedipine.
7. The composition of Claim 5, wherein the L-type calcium channel
antagonist has an onset of action of less than or equal to 10 minutes.
8. The composition of Claim 1, wherein the calcium channel antagonist
comprises nifedipine.
9. The composition of Claim 8, wherein ketoprofen and nifedipine are
included in the composition in a ketoprofen:nifedipine molar ratio of from 10:1 to
1:10.
-48-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

10. The composition of Claim 9, wherein ketoprofen and nifedipine are
included in the composition in a ketoprofen:nifedipine molar ratio of from 4:1 to 1:1.
11. The composition of Claim 10, wherein ketoprofen and nifedipine are
included in the composition in a ketoprofen:nifedipine molar ratio of 3:1.
12. The composition of Claim 8, wherein ketoprofen is included in the
compositon at a concentration of no more than 500,000 nanomolar and nifedipine is
included in the composition at a concentration of no more than 200,000 nanomolar.
13. The composition of Claim 12, wherein ketoprofen is included in the
compositon at a concentration of no more than 300,000 nanomolar and nifedipine is
included in the composition at a concentration of no more than 100,000 nanomolar.
14. The composition of Claim 13, wherein ketoprofen is included in the
compositon at a concentration of no more than 50,000 nanomolar and nifedipine is
included in the composition at a concentration of no more than 25,000 nanomolar.
15. The composition of Claim 8, wherein the carrier comprises an
aqueous carrier.
16. The composition of Claim 15, wherein the composition comprises at
least one stabilizing agent.
17. The composition of Claim 16, wherein the at least one stabilizing
agent comprises propyl gallate.
18. The composition of Claim 17, wherein the composition further
comprises sodium metabisulfite.
19. The composition of Claim 18, wherein the composition further
comprises polyethylene glycol .400 as a cosolvent.
20. The composition of Claim 19, wherein the composition further
comprises a citric acid buffer.
21. The composition of Claim 18, wherein the composition further
comprises a buffer.
-49-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

22. The composition of Claim 18, wherein the buffer comprises a citric
acid buffer.
23. The composition of Claim 15, wherein the composition further
comprises a cosolvent.
24. The composition of Claim 23, wherein the cosolvent comprises
polyethylene glycol.
25. The composition of Claim 24, wherein the cosolvent comprises
polyethylene glycol 400.
26. The composition of Claim 23, wherein the composition further
comprises propyl gallate.
27. The composition of Claim 8, wherein the composition further
comprises at least one stabilizing agent.
28. The composition of Claim 27, wherein the stabilizing agent comprises
propyl gallate.
29. The composition of Claim 28, wherein the composition further
comprises sodium metabilsulfite.
30. The composition of Claim 1, wherein ketoprofen is included in the
compositon at a concentration of no more than 500,000 nanomolar and the calcium
channel antagonist is included in the composition at a concentration of no more than
200,000 nanomolar.
31. The composition of Claim 30, wherein ketoprofen is included in the
compositon at a concentration of no more than 300,000 nanomolar and the calcium
channel antagonist is included in the composition at a concentration of no more than
100,000 nanomolar.
32. The composition of Claim 31, wherein ketoprofen is included in the
compositon at a concentration of no more than 50,000 nanomolar and the calcium
-50-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

channel antagonist is included in the composition at a concentration of no more than
25,000 nanomolar.
33. The composition of Claim 1, wherein ketoprofen comprises the S-(+)-
enantiomer, dexketoprofen.
34. The composition of Claim 1, wherein the carrier comprises an
aqueous carrier.
35. The composition of Claim 34, wherein the composition comprises at
least one stabilizing agent.
36. The composition of Claim 35, wherein the at least one stabilizing
agent comprises propyl gallate.
37. The composition of Claim 36, wherein the composition further
comprises sodium metabisulfite.
38. The composition of Claim 37, wherein the composition farther
comprises polyethylene glycol 400 as a co solvent.
39. The composition of Claim 38, wherein the composition further
comprises a citric acid buffer.
40. The composition of Claim 37, wherein the composition further
comprises a buffer.
41. The composition of Claim 40, wherein the buffer comprises a citric
acid buffer.
42. The composition of Claim 34, wherein the composition further
comprises a cosolvent.
43. The composition of Claim 42, wherein the cosolvent comprises
polyethylene glycol.
44. The composition of Claim 43, wherein the cosolvent comprises
polyethylene glycol 400.
-51-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

45. The composition of Claim 42, wherein the composition further
comprises propyl gallate.
46. The composition of Claim 1, wherein the carrier comprises a liquid
irrigation carrier.
47. The composition of Claim 1, wherein the composition is coated onto
an implantable device or medical instrument.
48. The composition of Claim 1, wherein the composition is impregnated
into an implantable device or medical instrument.
49. A locally deliverable composition for inhibiting pain/inflammation
and spasm, comprising a combination of a cyclooygenase inhibitor and a calcium
channel antagonist, each included in a therapeutically effective amount such that the
combination inhibits pain/inflammation and spasm at a site of local delivery, propyl
gallate as a stabilizing agent and a liquid carrier.
50. The composition of Claim 49, wherein the composition comprises
polyethylene glycol as a cosolvent.
51. The composition of Claim 50, wherein the composition comprises
polyethylene glycol 400.
52. The composition of Claim 49, wherein the composition comprises at
least a second stabilizing agent.
53. The composition of Claim 52, wherein the second stabilizing agent
comprises sodium metabisulfite.
54. The composition of Claim 53, wherein the composition further
comprises a citric acid buffer.
55. The composition of Claim 52, wherein the the composition further
comprises a citric acid buffer.
56. A.locally deliverable composition for inhibiting pain/inflammation
and spasm, comprising a combination of a cyclooxygenase inhibitor and a calcium
-52-
PH.1.0050.PCT.AP 03.17.06
3/17/2006 3:58:27 PM

-53-
channel antagonist, each included in a therapeutically effective amount such that the
combination inhibits pain/inflammation and spasm at a site of local delivery, an
aqueous liquid carrier, a cosolvent, at least one stabilizing agent and a buffer.


Compositions of a cyclooxygenase inhibitor and a calcium channel antagonist in a liquid carrier. The composition may be administered the the urinary tract during urological diagnostic, interventional, surgical and other medical procedures. One disclosed composition comprises ketoprofen and nifedipine in a liquid irrigation
carrier, and includes a solubilizing agent, stabilizing agents and a buffering agent.

Documents:

04766-kolnp-2007-abstract.pdf

04766-kolnp-2007-claims 1.0.pdf

04766-kolnp-2007-claims 1.1.pdf

04766-kolnp-2007-correspondence others.pdf

04766-kolnp-2007-description complete.pdf

04766-kolnp-2007-drawings.pdf

04766-kolnp-2007-form 1.pdf

04766-kolnp-2007-form 13.pdf

04766-kolnp-2007-form 3.pdf

04766-kolnp-2007-form 5.pdf

04766-kolnp-2007-gpa.pdf

04766-kolnp-2007-international search report.pdf

04766-kolnp-2007-pct request form.pdf

4766-KOLNP-2007-(14-11-2013)-CORRESPONDENCE.pdf

4766-KOLNP-2007-(16-01-2015)-CLAIMS.pdf

4766-KOLNP-2007-(16-01-2015)-CORRESPONDENCE.pdf

4766-KOLNP-2007-(16-01-2015)-FORM-13.pdf

4766-KOLNP-2007-(16-01-2015)-OTHERS.pdf

4766-KOLNP-2007-(17-02-2012)-CORRESPONDENCE.pdf

4766-KOLNP-2007-(20-01-2012)-ABSTRACT.pdf

4766-KOLNP-2007-(20-01-2012)-AMANDED CLAIMS.pdf

4766-KOLNP-2007-(20-01-2012)-CORRESPONDENCE.pdf

4766-KOLNP-2007-(20-01-2012)-DESCRIPTION (COMPLETE).pdf

4766-KOLNP-2007-(20-01-2012)-DRAWINGS.pdf

4766-KOLNP-2007-(20-01-2012)-FORM 1.pdf

4766-KOLNP-2007-(20-01-2012)-FORM 13.pdf

4766-KOLNP-2007-(20-01-2012)-FORM 2.pdf

4766-KOLNP-2007-(20-01-2012)-FORM 3.pdf

4766-KOLNP-2007-(20-01-2012)-FORM 5.pdf

4766-KOLNP-2007-(20-01-2012)-OTHERS.pdf

4766-KOLNP-2007-(20-01-2012)-PETITION UNDER RULE 137-1.1.pdf

4766-KOLNP-2007-(30-07-2012)-CORRESPONDENCE.pdf

4766-KOLNP-2007-(30-07-2012)-OTHERS.pdf

4766-kolnp-2007-ASSIGNMENT-1.1.pdf

4766-KOLNP-2007-ASSIGNMENT.pdf

4766-KOLNP-2007-CANCELLED PAGES.pdf

4766-KOLNP-2007-CORRESPONDENCE OTHERS 1.1.pdf

4766-KOLNP-2007-CORRESPONDENCE.pdf

4766-KOLNP-2007-DECISION.pdf

4766-kolnp-2007-drawings.pdf

4766-KOLNP-2007-EXAMINATION REPORT REPLY RECIEVED.pdf

4766-KOLNP-2007-EXAMINATION REPORT.pdf

4766-KOLNP-2007-FORM 13.pdf

4766-KOLNP-2007-FORM 18.pdf

4766-KOLNP-2007-FORM 3-1.1.pdf

4766-KOLNP-2007-GPA.pdf

4766-KOLNP-2007-GRANTED-ABSTRACT.pdf

4766-KOLNP-2007-GRANTED-CLAIMS.pdf

4766-KOLNP-2007-GRANTED-DESCRIPTION (COMPLETE).pdf

4766-KOLNP-2007-GRANTED-DRAWINGS.pdf

4766-KOLNP-2007-GRANTED-FORM 1.pdf

4766-KOLNP-2007-GRANTED-FORM 2.pdf

4766-KOLNP-2007-GRANTED-FORM 3.pdf

4766-KOLNP-2007-GRANTED-FORM 5.pdf

4766-KOLNP-2007-GRANTED-LETTER PATENT.pdf

4766-KOLNP-2007-GRANTED-SPECIFICATION-COMPLETE.pdf

4766-KOLNP-2007-INTERNATIONAL SEARCH REPORT & OTHERS.pdf

4766-KOLNP-2007-OTHERS 1.1.pdf

4766-KOLNP-2007-PETITION UNDER RULE 137.pdf

4766-KOLNP-2007-REPLY TO EXAMINATION REPORT.pdf


Patent Number 265090
Indian Patent Application Number 4766/KOLNP/2007
PG Journal Number 07/2015
Publication Date 13-Feb-2015
Grant Date 06-Feb-2015
Date of Filing 07-Dec-2007
Name of Patentee OMEROS CORPORATION
Applicant Address 1420 FIFTH AVENUE, SUITE 2600 SEATTLE, WASHINGTON
Inventors:
# Inventor's Name Inventor's Address
1 HERZ JEFFREY M 14427 12TH DRIVE SE, MILL CREEK, WASHINGTON 98012
2 DEMOPULOS GREGORY A 4845 FOREST AVENUE SW, MERCER ISLAND, WASHINGTON 98040
3 GOMBOTZ WAYNE R 15545 61ST AVENUE NE, KENMORE, WASHINGTON 98028
4 SHEN HUI-RONG 10376 NE 12TH STREET, SUITE J204, BELLEVUE, WASHINGTON 98004
PCT International Classification Number A61K 9/22
PCT International Application Number PCT/US2006/009771
PCT International Filing date 2006-03-17
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/683488 2005-05-20 U.S.A.