Formulations of low dose diclofenac and beta-cyclodextrin

Information

  • Patent Grant
  • 8946292
  • Patent Number
    8,946,292
  • Date Filed
    Thursday, May 12, 2011
    13 years ago
  • Date Issued
    Tuesday, February 3, 2015
    9 years ago
Abstract
The present invention is directed to a pharmaceutical composition containing a unit dose of a diclofenac compound effective to induce analgesia; and a beta-cyclodextrin compound; wherein the dose of the diclofenac compound is less than 75 mg, e.g., from about 18.75 mg to about 37.5 mg. The present invention is also directed to methods of treating a subject in need of analgesia with the pharmaceutical compositions of the invention.
Description
FIELD OF THE INVENTION

The present invention is directed to pharmaceutical compositions and methods of treating a subject in need of analgesia with pharmaceutical compositions which contain diclofenac and beta-cyclodextrin. Specifically, the compositions contain low doses of diclofenac, namely less than 10 mg.


BACKGROUND OF THE INVENTION

Diclofenac is a well-known non-steroidal anti-inflammatory drug (“NSAID”) used in acute and chronic pain in both parenteral and oral dosage forms. Oral dosages range from 100-200 mg/day, while parenteral dosages range from 75-150 mg/day (1-2 mg/kg/day) by either infusion or intermittent (divided) doses. Toxicity of oral and parenteral forms are well known, with gastro-intestinal, hemorrhagic, renal, hepatic, cardiovascular and allergic (anaphylactic and severe dermal allergy) adverse events being most significant.


Parenteral use of diclofenac has been limited due to limited solubility, such that parenteral preparations have had to include non-polar solvents in order to achieve concentrations (75 mg/3 ml) which would allow intra-muscular (IM) administration of the desired dose. This solubility has limited the parenteral use to IM use and/or slow intravenous (IV) administration of diluted (100-500 ml diluent) product.


U.S. Pat. No. 5,679,660 and co-pending application Ser. No. 10/999,155, filed Nov. 30, 2004,published as US 2005/0238674 A1 on Oct. 27, 2005,both of which are incorporated by reference, disclose novel formulations of diclofenac with hydroxypropyl-beta-cyclodextrin, which allows a more concentrated preparation and thus rapid intravenous administration. The data show that the more concentrated the diclofenac/beta-cyclodextrin formulation, the faster onset of action over current products.


Other than ease of administration and more rapid onset of action, consequent on the improvements in the pharmaceutical formulation, no other advantages were observed. The present invention arises, in part, from the surprising discovery that formulating a non-steroidal anti-inflammatory drug with beta-cyclodextrin not only improves solubility and stability of the drug, it also increases efficacy.


SUMMARY OF THE INVENTION

The present invention relates to a pharmaceutical composition comprising a unit dose of a diclofenac compound effective to induce analgesia; and a beta-cyclodextrin compound, in which the dose of the diclofenac compound is less than 10 mg. This dose is less than any effective dose previously reported or even suggested for a formulation of a diclofenac and a beta-cyclodextrin compound. The diclofenac compound can be a diclofenac salt, e.g., diclofenac sodium, as exemplified infra. The beta-cyclodextrin compound can be 2-hydroxypropyl-beta-cyclodextrin, as exemplified infra.


The pharmaceutical composition may further comprise a stabilizer, such as monothioglycerol.


In specific embodiments, the pharmaceutical composition provides a dose of diclofenac of about 9.4 mg, less than about 5 mg, and even about 3.75 mg.


The invention further provides a method for treating a mammal in need of analgesia by administering a pharmaceutical composition of the invention, as set forth above. In a specific embodiment, the mammal is a human. In particular, the pharmaceutical composition can be administered intravenously.


The advantageous methods of the invention pertain to other formulations as well. Thus, the invention provides a method for treating a mammal in need of analgesia by administering a pharmaceutical composition comprising a dosage of a diclofenac compound effective to induce analgesia; and a beta-cyclodextrin compound, in which the dosage of the diclofenac compound is less than about 1.3 mg/kg per day. In particular embodiments, the dosage of diclofenac is less than 0.65 mg/kg per day, less than 0.33 mg/kg per day, or less than 0.165 mg/kg per day.


In another embodiment of the methods of treatment, the invention provides a method for treating a mammal in need of analgesia by administering a pharmaceutical composition comprising a dosage of a diclofenac compound effective to induce analgesia; and a beta-cyclodextrin compound, wherein the dosage of the diclofenac compound is less than a minimum approved dose for a particular route of administration. The dose of the diclofenac compound can have the same efficacy of pain relief as the minimum approved dose, or it can have from about 70% to about 100% or from about 40% to about 70% of the efficacy of pain relief as the minimum approved dose. In addition, the dose of the diclofenac compound can have the same duration of pain relief as the minimum approved dose, or it can have from about two-thirds to the same duration of pain relief, or from about one-third to about two-thirds of the duration of pain relief, as the minimum approved dose.





DESCRIPTION OF THE FIGURES


FIG. 1 contains a graphical representation of the 100 mm visual analog pain relief (mm) afforded to patients over time (hours) based on the formulation strengths administered. The tested formulations include placebo, 3.75 mg Dyloject, 9.4 mg Dyloject, 18.75 mg Dyloject; 37.5 mg Dyloject, 75 mg Dyloject, and 30 mg Ketorolac.



FIG. 2 illustrates the dose-response curve for peak analgesia observed (mm VAS) over mg of formulation. Both diclofenac and ketorolac formulations were tested.



FIG. 3 illustrates the dose-duration relationship examined using the median time to re-medication (hours) in the single dose phase. Two formulations of diclofenac were studied.



FIG. 4 illustrates the percentage of patients with NSAID Adverse Events by dose of diclofenac (mg).





DETAILED DESCRIPTION OF THE INVENTION

The present invention provides formulations of a diclofenac compound with a beta-cyclodextrin compound. These formulations unexpectedly provide for significant efficacy and duration of pain relief at a lower dose than the current recommended doses of the diclofenac. More particularly, at a reduced dose and dosage, the formulation provides the same level of efficacy and the same duration of analgesia as at the minimum approved dose and dosage.


The invention is based, in part, on the results of a comparison of the efficacy of diclofenac solubilized with hydroxypropyl-beta-cyclodextrin to ketorolac and placebo for the treatment of moderate-to-severe post-surgical pain. The efficacy of diclofenac solubilized with hydroxypropyl-beta-cyclodextrin at several dose levels suggests a faster onset of action. Most notably, diclofenac formulated with hydroxypropyl-beta-cyclodextrin provides single-dose efficacy at about 50%, about 25%, about 12.5% and about 5% of the current recommended doses of diclofenac. This, in combination with the known human pharmacokinetic results for the formulation, supports reduced total daily doses of this NSAID with anticipated lower risk of toxicity by reducing the extent and duration of drug exposure. This is a novel finding and of clinical importance.


The minimum effective dose of diclofenac solubilized with hydroxypropyl-beta-cyclodextrin tested was 3.75 mg, demonstrating that diclofenac, if solubilized with hydroxypropyl-beta-cyclodextrin, may be administered at doses lower than those previously considered necessary for postoperative analgesia.


The term “diclofenac compound” refers to diclofenac or a pharmaceutically acceptable diclofenac salt. A pharmaceutically acceptable salt of diclofenac, can be an alkali metal salt, for example the sodium or the potassium salt, or the salt formed with an amine, e.g., a mono-, di- or tri-C1-C4 alkylamine, for example diethyl- or triethyl-amine, hydroxy-C2-C4 alkylamine, for example ethanolamine, or hydroxy-C2-C4 alkyl-C1-C4 alkylamine, for example dimethylethanolamine, or a quaternary ammonium salt, for example the tetramethylammonium salt or the choline salt of diclofenac (see, e.g., U.S. Pat. No. 5,389,681). Preferably the diclofenac salt is diclofenac sodium.


Suitable formulations of the present invention for parenteral administration include cyclodextrin inclusion complexes. One or more modified or unmodified cyclodextrins can be employed to stabilize and increase the water solubility and efficacy of compounds of the present invention. Useful cyclodextrins for this purpose include beta-cyclodextrins.


The term “beta-cyclodextrin” as used herein refers to cyclic alpha-1,4-linked oligosaccharides of a D-glucopyranose containing a relatively hydrophobic central cavity and hydrophilic outer surface. Particular efficacy has been observed in the present invention utilizing hydroxypropyl-beta-cyclodextrin, however, other substituted and unsubstituted beta-cyclodextrins can also be used in the practice of the invention. Additional examples of cyclodextrins that may be utilized are disclosed in U.S. Pat. Nos. 4,727,064, 4,764,604, 5,024,998, 6,407,079, 6,828,299, 6,869,939 and Jambhekar et al., 2004 Int. J Pharm. 2004, 270(1-2) 149-66.The formulations may be prepared as described in U.S. Pat. Nos. 5,679,660 and 5,674,854.


The “pharmaceutical compositions” for use in accordance with the present invention can be formulated in any conventional manner using one or more pharmaceutically acceptable carriers or excipients. A “pharmaceutically acceptable” carrier or excipient, as used herein, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in mammals, and more particularly in humans.


Pharmaceutical compositions include solid dosage forms, e.g., for perioral, transnasal (powder), or rectal (suppository) administration; and liquid dosage forms, e.g., for parenteral administration (injection), transnasal (spray), or perioral administration. In a specific embodiment, the pharmaceutical compositions of the present invention are liquid compositions formulated for intravenous or intramuscular administration, and particularly intravenous administration.


As used herein, the term “stabilizer” refers to a compound optionally used in the pharmaceutical compositions of the present invention in order to avoid the need for sulphite salts and increase storage life. Optimal stabilizers include antioxidants, specifically monothioglycerol and those described in U.S. Patent Publication 2005/0238674.


The term “dosage” is intended to encompass a formulation expressed in terms of mg/kg/day. The dosage is the amount of an ingredient administered in accordance with a particular dosage regimen. A “dose” is an amount of an agent administered to a subject in a unit volume or mass, e.g., an absolute unit dose expressed in mg of the agent. The dose depends on the concentration of the agent in the formulation, e.g., in moles per liter (M), mass per volume (m/v), or mass per mass (m/m). The two terms are closely related, as a particular dosage results from the regimen of administration of a dose or doses of the formulation. The particular meaning in any case will be apparent from context.


The term “mammal” is intended to include, any warm-blooded vertebrate having the skin more or less covered with hair. Most preferably, the mammal is a human subject, but the mammal can also be a non-human animal. Thus, the invention is useful in veterinary medicine as well, e.g., for treating pain in a domestic pet, such as a canine or feline, a farm animal, a work animal, or an animal in a circus or zoological garden. The invention has particular value in treating pain in a horse, particularly in sport, such as thoroughbred and other race horses, rodeo horses, circus horses, and dressage horses. A particular advantage of the invention is that, by increasing the efficacy of a dosage of diclofenac, it is possible to administer a therapeutic dosage that is below a maximum allowed dose permitted by the particular regulatory authorities of the sport.


The term “minimum approved dose” refers to the minimum dosage that has received full regulatory approval by the appropriate United States or foreign regulatory authority as safe and effective for human or veterinary use.


The term “therapeutically effective” as applied to dose or amount refers to that quantity of a compound or pharmaceutical composition that is sufficient to result in a desired activity upon administration to a mammal in need thereof. As used herein with respect to the pharmaceutical compositions comprising an antifungal, the term “therapeutically effective amount/dose” refers to the amount/dose of a compound or pharmaceutical composition that is sufficient to produce an effective response upon administration to a mammal.


The term “amount” as used herein refers to quantity or to concentration as appropriate to the context. In the present invention, the effective amount of a compound refers to an amount sufficient to treat a patient/subject in need of analgesia. The effective amount of a drug that constitutes a therapeutically effective amount varies according to factors such as the potency of the particular drug, the route of administration of the formulation, and the mechanical system used to administer the formulation. A therapeutically effective amount of a particular drug can be selected by those of ordinary skill in the art with due consideration of such factors.


The term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 3 or more than 3 standard deviations, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value.


As used herein, the term “treat” is used herein to mean to relieve or alleviate at least one symptom of a disease in a subject. Within the meaning of the present invention, the term “treat” also denotes to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disease) and/or reduce the risk of developing or worsening a disease.


Methods of Treatment

As noted above, the novel dosage formulations of the invention are suitable for administering diclofenac to treat pain, i.e., for analgesia. Various embodiments of the invention provide for administration of unit doses to achieve a total dosage for the desired effect. The examples demonstrate efficacy of a 3.75 mg dose of diclofenac, which is about 5% of the minimum approved dose (and about 5% or about 2.5% of the approved daily dosage). However, this dose provides about 40% of the pain relief and one-third of the duration as the minimum approved dose. Better results can be achieved by selecting a dosage regimen with this dose of diclofenac, e.g., increasing the frequency of administration, to achieve a level and duration of pain relief acceptable for the patient. Higher dose formulations likewise could provide such relief. Such higher dose formulations are nevertheless lower than any approved formulation, and the dosage regimen results in administration of less diclofenac than the current approved minimum dosage regimen.


A significant advantage of the invention results from the ability to achieve efficacy with lower doses and overall daily dosing of diclofenac. Consequently, it is possible to reduce the dosage, and thus reduce toxicity.


In specific embodiments, the unit dose (i.e., the amount of active drug administered at one time to a patient) is no more than about 75%, no more than about 50%, no more than about 25%, no more than about 12.5%, and no more than about 5%, of the approved minimum dose. Doses that are about or greater than about 50% of the approved minimum dose can show the same level and duration of pain relieve as the minimum effective dose. Furthermore, by increasing the frequency of administration of a lower dose formulation, the patient can achieve the same levels of efficacy and duration of pain relief as with the approved doses, with decreased toxicity.


In another embodiment, then, the invention provides for titrating the dose reduction of diclofenac and beta-cyclodextrin by decreasing the unit dose to achieve an analgesic effect that is sufficient, even at a reduced level, for the patient's needs, which can be met by increasing the dosing frequency to achieve an effective daily dosage that is still lower than the minimum approved dose. The term “effect” means that there is a statistically significant difference in a response in patients taking the formulation containing the diclofenac relative to patients taking a placebo.


The formulations of the invention can be administered via any route, including parenteral, perioral, transnasal, and rectal. Particular parenteral routes of administration include intravenous and intramuscular injection.


The formulations of the invention are suitable for treating pain by administration either or both prophylactically and after onset of the pain. Thus, as used herein, the term “treat” (in any of its grammatical forms) means to reduce pain through administration of a formulation of the invention prior to or after the onset of pain, or both. In particular, the formulations are suitable in the treatment of acute painful conditions in humans and animals such as headache, including migraine, trauma, dysmenorrhoea, renal or biliary colic, post-operative pain, gout, arthritis, cancer related pain, musculoskeletal pain, lower back pain, fibromyalgia, and pain of infectious origin. Indeed, the low dosage of diclofenac will have little or no anti-inflammatory activity, so in the treatment of pain of infectious origin it will have little effect on any immune response to the infectious organism while achieving analgesia. In a specific embodiment, exemplified below, the formulation is effective to treat post-surgical dental pain resulting from surgical extraction of one or more third molars. In addition, although not intending to be bound by any particular mechanism of action, the formulation of the invention may be used prophylactically to prevent the formation of prostaglandins during and after surgery, with subsequent reduction in immediate post-operative pain. Further, the formulation of the invention may be used to modulate nuclear transcription factors, such as NF-κB, or to modulate ion channel activity, for example as described in Ocana, Maria et al., Potassium Channels and Pain: Present Realities and Future Opportunities, 500 Eur. J. Pharm. 203 (2004).


EXAMPLES

The present invention will be better understood by reference to the following Examples, which are provided as exemplary of the invention, and not by way of limitation.


Example 1
Analysis of Pain Relief Afforded to Patients Based on Administered Dose

A 336-patient, seven treatment arm, randomized, double-blind, single-dose, and placebo- and comparator-controlled, parallel-group study was conducted. Patients were randomly assigned to receive a single dose of either diclofenac sodium solubilized with hydroxypropyl-beta-cyclodextrin (hereinafter “DIC”), ketorolac tromethamine, or placebo.


Bolus IV injectable 2 ml solutions were prepared by solubilizing diclofenac sodium with hydroxypropyl-beta-cyclodextrin. The formulation strengths were as follows:

    • Formulation: Diclofenac sodium solubilized with hydroxypropyl-β-cyclodextrin
    • Strengths: 75 mg, 37.5 mg, 18.75 mg, 9.4 mg and 3.75 mg
    • Dosage: Bolus IV injection (no less than 15 sec)
    • Batch Number: 063004 (PPS04010)
    • Manufacturer: Manufactured for Javelin by Precision Pharma
    • Storage Conditions: Room temperature
    • Active control/comparator:
    • Formulation: Ketorolac Tromethamine
    • Strength: 30 mg
    • Dosage: Bolus IV injection (no less than 15 sec)
    • Batch Number: 21-430-DK
    • Manufacturer: Abbott Labs
    • Storage Conditions: Room temperature


Pain was assessed by each patient at Baseline (0 hour: Visual Analog Scale (VAS) and categorical pain intensity only), at 5, 15, 30 and 45 minutes, and at 1, 1.5, 2, 3, 4, 5, 6, 7, 8, 10, 12 and 24 hours after administration of study medication and immediately prior to the first dose of rescue medication. At each post-dose time period, levels of pain intensity (categorical and VAS) and pain relief (categorical and VAS) was assessed by each patient. Patients were also provided with 2 stopwatches to measure perceptible and meaningful pain relief.


The presence of a dose-response was tested with a step-down testing procedure. Total pain relief (TOTPAR), peak pain relief, pain intensity difference (SPID), summed peak reduction in pain intensity difference (SPRID), and patient global evaluation was analyzed with analysis of variance (ANOVA) with treatment, center, and baseline categorical pain intensity as factors. The possibility of interactions was investigated. Comparisons of the DIC groups with the placebo and Ketorolac groups were performed with Dunnett's test. The presence of a linear dose response for the ordered DIC dose levels was tested with orthogonal contrasts for TOTPAR, SPID and SPRID. Tests of individual DIC dose levels versus placebo for TOTPAR, SPID, and SPRID were conducted with the Tukey, Ciminera, and Heyse step-down testing procedure. The mean, standard deviation, and sample size were presented for each treatment group. Significant p-values (those less than or equal to 0.05) were presented for each step of the procedure. Non-significant p-values were represented with three dashes. Time to onset of perceptible relief and time to onset of meaningful relief was analyzed with survival analysis techniques. These variables were summarized with number of observations, mean, standard deviation, median, and range. Log-rank tests were used to compare treatments with respect to survival distributions. The median time to event for each treatment group was estimated with the Kaplan-Meier product limit estimator. A 95% confidence interval for each estimated median time to event was calculated.


The results of the study were strongly positive, novel and could not have been anticipated from prior experience with diclofenac. The doses had been chosen based on the currently recommended minimally effective doses of 1 mg/kg (50 mg immediate-release or 100 mg sustained-release orally or 75 mg intramuscularly or intravenously). Based on these doses the test conditions were a full dose (75 mg), half dose (37.5) mg, a possibly effective dose (18.75 mg) and two placebo doses (9.75 & 3.4 mg). The findings were as follows:









TABLE 1







TOTPAR (Sum of Pain Relief VAS 0-100 mm ratings 0-6 hours)












Treatment Group

Result
% Maximum Effect
















Placebo
62.8
(SEM 9)
17%



DIC 3.75 mg
134.1
(SEM 8)
38%



DIC 9.4 mg
237.6
(SEM 15)
68%



DIC 18.75 mg
284.4
(SEM 21)
82%



DIC 37.5 mg
348.2
(SEM 30)
100% 



DIC 75 mg
346.3
(SEM 27)
100% 



ketorolac
400.3
(SEM 36)
100% 










See FIG. 1 for the corresponding graphical representation of the pain relief afforded to patients based on the formulation strengths administered.


Example 2
Analysis of Efficacy & Duration of Pain Relief at Lower Doses of Diclofenac

To explore this further, the dose-duration relationship in the same study was examined using the median time to remedication in the single-dose phase. Utilizing the results of study in Example 1,the efficacy and duration of pain relief were thoroughly analyzed. The lowest IV dose of DIC (3.75 mg) had 38% of the effect of the maximal dose, and the next lowest dose (9.4 mg) had 68% of the maximal possible effect, despite being 5% and 12% respectively of the current recommended minimally effective dose (1 mg/kg). FIG. 2 contains a graphical illustration of the dose-response for peak analgesia observed in the study.



FIG. 3 depicts the dose-duration relationship examined using the median time to remedication in the single dose phase. The peak analgesic response was about 80% pain relief, with a 50% response at a dose of 4-8 milligrams of Diclofenac in relation to dental pain. Similar peak analgesic response was seen for 30 milligrams of ketorolac. Given the shape of the dose response curve, it is clear that lower doses of the DIC formulation achieved the same results as the current established dose of diclofenac of 75 milligrams.


The findings show a 6 hour duration of effect for all doses above about 20 milligrams (18 milligrams).


For most drugs the findings of significant activity at doses far below the recommended doses would be of little significance due to large therapeutic indices (wide ranges between the effective and toxic doses). The opposite is true for parenteral NSAIDs; it has been well established in the prior art that increasing the dose of these drugs rapidly diminishes their utility due to increasing risk of toxicity.


Thus the finding that with the new formulation of diclofenac that 5%-12% of the usual dose can provide 38-68% of the desired therapeutic effect is remarkable and unanticipated. This leads to the possibility that the high early blood levels possible with the new formulation allow lower total daily doses resulting in less risk of toxicity.


This finding demonstrates efficacy with a lower daily dose (25-75 mg/day) than current dosing of diclofenac (75-200 mg/day), and anticipates better dosing paradigms (less than Q 12 hours) offering the expectation of lesser toxicity. Proof of lesser toxicity from available data from this study is suggestive, based on the known relationship of dose and toxicity.


The novel diclofenac formulation allowed by hydroxypropyl-β-cyclodextrin has been shown to provide proof of single-dose efficacy at 50%, 25%, 12.5% and 5% of the current recommended doses of diclofenac. This in combination with the known human pharmacokinetic results for the formulation supports reduced total daily doses of this NSAID with anticipated lower risk of toxicity by reducing the extent and duration of drug exposure.


The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims.


Patents, patent applications publications product descriptions, and protocols are cited throughout this application the disclosures of which are incorporated herein by reference in their entireties for all purposes.

Claims
  • 1. A method of providing analgesia to a subject in need thereof, which method comprises parenterally administering to the subject a pharmaceutical composition, wherein the composition comprises: (a) from about 18.75 mg to about 37.5 mg of a diclofenac compound; and(b) a beta-cyclodextrin compound;wherein the dosage of the diclofenac compound is less than about 1.3 mg/kg per day; andwherein the method achieves at least about 82% of maximum observable total pain relief assessed by a Visual Analog Scale.
  • 2. The method of claim 1, wherein the diclofenac compound is diclofenac sodium.
  • 3. The method of claim 1, wherein the pharmaceutical composition is administered intravenously.
  • 4. The method of claim 1, wherein the dose of the diclofenac compound is 18.75 mg (+/−20%).
  • 5. The method of claim 1, wherein the dose diclofenac compound is 37.5 mg (+/−20%).
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of application Ser. No. 11/689,931, filed Mar. 22, 2007 now U.S. Pat. No. 8,580,954, which claims priority under 35 U.S.C. §119, based on U.S. Provisional Application 60/786,486, filed Mar. 28, 2006. The disclosures of these applications are incorporated herein by reference in their entireties.

US Referenced Citations (76)
Number Name Date Kind
4711906 von Stetten et al. Dec 1987 A
4727064 Pitha Feb 1988 A
4764604 Muller Aug 1988 A
4983586 Bodor Jan 1991 A
5017566 Bodor May 1991 A
5024998 Bodor Jun 1991 A
5354560 Lovrecich Oct 1994 A
5389681 Galli Feb 1995 A
5449521 Lovrecich Sep 1995 A
5464633 Conte et al. Nov 1995 A
5674854 Bodley et al. Oct 1997 A
5679660 Bodley et al. Oct 1997 A
5681583 Conte et al. Oct 1997 A
5690954 Illum Nov 1997 A
5702724 Stahl et al. Dec 1997 A
5738874 Conte et al. Apr 1998 A
5747058 Tipton et al. May 1998 A
5747061 Amselem et al. May 1998 A
5785989 Stanley et al. Jul 1998 A
5811547 Nakamichi et al. Sep 1998 A
5821237 Bissett et al. Oct 1998 A
5854226 Penkler et al. Dec 1998 A
5866162 Grattan Feb 1999 A
5891913 Sallmann et al. Apr 1999 A
5929115 Takeuchi et al. Jul 1999 A
6028222 Dietlin et al. Feb 2000 A
6071964 Fischer et al. Jun 2000 A
6107343 Sallmann et al. Aug 2000 A
6294192 Patel et al. Sep 2001 B1
6365180 Meyer et al. Apr 2002 B1
6407079 Müller et al. Jun 2002 B1
6828299 Yang et al. Dec 2004 B2
6869939 Mosher et al. Mar 2005 B2
6923988 Patel et al. Aug 2005 B2
7115586 Loftsson Oct 2006 B2
7186260 Hyson Mar 2007 B2
7423028 Zoppetti et al. Sep 2008 B2
7655231 Shelton et al. Feb 2010 B2
20020012680 Patel et al. Jan 2002 A1
20020034539 Esposito et al. Mar 2002 A1
20020035264 Kararli et al. Mar 2002 A1
20020107265 Chen et al. Aug 2002 A1
20030082234 Seo et al. May 2003 A1
20030113367 Penkler Jun 2003 A1
20030139698 Hyson Jul 2003 A1
20030232089 Singh et al. Dec 2003 A1
20040024044 Di Salle et al. Feb 2004 A1
20040072798 Naggi et al. Apr 2004 A1
20040137062 Chopra Jul 2004 A1
20040151774 Pauletti et al. Aug 2004 A1
20040157796 Gokarn Aug 2004 A1
20050043481 Gref et al. Feb 2005 A1
20050085446 Babu et al. Apr 2005 A1
20050095205 Krishnamoorthy May 2005 A1
20050118272 Besse et al. Jun 2005 A1
20050197303 Krishnamoorthy Sep 2005 A1
20050203115 Sancilio et al. Sep 2005 A1
20050238674 Penkler et al. Oct 2005 A1
20050276841 Davis et al. Dec 2005 A1
20060024238 Barth et al. Feb 2006 A1
20060121085 Warren et al. Jun 2006 A1
20060134095 Ito et al. Jun 2006 A1
20060188530 Yoo Aug 2006 A1
20060210604 Dadey et al. Sep 2006 A1
20060211599 Suzuki Sep 2006 A1
20070049552 Babu et al. Mar 2007 A1
20070116730 Simmons May 2007 A1
20070207222 Yu Sep 2007 A1
20070219170 Samson Sep 2007 A1
20070232566 Wright et al. Oct 2007 A1
20070232567 Wright et al. Oct 2007 A1
20080171014 Wu et al. Jul 2008 A1
20080200533 Krishnan Aug 2008 A1
20080220441 Birnbaum et al. Sep 2008 A1
20090292022 Kowalski et al. Nov 2009 A1
20120142779 Penkler et al. Jun 2012 A1
Foreign Referenced Citations (117)
Number Date Country
4207922 Sep 1993 DE
69820973 Dec 2004 DE
69434356 May 2006 DE
0268215 May 1988 EP
0335545 Oct 1989 EP
0371431 Jun 1990 EP
446753 Sep 1991 EP
0519428 Dec 1992 EP
598337 May 1994 EP
0647451 Apr 1995 EP
0658347 Jun 1995 EP
807434 Nov 1997 EP
868915 Oct 1998 EP
1004318 May 2000 EP
862414 Dec 2001 EP
833618 Apr 2002 EP
730443 May 2002 EP
1219304 Jul 2002 EP
1219306 Jul 2002 EP
729748 Feb 2003 EP
0658347 Aug 2003 EP
0701449 Aug 2003 EP
917457 Jan 2004 EP
792147 Mar 2004 EP
1574221 Sep 2005 EP
1595936 Nov 2005 EP
1609481 Dec 2005 EP
1681065 Jul 2006 EP
1767219 Mar 2007 EP
1524968 Dec 2007 EP
1515729 Feb 2008 EP
1677761 Apr 2008 EP
1967186 Sep 2008 EP
1974751 Oct 2008 EP
2 230 440 Oct 1990 GB
2417900 Mar 2006 GB
59-084821 May 1984 JP
62138437 Jun 1987 JP
1-287094 Nov 1989 JP
06-16547 Jan 1994 JP
7196484 Aug 1995 JP
7252144 Oct 1995 JP
9-48737 Feb 1997 JP
2006-502185 Jan 2006 JP
WO9002141 Mar 1990 WO
WO9200725 Jan 1992 WO
WO9203122 Mar 1992 WO
WO9406416 Mar 1994 WO
WO9428936 Dec 1994 WO
WO9501781 Jan 1995 WO
WO9504528 Feb 1995 WO
WO9507104 Mar 1995 WO
WO9511669 May 1995 WO
WO9532737 Dec 1995 WO
WO9611003 Apr 1996 WO
WO9614839 May 1996 WO
WO9622088 Jul 1996 WO
WO 9641646 Dec 1996 WO
WO9641617 Dec 1996 WO
WO9710805 Mar 1997 WO
WO9735568 Oct 1997 WO
WO9851280 Nov 1998 WO
WO9936060 Jul 1999 WO
WO0050007 Aug 2000 WO
WO0059475 Oct 2000 WO
WO 0066099 Nov 2000 WO
WO0071098 Nov 2000 WO
WO0113897 Mar 2001 WO
WO0128555 Apr 2001 WO
WO 0141760 Jun 2001 WO
WO0145742 Jun 2001 WO
WO0180797 Nov 2001 WO
WO0205815 Jan 2002 WO
WO0220020 Mar 2002 WO
WO02053188 Jul 2002 WO
WO02072106 Sep 2002 WO
WO 03018009 Mar 2003 WO
WO03033025 Apr 2003 WO
WO 03063824 Aug 2003 WO
WO 03095498 Nov 2003 WO
WO03097011 Nov 2003 WO
WO03105867 Dec 2003 WO
WO2004041118 May 2004 WO
WO2004081196 Sep 2004 WO
WO2004089418 Oct 2004 WO
WO2005044231 May 2005 WO
WO2005074887 Aug 2005 WO
WO2005077336 Aug 2005 WO
WO2005086763 Sep 2005 WO
WO2005092387 Oct 2005 WO
WO2005120578 Dec 2005 WO
WO2005123193 Dec 2005 WO
WO2006007753 Jan 2006 WO
WO2006041942 Apr 2006 WO
WO2006082588 Aug 2006 WO
WO2007004236 Jan 2007 WO
WO2007005608 Jan 2007 WO
WO2007044062 Apr 2007 WO
WO2007052289 May 2007 WO
WO2007059591 May 2007 WO
WO2007061828 May 2007 WO
WO2007103435 Sep 2007 WO
WO2007103687 Sep 2007 WO
WO2007106381 Sep 2007 WO
WO2007112272 Oct 2007 WO
WO2007112274 Oct 2007 WO
WO2008006216 Jan 2008 WO
WO2008040534 Apr 2008 WO
WO2008071851 Jun 2008 WO
WO2008074087 Jun 2008 WO
WO2008076703 Jun 2008 WO
WO2008115572 Sep 2008 WO
WO2008127271 Oct 2008 WO
WO2008127291 Oct 2008 WO
WO2008133982 Nov 2008 WO
WO2008134600 Nov 2008 WO
WO2008134601 Nov 2008 WO
Non-Patent Literature Citations (149)
Entry
Nagarsenker et al., “Solid dispersion of hydroxyproply beta-cyclodextrin and ketrolac: enhancement of in-vitro dissolution rates, improvement in anti-inflammatory activity and reduction in ulcerogenicity in rats”, Journal of Pharmacy and Pharmacology, 52(8):949-956, Aug. 2000.
U.S. Appl. No. 11/689,931, filed Mar. 22, 2007.
U.S. Appl. No. 11/689,939, filed Mar. 22, 2007.
U.S. Appl. No. 11/689,931, Apr. 6, 2011 Non-Final Office Action.
U.S. Appl. No. 11/689,931, Feb. 10, 2011 Respone to Non-Final Office Action.
U.S. Appl. No. 11/689,931, Nov. 10, 2011 Non-Final Office Action.
U.S. Appl. No. 11/689,931, Sep. 9, 2010 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,931, Mar. 10, 2010 Non-Final Office Action.
U.S. Appl. No. 11/689,931, Oct. 16, 2009 Amendment and Request for Continued Examination (RCE).
U.S. Appl. No. 11/689,931, Apr. 17, 2009 Final Office Action.
U.S. Appl. No. 11/689,931, Jan. 7, 2009 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,931, Sep. 25, 2008 Non-Final Office Action.
U.S. Appl. No. 11/689,939, Apr. 1, 2011 Final Office Action.
U.S. Appl. No. 11/689,939, Feb. 10, 2011 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,939, Sep. 10, 2011 Non-Final Office Action.
U.S. Appl. No. 11/689,939, Jul. 1, 2010 Examiner's Interview Summary.
U.S. Appl. No. 11/689,939, Jun. 23, 2010 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,939, Dec. 31, 2009 Non-Final Office Action.
U.S. Appl. No. 11/689,939, Oct. 28, 2009 Amendment and Request for Examination (RCE).
U.S. Appl. No. 11/689,939, Apr. 29, 2009 Final Office Action.
U.S. Appl. No. 11/689,939, Jan. 7, 2009 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,939, Sep. 19, 2008 Non-Final Office Action.
Adis Drugs Monograph on Diclofenac (2008).
Anderson et al., “Diclofenac in Combination With Opiate Infusion After Joint Replacement Surgery”, Anaesth. Intens. Care, (1991), 19:535-538.
Backensfeld et al., “Interaction of NSA with Cyclodextrins and Hydroxypropyl Cyclodextrin Derivatives”, Int. J. Pharm., (1991), 74:85-93.
Backensfeld et al., “Solubilization and Stabilization of Non-Steroidal Antirheumatics with Cyclodextrins and Cyclodextrin Ethers”, Arch. Pharm., (1990), 323:690.
Barbato F. et al. “Diclofenac beta-Cyclodextrin Binary Systems: A Study in Solution and in the Solid State,” J. Inclusion Phenomena and Macrocyclic Chem. vol. 46, Aug. 2003, pp. 179-185.
Blaicher et al., “Effect of non-selective, non-steroidal anti-inflammatory drugs and cyclo-oxygenase-2 selective inhibitors on the PFA-100 closure time”, Anaesthesia, (2004), 59:1100-1103.
Bricker et al., “Peri-operative blood loss and non-steriodal anti-inflammatory drugs: an investigation using diclofenac in patients undergoing transurethral resection of the prostate”, European Journal of Anaesthesiology, (1987), 4:429-434.
Brogden et al., Drugs 20(1): 24-48 (1980).
Campbell et al., “Intravenous diclofenac sodium”, Anaethesia, (1990), 45:763-766.
Coakley et al., “Use of the platelet function analyser (PFA-100®) to quantify the effect of low dose aspirin in patients with ischaemic heart disease”, Anaesthesia, (2005), 60:1173-1178.
Dahl et al., “High-dose diclofenac for postoperative analgesia after elective caesarean section in regional anaesthesia”, Int. J. Obstet. Anesth., (2002), 11(2):91-94.
Devi et al., “Albumin Microspheres and beta-Cyclodextrin Inclusion Complex Containing Diclofenac Sodium”, Ind. J. Pharm. Sci, (1992), 54:259-261.
Ejnell et al., “Treatment of postoperative pain with diclofenac in uvulopalatopharyngoplasty”, British Journal of Anaesthesia, (1992), 68:76-80.
European Search Report for EP Patent Application No. 07759156.
European Search Report (Appln No. 04257437.6-1219, dated Jun. 15, 2005).
FDA label for BEXTRA® valdecoxib tablets, revised Nov. 2004, downloaded from www.fda.gov.
FDA product label for diclofenac, http://www.drugs.com/pro/ diclofenac.html.
Final Study Report, Randomized, double-blind, placebo and comparator controlled, single-dose, parallel-group comparison of the analgesic efficacy and safety of intravenous DIC075V (diclofenac sodium) injection, ketorolac tromethamine, and placebo following study (Jun. 4, 2009).
Graham, “COX-2 Inhibitors, Other NSAIDs, and Cardiovascular Risk”, JAMA., (2006), 296.
Gunnison et al., “Sulfite Hypersensitivity, a Critical Review”, CRC Critical Reviews in Toxicology, (1987), 17(3):185-214.
Hawley's Condensed Chemical Dictionary, 1997, p. 603.
Heaney et al., “Sequential clot strength analyses following diclofenac in pediatric adenotonsillectomy”, Pediatric Anethesia, (2007), 17:1078-1082.
Hegi et al., “Effect of rofecoxib on platelet aggregation and blood loss in gynaecological and breast surgery compared with diclofenac”, British Journal of Anaesthesia, (2004), 92(4):523-531.
Hersh et al, Clin. Ther. 26(8):1215-27 (2004).
Hyrkäs, “Effect of Preoperative Single Doses of Diclofenac and Methylprednisolone on Wound Healing”, Scand. J. Plast. Reconstr. Hand Surg., (1994), 28:275-278.
Ikeda et al., “Inclusion Complexes of beta-Cyclodextrin with Antiinflammatory Drugs Fenamates in Aqueous Solution”, Chem. Pharm. Bull., (1975), 23(1):201-208.
Jambhekar et al., “The physicochemical characteristics and bioavailability of indomethacin from beta-cyclodextrin, hydroxyethyl-beta-cyclodextrin, and hydroxypropyl-beta-cyclodextrin complexes”, (2004), Int. J. Pharm., 270(1-2):149-66.
Kasrarian et al., “Developing an injectable formula containing an oxygen-sensitive drug: a case study of danofloxacin injectable” Pharm. Dev. Technol., (1999), 4(4):475-80.
Kokki, “Nonsteroidal anti-inflammatory drugs for postoperative pain: a focus on children”, Paediatr. Drugs, (2003), 5(2):103-123.
Kundu et al., “Description of an in Vitro Platelet Function Analyzer—PFA-100™”, Seminars in Thrombosis and Hemostasis, (1995), 21(2):106-112.
Kurozumi et al., “Inclusion Compounds of Non-Steroidal Antiinflammatory and Other Slightly Water Soluble Drugs with alpha-and beta-Cyclodextrins in Powdered Form,” Chem. Pharm. Bull., (1975), 23:3062-3068.
Laitinen et al., “Intravenous Diclofenac Coupled with PCA Fentanyl for Pain Relief after Total Hip Replacement”, Anesthesiology, (1992), 76:194-198.
Laitinen et al., “Effect of a non-steriodal anti-inflammatory drug, diclofenac, on haemostasis in patients undergoing total hip replacement”, Acta. Anaesthesiol. Scand., (1992), 36:486-489.
Leese et al., “Valdecoxib Does Not Impair Platelet Function”, American Journal of Emergency Medicine, (2002), 20(4):275-281.
Legeby et al., “Analgesic efficacy of diclofenac in combination with morphine and paracetamol after mastectomy and immediate breast reconstruction”, Acta. Anaethesiol. Scand., (2005), 49:1360-1366.
Lewis et al., “Dose-response relationships between individual nonaspirin nonsteroidal anti-inflammatory drugs (NANSAIDs) and serious upper gastrointestinal bleeding: a meta-analysis based on individual patient data”, Clin. Pharmacol., (2002), 54:320-326.
Mammen et al., “PFA-100™ System: A New Method for Assessment of Platelet Dysfunction”, Seminars in Thrombosis and Hemostasis, (1998), 24(2):195-202.
Mandell, “General Tolerability and Use of Nonsteroidal Anti-Inflammatory Drugs”, The American Journal of Medicine, (1999), 107(6A):72S-76S.
McCabe et al., “Assessment of the antiplatelet effects of low to medium dose aspirin in the early and late phase after ischaemic stroke and TIA”, Platelets, (2005), 16(5):269-280.
McGettigan et al., “Cardiovascular Risk and Inhibition of Cyclooxygenase. A Systematic Review of the Observational Studies of Selective and Nonselective Inhibitors of Cyclooxygenase 2”, JAMA, (2006), 296(13):E1-E12.
Menasse et al, Scand. J. Rheumatol. Suppl. 22:5-16 (1978).
Maier et al., Rheumatol. Rehabil. Suppl. 2:11-21 (1979).
Mendham et al., “Comparison of diclofenac and tenoxicam for postoperative analgesia with and without fentanyl in children undergoing adenotonsillectomy or tonsillectomy”, Paediatric Anaesthesia, (1996), 6:467-473.
Munsterhjelm et al., “Propacetamol augments inhibition of platelet function by diclofenac in volunteers”, British Journal of Anaesthesia, (2003), 91(3):357-362.
Munsterhjelm et al., “Characterization of inhibition of platelet function by paracetamol and its interaction with diclofenac in vitro”, Acta. Anaesthesiol. Scand., (2005), 49(6):840-846.
Niemi et al., “Comparison of the effect of intravenous ketoprofen, ketorolac and diclofenac on platelet function in volunteers”, Acta. Anaesthesiol. Scand., (1997), 41:1353-1358.
Nekroshus et al., English Abstract of Russian Article, “Preparing the Inclusion Compounds Orthophen and Indomethcin with .beta.-Cyclodextrin and their Derivatographic Analysis”, Farmatsiya Moscow, (1989), 38:29-34.
Nordbladh et al., “Analgesia in tonsillectomy: a double-blind study on pre and post-operative treatment with diclofenac”, Clin. Otolaryngol., (1991), 16:554-558.
Ocana et al., “Potassium Channels and Pain: Present Realities and Future Opportunities”, European Journal of Pharmacology, (2004), 500:203-219.
O'Hanlon et al., “A comparison of the effect of intramuscular diclofenac, ketorolac or piroxicam on post-operative pain following laparoscopy”, European Journal of Anaesthesiology, (1996), 13:404-407.
Orienti et al., “Inclusion Complexes Between Non Steroidal Antiinflammatory Drugs and beta-Cyclodextrin”, Dur. J. Pharm. Biopharm., (1991), 37(2):110-112.
Orienti et al., “Availability of NSAIDH beta-Cyclodextrin Inclusion Complexes”, Arch. Pharm. (Weinheim), (1989), 322:207-211.
Otagiri et al., “Comparative Study on Inclusion Complexation of beta-Cyclodextrin and Tri-O-Methyl-beta-Cyclodextrin with Several Drugs in Aqueous Solution”, Acta Pharm. Suec., (1984), 21:357-366.
Perttunen et al., “I.V. Diclofenac in post-thoracotomy pain”, British Journal of Anaesthesia, (1992), 68:474-480.
Pilotto et al., “The risk of upper gastrointestinal bleeding in elderly users of aspirin and other non-steroidal anti-inflammatory drugs: the role of gastroprotective drugs”, Aging Clin. Exp. Res., (2003), 15(6):494-499.
Pose-Vilarnovo et al., “Modulating Drug Release With Cyclodextrins in Hydroxypropyl methylcellulose gels and tablets”, Journal of Controlled Release, (2004), 94:351-363.
Power et al., “Platelet function after intramuscular diclofenac”, Anaesthesia, (1990), 45:910-919.
Reddy et al. “Beta Cyclodextrin Complexes of Celecoxib: Molecular Modeling, Characterization, and Dissolution Studies,” AAPS Pharm. Sci., vol. 6, No. 1, 7, Mar. 5, 2005, pp. 1-9.
Reed et al., “Lysis of Human Red Blood Cells in the Presence of Various Cosolvents”, Journal of Parenteral Science and Technology, (1985), 39(2):64-69.
Reer et al., “In Vitro Corneal Permeability of Diclofenac Sodium in Formulations Containing Cyclodextrins Compared to the Commercial Product Voltraren Ophtha”, Journal of Pharmaceutical Sciences, (1994), 83(9):1345-1349.
Robinson et al., “Diclofenac and post-tonsillectomy haemorrhage”, Clin. Otolatryngol., (1994), 19:344-345.
Robinson, “Correspondence: Diclofenac and post tonsillectomy haemorrhage”, Clin. Otolatryngol., (1995), 20:483-484.
Romsing et al., “Diclofenac or acetaminophen for analgesia in paediatric tonsillectomy outpatients”, Acta. Anaesthesiol. Scand., (2000), 44(3):291-295.
Rorarius et al., “Diclofenac versus indomethacin given as intravenous infusions: their effect on haemodynamics and bleeding time, and side-effects in healthy subjects”, Annals of Clinical Research, (1985), 17:306-309.
Rorarius et al., “Effects of peri-operatively administered diclofenac and indomethacin on blood loss, bleeding time and plasma prostandoids in man”, European Journal of Anaesthesiology, (1989), 6:335-342.
Saray et al., “Diclofenac and metamizol in postoperative analgesia in plastic surgery”, Acta. Chirurgiae. Plasticae., (2001), 43(3):71-76.
Sia et al., “Combination of suppository diclofenac and intravenous morphine infusion in post-caesarean section pain relief—a step towards balanced analgesia?”, Singapore Med. J., (1997), 38(2):68-70.
Schmidt et al., “Preoperative rectal diclofenac versus paracetamol for tonsillectomy: effects on pain and blood loss”, Acta. Anaesthesiol. Scand., (2001), 45:48-52.
Seymour et al., Eur. J. Clin. Pharmacol. 56:447-452 (2000).
Sun et al., “Effects of Epidural Morphine and Intramuscular Diclofenac Combination in Postcesarean Analgesia: A Dose-Range Study”, Anasth. Analg., (1993), 76:284-288.
Tai et al., “Comparison of controlled-release ketoprofen and diclofenac in the control of post-surgical dental pain”, Journal of the Royal Society of Medicine, (1992), 85:16-18.
Thiagarajan et al. “Blood loss following tonsillectomy in children”, Anaesthesia, (1993), 47:132-135.
Todd et al., Drugs 35(3): 244-85 (1988).
Uekama, “Cyclodextrin Inclusion Compounds: Effects on Stability and Bio-Pharmaceutical Properties”, Elsevier Science Publishers B.V. (Biomedical Division), (1987), pp. 181-193.
Van Hecken et al., “Comparative Inhibitory Activity of Rofecoxib, Meloxicam, Diclofenac, Ibuprofen, and Naproxen on COX-2 versus COX-1 in Healthy Volunteers”, J. Clin. Pharmacol., (2000), 40:1109-1120.
Yee et al., “Platelet hyperractivity generalizes to multiple forms of stimulation”, Journal of Thrombosis and Haemostasis, (2006), 4:2043-2050.
U.S. Appl. No. 11/689,931, Oct. 14, 2011 Final Office Action.
U.S. Appl. No. 11/689,931, Aug. 4, 2011 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,939, Aug. 4, 2011 Supplemental Amendment.
U.S. Appl. No. 11/689,939, Aug. 1, 2011 Amendment and Request for Continued Examination (RCE).
U.S. Appl. No. 11/689,931, Jan. 13, 2012 Amendment and Request for Continued Examination (RCE).
Cwiertni et al., “Stability of Dicolfenac Sodium in the Inclusion Complex with b-Cyclodextrin in the Solid State”, J. Pharm. Pharmacol, 1999, vol. 51, pp. 1213-1218.
U.S. Appl. No. 11/689,939, Aug. 27, 2012 Non-Final Office Action.
U.S. Appl. No. 11/689,931, Aug. 28, 2012 Non-Final Office Action.
Japanese Office Action dated Aug. 29, 2012 from Japanese Application No. 2009-503166, (corresponding to U.S. Appl. Nos. 11/689,931 and 13/106,697).
Kore, et al., “Toxicology of Nonsteroidal Antiinflammatory Drugs”, Veterinary Clinics of North America, Small Animal Practice, 20(2):419-430 (1990).
Gazarian, et al., “Safe use of NSAIDs in infants and children”, Medicine Today, 7(11):7173 (2006).
Rekkas, et al., “Oral and intramuscular absorption of ibuprofen after administration of a freeze-dried ibuprofen 2-hp-beta-CD complex in dogs”, Proceedings of the International Symposium on Controlled Release of Bioactive Materials, 24:557-558 (1997).
FDA Orange Book entry for ibuprofen intravenous solution, downloaded Aug. 20, 2012 from http://www.accessdata.fda.gov.
FDA Orange Book entry for ketorolac injectable solution, downloaded Aug. 20, 2012 from http://www.accessdata.fda.gov.
Routes of Administration Requiring Sterile Formulations, The Pharmaceutics and Compounding Laboratory, downloaded Aug. 20, 2012, from http://pharmlabs.unc.edu.
U.S. Appl. No. 13/153,283, Apr. 29, 2013 Response to Non-Final Office Action.
U.S. Appl. No. 11/689,931, Jul. 10, 2013 Notice of Allowance.
U.S. Appl. No. 13/153,283, Aug. 13, 2013 Final Office Action.
U.S. Appl. No. 14/051,021, filed Oct. 10, 2013.
U.S. Appl. No. 11/689,931, Oct. 9, 2013 Issue Fee payment.
U.S. Appl. No. 13/153,283, Nov. 29, 2012 Non-Final Office Action.
U.S. Appl. No. 11/689,931, Apr. 1, 2013 Amendment and Request for Continued Examination (RCE).
U.S. Appl. No. 11/689,931, Dec. 31, 2012 Final Office Action.
U.S. Appl. No. 11/689,931, Nov. 28, 2012 Response to Non-Final Office Action.
U.S. Appl. No. 10/999,155, Aug. 16, 2011 Notice of Abandonment.
U.S. Appl. No. 10/999,155, Dec. 6, 2010 Final Office Action.
U.S. Appl. No. 10/999,155, Nov. 4, 2010 Response to Non-Final Office Action.
U.S. Appl. No. 10/999,155, May 5, 2010 Non-Final Office Action.
U.S. Appl. No. 10/999,155, Mar. 18, 2010 Amendment and Request for Continued Examination (RCE).
U.S. Appl. No. 10/999,155, Aug. 18, 2009 Notice of Appeal.
U.S. Appl. No. 10/999,155, Feb. 20, 2009 Final Office Action.
U.S. Appl. No. 10/999,155, Sep. 15, 2008 Response to Non-Final Office Action.
U.S. Appl. No. 10/999,155, Jun. 11, 2008 Non-Final Office Action.
U.S. Appl. No. 10/999,155, Oct. 26, 2007 Response to Non-Final Office Action.
U.S. Appl. No. 10/999,155, Jul. 26, 2007 Non-Final Office Action.
Ammon, et al., “Diclofenac does not Interact with Codeine Metabolism in Vivo: A Study of Healthy Volunteers”, BMC Clinical Pharmacology, 2(2):1-10 (2002).
Christensen, et al., “A Double-Blind Placebo-Controlled Comparison of a Novel Formulation of Intravenous Diclofenac and Ketorolac for Postoperative Third Molar Extraction Pain”, Anesth. Prog., 58:73-81 (2011).
Eddaoudi, et al., “Solubilities of the Cyclodextrins in the Presence of Transition Metal Salts”, Journal of Inclusion Phenomena and Molecular Recognition in Chemistry, 26:133-151 (1996).
Eyjolfsson, “Diclofenac Sodium: Oxidative Degradation in Solution and Solid State”, Drug Development and Industrial Pharmacy, 26(4):451-453 (2000).
Gaudiano, et al., “RP-HPLC Study of the Degradation of Diclofenac and Piroxican in the Presence of Hydroxyl Radicals”, Journal of Pharmaceutical and Biomedical Analysis, 32:151-158 (2003).
Jansook, et al. “CD as Solubilizers: Effects of Excipients and Competing Drugs”, Int. J. Pharm, 379(1):32-40 (2009).
Ledwidge, et al., “Effects of Surface Active Characteristics and Solid State Forms on the pH Solubility Profiles of Drug-Salt Systems”, Int. J. Pharm., 174(1-2):187-200 (1998).
Loftsson, et al., “Pharmaceutical Applications of Cyclodextrins: 1. Drug Solubilization and Stabilization”, Journal of Pharmaceutical Sciences, 85(10):1017-1025 (1996).
Martin Del Valle, et al., “Cyclodextrins and Their Uses: A Review”, Process Biochemistry, 39:1033-1046 (2004).
International Search Report and Written Opinion for PCT/IB2004/003918, dated Jun. 15, 2005.
International Preliminary Report on Patentability for PCT/IB2004/003918, dated Sep. 13, 2006.
International Search Report and Written Opinion for PCT/US2005/007354, dated Dec. 22, 2005.
International Preliminary Report on Patentability for PCT/US2005/007354, dated Sep. 13, 2006.
International Search Report and Written Opinion for PCT/US2012/047453, dated Oct. 4, 2012.
U.S. Appl. No. 13/153,283, Feb. 13, 2014 Non-Final Office Action.
Excerpts from Nursing Procedures: Intramuscular Injection Technique, AstraZeneca, 2000; pp. 243-247.
Related Publications (1)
Number Date Country
20110218247 A1 Sep 2011 US
Provisional Applications (1)
Number Date Country
60786486 Mar 2006 US
Continuations (1)
Number Date Country
Parent 11689931 Mar 2007 US
Child 13106697 US