WO1999032151A1 - Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions - Google Patents

Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions

Info

Publication number
WO1999032151A1
WO1999032151A1 PCT/US1997/023865 US9723865W WO9932151A1 WO 1999032151 A1 WO1999032151 A1 WO 1999032151A1 US 9723865 W US9723865 W US 9723865W WO 9932151 A1 WO9932151 A1 WO 9932151A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
polymer
polyoxyalkylene
weight
copolymer
Prior art date
Application number
PCT/US1997/023865
Other languages
French (fr)
Inventor
Stephen G. Flore
Luis A. Dellamary
Lorraine E. Reeve
Jeffry G. Weers
Original Assignee
Alliance Pharmaceutical Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alliance Pharmaceutical Corporation filed Critical Alliance Pharmaceutical Corporation
Priority to EP97951781A priority Critical patent/EP1039932A1/en
Priority to US09/582,508 priority patent/US6280745B1/en
Priority to CA002316248A priority patent/CA2316248A1/en
Priority to AU55335/98A priority patent/AU753206B2/en
Priority to JP2000525141A priority patent/JP2001526246A/en
Priority to PCT/US1997/023865 priority patent/WO1999032151A1/en
Priority to AU19438/99A priority patent/AU1943899A/en
Priority to PCT/US1998/027410 priority patent/WO1999032152A2/en
Publication of WO1999032151A1 publication Critical patent/WO1999032151A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0065Forms with gastric retention, e.g. floating on gastric juice, adhering to gastric mucosa, expanding to prevent passage through the pylorus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/04Macromolecular materials
    • A61L31/06Macromolecular materials obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels

Definitions

  • the present invention generally relates to pharmaceutical preparations and methods of their use. More particularly, the present invention relates to preparations suitable for the reduction of adhesion formation in mammals or the delivery of pharmaceutically active compounds.
  • Aqueous liquids which can be applied at room temperature in a free flowing state but which forms a semi- solid gel when warmed to body temperature have been used in such capacities for some time.
  • Such systems combine ease of application with greater retention at the site of application than the use of exclusively free flowing vehicles.
  • Pluronic ® polyols are used in aqueous compositions to provide thermally gelling aqueous systems. Adjusting the concentration provides the desired sol-gel transition temperature. More particularly, the lower the concentration of the incorporated polymer the higher the sol-gel transition temperature.
  • Adhesions are thought to form following a trauma or injury to the peritoneum. This results in increased vascular permeability, which produces an inflammatory exudate and results in the formation of a fibrin matrix.
  • the fibrin matrix is removed by fibrinolysis, and subsequent fibroblast proliferation results in remesothelialization.
  • the fibrinolytic process is suppressed and the fibrin matrix may persist. If it persists until about day three, significant collagen deposition within the fibrin matrix, can lead to adhesion formation.
  • dextran 70 in preventing post-operative adhesions was shown to be limited to the more dependent regions of the pelvis.
  • TC-7 Johnson and Johnson Products, Inc., New Brunswick, N.J.
  • Other solid sheet devices include polytetrafluoroethylene (Gore-Tex®, W. L. Gore) and crosslinked hyaluronic acid (Seprafilm ⁇ Genzyme Corp.).
  • Chondroitin sulfate and sodium carboxymethyl cellulose have also been used to prevent the formation of postoperative adhesions in the rabbit uterus (Oelsner et al., J Reprod. Med. 32:812-814 (1987)). Chondroitin sulfate solutions have also been proposed for intraperitoneal use in the prevention of adhesions in rabbits
  • aqueous gel compositions comprising polyalkylene polymers have been shown to successfully reduce adhesions (U.S. Pat. No. 5,366,735, incorporated herein by reference). These compositions can be applied below room temperature as a liquid and form semi-solid gels when warmed to body temperature.
  • precise control of the sol-gel transition temperature and dissolution rate of the gel within the physiological environment still present problems in many cases. Accordingly, despite these previous efforts, a need exists for improved means to treating and/or preventing post-surgical adhesions. As such, it is an object of the present invention to provide polymeric gel compositions which allow for precise control of the sol-gel transition temperature and/or dissolution rate of the gel once formed.
  • the present invention accomplishes these and other objectives by providing polymeric compositions that exhibit well defined sol-gel transition temperatures (or defined ranges of temperatures) and/or established dissolution rates.
  • the disclosed compositions generally comprise at least one constitutive polymer and at least one modifier polymer that may be used to modify or control the dissolution rate of gel once it has been formed.
  • the compositions of the present invention comprise at least one constitutive polymer and at least one hydrophilic co-surfactant whereby the gelation temperature (or sol-gel transition temperature) of the composition may be controlled or modified.
  • compositions may comprise at least one constitutive polymer in combination with both at least one modifier polymer and at least one hydrophilic co-surfactant to provide preparations having both selected gelation temperatures and superior dissolution times.
  • compositions may comprise at least one constitutive polymer in combination with both at least one modifier polymer and at least one hydrophilic co-surfactant to provide preparations having both selected gelation temperatures and superior dissolution times.
  • preferred embodiments of the invention will comprise the aforementioned preparations and at least one bioactive agent.
  • the polyphase preparations of the present invention may be used to retard or prevent the formation of scar tissue or adhesions in a mammal, for the prolonged delivery of a bioactive agent or both.
  • the present invention comprises methods for the reduction of adhesion or scar tissue formation comprising the administration of the disclosed preparations to a mammal in need thereof.
  • compositions of the present invention comprise methods of delivering a bioactive agent to a mammal comprising administering the disclosed polyphase compositions incorporating a pharmaceutically effective amount of at least one bioactive agent to a mammal in need thereof.
  • the compositions of the present invention will be administered as a relatively free flowing liquid that gels upon contact with the mammalian tissue to provide a viscoelastic semi-solid barrier or mask that may remain in place for an extended period.
  • the constitutive polymer will be a polyoxyalkylene copolymer. More particularly, in selected embodiments the constitutive polymer will be selected from the group consisting of polyoxyalkylene block copolymers, polyoxyalkylene polyethers and combinations thereof. In especially preferred embodiments of the invention, the constitutive polymer will comprise Poloxamer 407.
  • the constitutive polymer or polymers may be present at any concentration that provides the desired gel viscosity and/or viscoelastic properties. Preferably, the constitutive polymers are present in a concentration which, when combined with the other components of the preparation, allows for the administration of the composition as a relatively free flowing liquid which gels upon contact with mammalian tissue.
  • modifier polymers compatible with the present invention comprise any polymeric entity capable of slowing or retarding the dissolution rate of the constitutive polymer once it has gelled. That is, the modifier polymers of the present invention comprise any polymer that, when added to the constitutive polymer(s), provides for a slower dissolving or diffusing gel when compared with a gel formed from pure constitutive polymer(s) under equivalent conditions.
  • Preferred modifier polymers typically have a relatively high average molecular weight on the order of tens or hundreds of thousands.
  • modifier polymers While a large number of polymeric compounds are suitable for use as modifier polymers, particularly preferred compounds comprise cellulose ethers (carboxymethyl cellulose) and Carbopols (e.g. Carbopol 940-NF).
  • the absolute incorporated concentration of the modifier polymers in the compositions of the present invention is not critical and may be adjusted to provide the desired dissolution rates and/or retention times.
  • compositions disclosed herein may further comprise one or more hydrophilic co-surfactants which may be used to modify the gelation temperature (sol-gel transition temperature) of the resulting preparation. More particularly, selected hydrophilic co-surfactant(s) may be added to compositions comprising a constitutive copolymer(s) or compositions comprising constitutive copolymer(s) and modifier polymer(s) to alter or modify the gelation temperature of the resulting composition when compared to similar compositions not comprising the hydrophilic co-surfactant.
  • the hydrophilic may be added at an effective concentration to lower the gelation temperature of the composition so as to provide for more rapid and complete gelation upon contact with the relatively high temperature mammalian tissue.
  • hydrophilic co-surfactants include fatty acid soaps such as sodium laureate, sodium caprate or sodium caprylate.
  • combinations of hydrophilic co- surfactants may be incorporated in the compositions of the present invention to provide the desired transition temperature or transition temperature range.
  • bioactive agents compatible with the present invention include, but are not limited to, antibiotics, antivirals, mydriatics, antiglaucomas, anti-inflammatories, -mtihistaminics, antineoplastics, anesthetics, ophthalmic agents, enzymes, cardiovascular agents, polynucleotides, genetic material, viral vectors, immunoactive agents, imaging agents, immunosuppressive agents, peptides, proteins, physiological gases, gastrointestinal agents and combinations thereof.
  • compositions may comprise one or more humectants, bactericides, bacteriostatic agents, fibrinolytic agents or agents effective in preventing leukocyte migration into the area of surgical injury.
  • Pharmaceutically effective amounts of the selected bioactive agents may be determined using techniques well known in the art. It will further be appreciated that the bioactive agents may be incorporated in the form of relatively insoluble solid particulates or associated with insoluble polymeric particulates.
  • the preparations of the present invention may be administered to a patient using a route of administration selected from the group consisting of topical, subcutaneous, pulmonary, synovial, intramuscular, intraperitoneal, nasal, vaginal, rectal, aural, oral and ocular routes.
  • the administered composition preferably gels upon contact with the relatively warm mammalian tissue and may act as a depot for the prolonged delivery of one or more incorporated bioactive agents.
  • the gelled compositions may act as a barrier or film which prevents or retards the formation of adhesion or scar tissue.
  • the present invention provides particularly effective methods for the prevention of post-surgical and other adhesion formation, administration of pharmaceutically effective amounts to the peritoneal, pelvic or pleural cavity is especially preferred.
  • adhesions are often associated with injury to mammalian organs, those skilled in the art will appreciate that the compositions are particularly useful when applied to the selected area during or immediately following surgery.
  • compositions of the present invention may further comprise pharmaceutically acceptable stabilizers, preservatives and buffers, preferably in an amount sufficient to maintain the pH of the composition at about pH 7.4 ⁇ 0.2.
  • Figure 1 is a graphical representation of an exemplary phase diagram for a prior art composition comprising a constitutive polymer
  • Figure 2 illustrates an equilibrium phase diagram of 20% w/w poloxamer 407 (407F) in tromethamine/maleate buffer with added sodium caprate with arrows indicating that the cloud point temperature is greater than the highest temperature measured, i.e. 140 °C;
  • Figure 3 is a graphical representation of a gelation profile of 1%, 3% and 5% (w/w) sodium caprylate in 20% w/w poloxamer 407 (407F) in hypotonic tromethamine/maleic acid buffer
  • Figure 4 is a graphical representation illustrating the effect of fatty acid soap concentration on the lower gelation temperature (LGT) of 20% w/w poloxamer 407F solutions for soaps of varying alkyl chain lengths and degrees of saturation.
  • LGT lower gelation temperature
  • compositions and methods are disclosed herein for delivering bioactive agents and/or reducing post-surgical adhesion formation reformation in mammals following injury to the organs or tissues, particularly those of the peritoneal, pelvic or pleural cavity.
  • the compositions of the invention are also useful in reducing adhesion formation/reformation in other body spaces such as the subdural, extraocular, intraocular, otic, synovial, tendon sheath, or those body spaces created either surgically or accidentally.
  • the concentration of the constitutive polymer in the disclosed compositions may be adjusted to take advantage of the gelation properties of certain polyoxyalkylene polymers.
  • compositions may be formed in accordance with the teachings herein that do not gel or thicken following application to the selected area or tissue.
  • the osmolality and pH of the compositions are adjusted to match the pH and osmotic pressure of mammalian bodily fluids, i.e. approximately pH 7.4.
  • the constitutive polymer i.e., a polyoxyalkylene block copolymer
  • compositions may also be used as a distending medium during diagnostic or operative endoscopic procedures, such as, for example, for intrauterine procedures.
  • diagnostic or operative endoscopic procedures such as, for example, for intrauterine procedures.
  • certain aqueous concentrations of the preferred polyoxyalkylene block copolymers form a clear gel, their use is well suited for visualization of interior cavities.
  • the disclosed formulations provide a barrier between tissues for hours or days. Because they are applied as liquids, they are easier to use, particularly for laparoscopic surgical procedures. That is, the compositions of the instant invention may be administered though a relatively small incision using a cannula or catheter assembly.
  • the disclosed compositions of the present invention are preferably aqueous based preparations.
  • the compositions typically comprise water in an amount of from about 60% to about 90%, by weight, preferably, about 70% to about 85%, by weight, and most preferably, about 75% to about 82% by weight, based upon the total weight of the composition.
  • the terms "peritoneal” and “abdominal” cavity are used as synonyms, as are the terms “pleural” and "thoracic” cavity.
  • polyalkylene block polymers include those polymers which form clear gels at mammalian body temperatures but are liquids at ambient temperatures or below.
  • gel is defined as a solid or semisolid colloid containing a certain quantity of water. The colloidal solution with water is often called a "hydrosol”.
  • the constitutive polymer will be a polyoxyalkylene polymer. More particularly, in selected embodiments the constitutive polymer will be selected from the group consisting of polyoxyalkylene block copolymers, polyoxyalkylene polyethers and combinations thereof. In especially preferred embodiments of the invention, the constitutive polymer will comprise poloxamer 407.
  • the constitutive polymer or polymers may be present at any concentration that provides the desired gel viscosity and/or viscoelastic properties. Preferably, the constitutive polymers are present in a concentration which, when combined with the other components of the preparation, allows for the administration of the composition as a relatively free flowing liquid which gels upon contact with mammalian tissue.
  • compositions comprise one or more polyoxyalkylene block copolymers of the formula
  • A is a polyoxyalkylene moiety; x is at least 2; Y is derived from water or an organic compound containing x reactive hydrogen atoms;
  • E is a polyoxyethylene moiety; n has a value such that the average molecular weight of A is at least about 500; and the total average molecular weight of the copolymer is at least about 5000.
  • the polyoxyalkylene moiety A has an oxygen/carbon atom ratio of less than 0.5.
  • A is derived from an alkylene oxide selected from the group consisting of butylene oxide, propylene oxide or a mixture thereof.
  • A is a polyoxypropylene moiety, and preferably has an average molecular weight of from about 3,000 to about 4,000 g mol "1 .
  • the polyoxyethylene moiety E preferably constitutes from about 60 to about 85% by weight of the copolymer, more preferably at least about 70%.
  • Y is derived from a water soluble organic compound having 1 to about 6 carbon atoms. In another embodiment, Y is derived from an organic compound selected from the group consisting of propylene glycol, glycerin, pentaerythritol trimethylolpropane, ethylenediamine and mixtures thereof.
  • the copolymer has the formula: HO(C 2 H 4 O)£(C 4 H 8 O) fl (C 2 H 4 O) 6 H (H)
  • a and b are integers such that (C 4 H 8 O) ⁇ has a molecular weight of at least about 500.
  • Useful polyoxyalkylene block copolymers which will form gels in aqueous solutions can be prepared using a hydrophobe base (such as A in Formulas (I) and (II)) derived from propylene oxide, butylene oxide or mixtures thereof. These block copolymers and representative methods of preparation are further generally described in U.S. Pat. Nos. 2,677,700; 2,674,619; and U.S. Pat. No. 2,979,528, inco ⁇ orated herein by reference.
  • the polyoxybutylene-based block copolymers useful in the compositions of the invention are prepared by first condensing 1,2 butylene oxide with a water soluble organic compound initiator containing 1 to about 6 carbon atoms such as 1,4 butylene glycol or propylene glycol and at least 2 reactive hydrogen atoms to prepare a polyoxyalkylene polymer hydrophobe of at least about 500, preferably at least about 1000, most preferably at least about 1500 average molecular weight. Subsequently, the hydrophobe is capped with an ethylene oxide residue. Specific methods for preparing these compounds are described in U.S. Pat. No. 2,828,345 and British Patent No. 722,746, both of which are herein incorporated by reference.
  • compositions comprise polyoxyethylene- polyoxypropylene block copolymers of the formula (III):
  • compositions comprise a polyxyethylene-polyoxyproplyene block copolymer of formula (III), having a polyoxyproplyene hydrophobe base average molecular weight of about 4000, a total average molecular weight of about 12,000 and containing oxyethylene groups in the amount of about 70% by weight of the total weight of the copolymer.
  • poloxamer 407 is a tri-block copolymer containing two polyoxyethylene blocks flanking a central polyoxypropylene block.
  • the USP material has an average molecular formula of (EO) 101 -(PO) 56 -(EO) 101 , and average molecular weight of ca. 12,000 g mol "1 .
  • poloxamer 407 self-assembles so as to remove contact between the polyoxypropylene groups and water (i.e.
  • the self-assembly is driven by the hydrophobic effect).
  • the self-assembled units are termed micelles.
  • the structure of the micelles and the interactions between them is strongly dependent on temperature.
  • a large increase in solution viscosity i.e. gel-phase formation
  • Gel phase formation occurs as a result of organization of the micelles into a three- dimensional cubic array.
  • the copolymer has the formula:
  • R is H(OC 2 H 4 ) b (OC 3 H 6 ) a -; and a and b are integers such that the hydrophobe base represented by (C 3 H 6 O) a has a sum average molecular weight of at least about 2000, about 3 to about 5%.
  • the hydrophobe base is prepared by adding propylene oxide for reaction at the site of the four reactive hydrogen atoms on the amine groups of ethylenediamine. An ethylene oxide residue is used to cap the hydrophobe base.
  • the polyoxyethylene chain constitute from about 60 to about 85% by weight of the colpolymer, preferably at least about 10%. It is further preferred that the copolymer have a total average molecular weight of at least about 5000, preferably from about 9,000 to about 15,000 as specified in the USP).
  • aqueous solutions which form gels of the polyoxyalkylene block copolymers is well known.
  • Either a hot or cold process for forming the solutions can be used.
  • a cold technique involves the steps of dissolving the polyoxyalkylene block copolymer at a temperature of about 5° to about 10° C in water. When solution is complete the system is brought to room temperature whereupon it forms a gel. If the hot process of forming the gel is used the polymer is added to water heated to a temperature of about 75 °C to about 85 °C, with slow stirring until a clear homogeneous solution is obtained. Upon cooling, a clear gel is formed.
  • Block copolymer gels containing polyoxybutylene hydrophobes must be prepared by the above hot process, since these will not liquefy at low temperatures.
  • the organic compound initiator which is utilized in the preparation of the polyoxyalkylene block copolymers generally is water or an organic compound, and can contain a plurality of reactive hydrogen atoms.
  • Y in formulas (I) and (II) above is defined as derived from a water soluble organic compound having 1 to about 6 carbon atoms and containing x reactive hydrogen atoms where x has a value generally, of at least 1, preferably, a value of at least 2.
  • Y is derived from water soluble organic compounds having at least two reactive hydrogen atoms
  • water soluble organic compounds such as propylene glycol, glycerin, pentaerythritol, trimethylolpropane, ethylenediamine, and mixtures thereof and the like.
  • the oxypropylene chains can optionally contain small amounts of at least one of oxy ethylene or oxy butylene groups.
  • Oxy ethylene chains can optionally contain small amounts of at least one of oxypropylene or oxybutylene groups.
  • Oxy butylene chains can optionally contain small amounts of at least one of oxy ethylene or oxypropylene groups.
  • the physical form of the polyoxyalkylene block copolymers can be a viscous liquid, a paste or a solid granular material depending upon the molecular weight of the polymer.
  • the present compositions may comprise other polyoxyalkylene polymers which form gels at low concentrations in water.
  • polymers are described in U.S. Pat. No. 4,810,503, incorporated herein by reference. These polymers are prepared by capping conventional polyoxyalkylene polyether polyols with an alphaolefin epoxide having an average of about 20 to about 45 carbon atoms, or mixtures thereof. Aqueous solutions of these polymers gel in combination with surfactants, which can be ionic or nonionic. The combination of the capped polyether polymers and the surfactants provide aqueous gels at low concentrations of the capped polymer and surfactant which generally do not exceed 10% by weight total.
  • copolymer polyether polyols are prepared by preparing block or heteric intermediate polymers of ethylene oxide and at least one lower alkylene oxide having 3 to 4 carbon atoms as intermediates. These are then capped with the alpha-olefin epoxide. Ethylene oxide homopolymers capped with the alpha-olefin oxides are also useful as intermediates.
  • the heteric copolymer intermediate is prepared by mixing ethylene oxide and at least one lower alkylene oxide having 3 to 4 carbon atoms with a low molecular weight active hydrogen-containing compound initiator having at least two active hydrogens and preferably,
  • 2 to 6 active hydrogen atoms such as a polyhydric alcohol, containing from 2 to 10 carbon atoms and from 2 to 6 hydroxyl groups, heating said mixture to a temperature in the range of about 50° C to 150° C, preferably, from 80° C to 130°, under an inert gas pressure, preferably, from about 30 psig to 90 psig.
  • a block copolymer intermediate is prepared by reacting either the ethylene oxide or the alkylene oxide having 3 to 4 carbon atoms with the active hydrogen-containing compound followed by reaction with the other alkylene oxide.
  • the ethylene oxide and the alkylene oxides having from 3 to 4 carbon atoms are used in the intermediates in amounts so that the resulting polyether product will contain at least 10 percent by weight, preferably about 70 percent to about 90 percent by weight, ethylene oxide residue.
  • the ethylene oxide homopolymer intermediate is prepared by reacting ethylene oxide with the active hydrogen-containing compound.
  • the reaction conditions for preparing the block copolymer and ethylene oxide homopolymer intermediates are similar to those for the heteric copolymer intermediate.
  • the temperature and pressure are maintained in the above ranges for a period of about one hour to ten hours, preferably one to three hours.
  • the alpha-olefin oxides which are utilized to modify the conventional polyether intermediates are those oxides, and commercially available mixtures thereof, generally containing an average of about 20 to 45, preferably about 20 to 30, carbon atoms.
  • the amount of alpha-olefin required to obtain the more efficient capped polyethers is generally about 0.3 to 10 percent, preferably about 4 to 8 percent, of the total weight of the polyethers. Further description regarding the preparation of heteric and block copolymers of alkylene oxides and ethylene oxide homopolymers is described in the art (U.S. Pat. Nos.
  • One major advantage of the present invention is that the desired gelation temperatures and viscosity of the resulting gels may be adjusted through the addition of modifier polymers and hydrophilic co-surfactants. This allows the use of lower concentrations of constitutive polymer without markedly reducing the ultimate gel characteristics of the composition.
  • exemplary concentrations of constitutive polymer may range from approximately 2% w/w to 50% w/w and more preferably from 4% to 30% w/w and even more preferably from 16% to 28% w/w.
  • any biocompatible polymeric entity that modifies the dissolution time of the gel resulting from the administration of the compositions of the present invention may be used in accordance with the teachings herein.
  • preferred modifier polymers to alter the dissolution time should preferably have the following characteristics: (a) high molecular weight; (b) effective swelling in water but poor dissolution: (c) compatibility with the constitutive polymer and, in particular, poloxamers; and (d) stability to extremes in heat and pH.
  • alter the dissolution time is held to mean the alteration of the gel dissolution time in vitro or in vivo with respect to a gel comprising constitutive polymer without the modifier polymer under similar conditions.
  • the alteration of dissolution times or release rates of the constitutive polymer from the gel matrix may be used to optimize formulations for antiadhesion applications as well as for other applications including controlled drug delivery.
  • release is a function of several physicochemical characteristics within the gel, and can be modified by the addition of high molecular weight polymers such as sodium carboxymethyl cellulose, polyacrylates (i.e. Carbopols) or other polyester based polymers. It appears that the dissolution rate is modified by the formation of a strong polymeric matrix (i.e. the modifier polymeric matrix) that controls the release of the constitutive polymer via diffusion through the formed modifier polymer interstices. One possible reason for this effect may be that the constitutive polymer has to diffuse around the long linear molecules of the incorporated modifier polymer.
  • the selected modifier polymer(s) have a molecular weight greater than or equal to approximately 500,000 although modifier polymers of much lower molecular weight (i.e. on the order of 50,000).
  • the selected modifier polymers will combine a relatively high molecular weight with a biodegradable moiety in their structure to speed excretion.
  • High molecular weight polylactic-glycolide copolymers which are broken down by hydrolytic decomposition are one example of such a polymer. It should be emphasized that these modifier polymers may also be used to slow the dissolution (and hence prolong delivery time) of any incorporated bioactive agent.
  • exemplary polymers compatible with the teachings herein include, but are not limited to: poly(acrylic acid), poly(styrene sulfonate), carboxymethylcellulose, poly(vinyl alcohol), poly(ethylene oxide), poly(vinylpyrrolidone), shellac, cellulose acetate phthalate, cellulose acetate succinate, polyvinyl acetate phthalate, hydroxypropylmethylcellulose acetate, poly(methacrylic acid-co- methylmethacrylate), poly(methyl acrylate), poly(methyl methacrylate), poly(glutamic acid), poly(lactic acid), poly(lactic-glycolide), poly(glycolic acid), poly( ⁇ -caprolactone), poly( ⁇ - hydroxybutyric acid), poly( ⁇ -hydroxy valeric acid), polydioxanone, poly(ethylene terephthalate), poly (malic acid), poly(tartronic acid), poly
  • any pharmaceutically acceptable salt of the foregoing compounds may be used in the disclosed compositions with compromising the effectiveness thereof.
  • the modifier polymers of the present invention may be used in suprisingly low concentrations to provide extended dissolution times or release times.
  • the selected modifier polymers are preferably inco ⁇ orated in a range between about 0.05% and about 25% by weight and more preferably in a range of from approximately 0.5% to approximately 5% by weight.
  • the absolute amount of modifier polymer included in the composition will depend on factors such as the constitutive polymer selected, the molecular weight of the modifier polymer and the physiochemical properties of the various composition components. These determinations are well within the purview of the skilled artisan and may easily be determined without undue experimentation.
  • hydrophilic Co-Surfactants Yet another aspect of the present invention comprises the addition of a hydrophilic co- surfactant to the disclosed compositions (i.e. constitutive polymer preparations and constitutive polymer + modifier polymer preparations) to alter the physiochemical properties thereof. That is, the inco ⁇ oration of a hydrophilic co-surfactant in accordance with the teachings herein may provide several advantages over prior art formulations. These advantages are most easily understood in conjunction with a graphical representation of a polyphase system of the instant invention and examples set forth below.
  • Fig. 1 a phase diagram for a constitutive polymer solution (poloxamer 407) is shown.
  • the lower gelation temperature (LGT) refers to the temperature at which the poloxamer micelles (sol phase) self-assemble into the cubic array (i.e. the gel phase).
  • UGT upper gelation temperature
  • the micelles change their shape from spheres to prolates, thereby negating their ability to assemble in a cubic packing. This leads to the reformation of the low viscosity sol phase.
  • Above another critical temperature, termed the cloud point (CP) the micelles separate into their own coacervate phase in excess water. The solution clouds due to mixing of the two insoluble phases.
  • the lower gelation temperature (LGT) of the constitutive polymer solutions in water is largely dependent upon the total constitutive polymer concentration, such that increases in concentration lead to decreases in the LGT.
  • Fractionation of the constitutive polymer fractionated using organic phase separation or other means known in the art, such as described, for example, in Textbook of Polymer Science. F. Billmeyer, Wiley-Interscience, pp. 45-56 (1971)
  • CMC carboxymethylcellulose
  • FloGel 28 (28% w/w poloxamer 407) has an LGT of 13 °C, and is currently applied surgically at a temperature of 0°C. Therefore, application of the product will have to be done in a timely fashion to avoid gelation in the application catheter. Additionally, it has been hypothesized that increases in the LGT to a temperature close to or above room temperature may be advantageous.
  • hydrophilic co-surfactants comprising fatty acid soaps are particularly compatible with the present invention.
  • long chain, saturated soaps appear to be particularly efficient at altering the phase behavior to provide the desired composition characteristics.
  • the rheological properties of the gelled compositions of the present invention are unaltered by the presence of the fatty acid soaps, indicating that, as long as the critical packing volume of the cubic phase is exceeded, the rheology will remain virtually unchanged.
  • the addition of hydrophilic co-surfactants to the disclosed polyphase systems provides an efficient method for modifying the gelation temperature and cloud point temperature. These changes in phase behavior are particularly advantageous for a drug delivery vehicle or antiadhesion product as they allow for storage and application at temperatures near room temperature. Moreover, these characteristics reduce the potential for significant syneresis during terminal sterilization.
  • preferred embodiments of the present invention may comprise effective amounts of at least one hydrophilic co-surfactant.
  • the inco ⁇ orated hydrophilic co-surfactant will comprise a fatty acid soap.
  • fatty acid soaps are GRAS (generally regarded as safe) materials, present naturally in the human body, and included in many pharmaceutical products including large volume parenterals (e.g. Fluosol ® ). Their toxicological profile is well understood and, at the concentrations compatible with the present invention, they pose no toxicological risk. While several compounds comprising fatty acids are useful in the present invention, especially compatible fatty acid soaps comprise sodium oleate, sodium laurate, sodium caprate, sodium caprylate and combinations thereof.
  • the hydrophilic co-surfactants of the present invention may be inco ⁇ orated in relatively low concentrations to provide the desired gelation properties. It will be appreciated that the selected hydrophilic co-surfactant or surfactants may comprise any concentration that provides for the preferred gelation temperatures. However, exemplary concentrations of hydrophilic co-surfactants compatible with the instant invention are typically in a range between about 0.05% and about 25% by weight and more preferably in a range of from approximately 0.5% to approximately 5% by weight.
  • the preparations also provide for the efficient delivery of bioactive agents.
  • bioactive agents may increase the solubilization and bioavailability of inco ⁇ orated pharmaceutical compounds.
  • the micelle core of the gelled compositions of the present invention may serve as a reservoir for solubilizing nonpolar solutes such as hydrophobic drugs.
  • the micelles may also self-assemble to form stiff gels above a critical temperature. As previously discussed, gel formation appears to occur when the micelles behave as hard spheres in a close-packed simple cubic array.
  • poloxamer gels may also be an ideal drug delivery vehicle, owing to their low toxicity and ability to impede drug diffusion.
  • compositions disclosed herein may further optionally comprise one or more pharmaceutically acceptable adjuvants such as a humectant, a bactericide, a bacteriostatic agent, an antihistamine, or a decongestant, an agent to prevent leucocyte migration into the area of surgical injury, or a fibrinolytic agent.
  • humectants include, but are not limited to, glycerin, propylene glycol and sorbitol.
  • Useful bactericides include, by way of example, antibacterial substances such as ⁇ -lactam antibiotics, such as cefoxitin, n- formamidoyl thienamycin and other thienamycin derivatives, tetracyclines, chloramphenicol, neomycin, gramicidin, bacitracin, sulfonamides; aminoglycoside antibiotics such as gentamycin, kanamycin, amikacin, sisomicin and tobramycin; nalidixic acids and analogs such as norfloxacin and the antimicrobial combination of fluoroalanine/pentizidone; nitrofurazones, and the like.
  • antibacterial substances such as ⁇ -lactam antibiotics, such as cefoxitin, n- formamidoyl thienamycin and other thienamycin derivatives, tetracyclines, chloramphenicol, neomycin, gramicidin, bacitracin, s
  • Antihistamines and decongestants such as pyrilamine, chlo ⁇ heniramine, tetrahydrozoline, antazoline, and the like, can also be used in admixtures as well as anti-inflammatories such as cortisone, hydrocortisone, beta-methasone, dexamethasone, fluocortolone, prednisolone, triamcinolone, indomethacin, sulindac, its salts and its corresponding sulfide, and the like. Both steroidal and nonsteroidal compounds are particularly compatible with the compositions and methods of the present invention. With regard to the latter, ketoprofen, indomethacin and tolmetin sodium are particularly preferred.
  • Nitric oxide donors such as nononates and nitrosylated compounds may also be inco ⁇ orated.
  • Useful leucocyte migration preventing agents which can be used in admixtures include but are not limited to silver sulfadiazine, acetylsalicylic acid, indomethacin and Nafazatrom.
  • Useful fibrinolytic agents include urokinase, streptokinase, tissue plasminogen activator (TPA) and acylated plasmin.
  • compatible bioactive agents comprise both hydrophilic and lipophilic compounds including antibiotics, antivirals, mydriatics, antiglaucomas, anti- inflammatories, antihistaminics, antineoplastics, anesthetics, ophthalmic agents including anti- glaucomics, enzymes, cardiovascular agents, polynucleotides, genetic material, viral vectors, immunoactive agents, imaging agents, immunosuppressive agents, peptides, proteins, physiological gases, gastrointestinal agents and combinations thereof.
  • hydrophilic and lipophilic compounds including antibiotics, antivirals, mydriatics, antiglaucomas, anti- inflammatories, antihistaminics, antineoplastics, anesthetics, ophthalmic agents including anti- glaucomics, enzymes, cardiovascular agents, polynucleotides, genetic material, viral vectors, immunoactive agents, imaging agents, immunosuppressive agents, peptides, proteins, physiological gases, gastrointestinal agents and combinations thereof.
  • the preparations of the present invention are uniquely suited for various administrative techniques such as ocular, oral, pulmonary, rectal, synovial, subcutaneous, intramuscular, intraperitoneal, nasal, vaginal, or aural administration of medicaments or diagnostic compounds, they are compatible for use with a wide variety of bioactive agents.
  • bioactive agents for example, ophthalmic applications involving topical administration of the disclosed preparations are particularly preferred.
  • the foregoing list of compounds is exemplary only and not intended to be limiting. It will also be appreciated by those skilled in the art that the proper amount of bioactive agent and the timing of the dosages may be determined for the formulations in accordance with already-existing information and without undue experimentation.
  • the compositions are applied to surgically injured tissue as an aqueous solution which upon contact with living mammalian tissue forms a firm, adherent gel.
  • the composition is a viscous liquid or paste
  • these compositions can be applied without dilution to areas of surgical injury in the abdominal or thoracic cavities.
  • the formulations adhere to the site of tissue injury and reduce or prevent the formation of postsurgical adhesions during the healing process.
  • the preparations of the invention may also be used to deliver therapeutic and diagnostic agents to the gastrointestinal tract by, for example, the oral or direct routes of administration.
  • a contemplated example would be the delivery of antibiotics to the lining of the gastrointestinal tract in the treatment of Heliobacter pylori infections.
  • H. pylori has been implicated in the cause of gastric ulcers and stomach cancer.
  • Antibiotics effective in the treatment of H pylori infections could be administered in the form of a free flowing liquid that gels and adheres to the sites of infection.
  • compositions of the present invention may further contain preservatives, cosolvents, suspending agents, viscosity enhancing agents, ionic- strength and osmolality adjustors and other excipients in addition to buffering agents.
  • Suitable water soluble preservatives which may be employed are sodium bisulfite, sodium thiosulfate, ascorbate, benzalkonium chloride, chlorabutanol, thimerosal, phenylmercuric borate, parabens, benzylalcohol phenylethanol and others. These agents may be present, generally, in amounts of about 0.001% to about 5% by weight and, preferably, in the amount of about 0.01 to about 2% by weight.
  • Suitable buffering agents or salts useful in maintaining p ⁇ include alkali or alkaline earth metal carbonates, chlorides, sulfates, phosphates, bicarbonates, citrates, borates, acetates and succinates such as sodium phosphate, citrate, borate, acetate, bicarbonate, carbonate and tromethamine (TRIS).
  • these agents are present in amounts sufficient to maintain the p ⁇ of the system at 7.4+ 0.2 and preferably, 7.4.
  • the buffering agent can be as much as 5% by weight.
  • preparations of the present invention may be sterilized, for example, by heat, irradiation, ultrafiltration or combinations of any of these or equivalent techniques.
  • preparations of the invention may be sterilized, for example, by autoclaving at 121 °C for 15 minutes or by filtration through a 0.22 mm filter.
  • the high bioavailability bioactive preparations of the present invention may advantageously be supplied to the physician in a sterile prepackaged form. More particularly, the formulations may be supplied as stable, preformed preparations, ready for administration or as separate, ready to mix components. When supplied as components the final preparation of the polyphase material could easily be performed in the pharmacy just prior to administration.
  • compositions comprising a constitutive polymer (poloxamer 407) and a modifier polymer (sodium carboxymethylcellulose) were prepared by dissolving the poloxamer in distilled water (4°C) to give a concentration of 28% by weight in accordance with the cold process described above for forming aqueous solutions.
  • Example 1 The following test procedure was utilized to determine the effect of the formulations of Example 1 on surgically injured rats.
  • Female Sprague-Dawley rats having a 300-400 gram body weight were anesthetized with pentobarbital sodium (30 milligrams per kilogram of body weight) by application intrapertoneally through the left lumbar region of the ventral abdominal wall.
  • Surgical defects (2) were created in directly opposed proximity by excising the peritoneal membrane and thereby exposing sidewall muscle tissue (2x1 cm).
  • the outer membrane of the cecum was removed by surgical peeling, thus exposing blood vessel loops (2xlcm). Both exposed defects were abraded to cause petechial bleeding, and then exposed to direct radiant heat source for 15 minutes to accelerate desiccation.
  • One ml of the compositions of Example 1 application temperature of 0 °C was applied to one injured site. The other injured site was left untreated.
  • the following example is directed to the determination of dissolution rates for various formulations prepared in accordance with the teachings herein.
  • Several of the formulations inco ⁇ orate at least one modifier polymer.
  • Poloxamer 407 (BASF. Mount Olive, NJ) Fractionated Poloxamer 407 (APC Lot #9630201)
  • Carbopol 940 NF (BF-Goodrich. Cleveland, OH) Hydroxypropylmethylcellulose K100M, HPMC-K100M, (Dow Chemical Company, Midland, MI)
  • Carboxymethylcellulose high viscosity, CMC (Spectrum Chemical Co., Gardena, CA)
  • Carboxymethylcellulose medium viscosity CMC-MV Purcosity
  • Polymer solutions were prepared by first dispersing the modifier polymer (i.e., CMC, hydropropylmethylcellulose (HPMC) or Carbopol) in the Tris/maleate buffer solution (0.1515 g of tris(hydroxymethyl)-aminomethane and 0.1726 g of sodium maleate were dissolved and brought up to 100 g with DI water) until fully hydrated.
  • the modifier polymer i.e., CMC, hydropropylmethylcellulose (HPMC) or Carbopol
  • Tris/maleate buffer solution 0.1515 g of tris(hydroxymethyl)-aminomethane and 0.1726 g of sodium maleate were dissolved and brought up to 100 g with DI water
  • In-vitro dissolution rate of poloxamer gels were determined using a modified USP dissolution apparatus (Hanson Research model SR6,) equipped with enhancer cells. Each of the dissolution vessels were filled with 25 mL of 0.1 M phosphate buffer (pH 7.4) (26.78 g of sodium phosphate dibasic (Na 2 HPO 4 »7H 2 0) was brought to a volume of 1L with DI water) and left to equilibrate for about 20 minutes to 36.8°C.
  • Membranes 1.2 ⁇ m cellulose ester membranes, 25 mm diameter, type RAWP
  • the determination of poloxamer concentration in the aqueous phase was carried out using the potassium picrate spectrophotometric method. This method is based on the extraction of picrate ion from water into an organic solvent in association with potassium ion complexes of polyoxyethylene chains.
  • the procedure consists of mixing 250 ⁇ L of potassium picrate solution (0.23g of picric acid (wet-based) dissolved in 10 mL of potassium hydroxide solution and brought to a volume of 50 mL with DI water) with 1 mL of 2.5 M potassium nitrate solution (50.55g of potassium nitrate was brought up to a volume of 200 mL with DI water; the pH was then adjusted to 12 with 0.1 N KOH) and 0.1 mL of the sample containing the poloxamer solution in a 16x250 mm test tube. The mixture is then vortexed and extracted with 3 mL of CH 2 C1 2 . The absorbance of the organic phase was measured at 378 run vs.
  • a reagent blank with the concentration determined from a calibration curve (Table 1) prepared by adding aliquots of surfactant standard solution (1024 ug/mL).
  • a Beckman UN/Vis spectrophotometer model DU-65 was used to measure poloxamer concentration. The pH was adjusted to 7.4 with IN HCl using a Sentron pH meter.
  • FloGel 25 refers to a 25% w/w formulation of poloxamer 407 in the Tris/maleate buffer system. Should the letter F follow the number, the poloxamer 407 has been fractionated to remove low molecular weight impurities. For the pu ⁇ oses of this example, poloxamer 407 is the constitutive polymer. Should the formulation contain a modifier polymer, it follows after a slash. Thus, FloGel 20F/0.5C, contains 20% w/w fractionated poloxamer 407 and 0.5% w/w high viscosity grade CMC. The acronyms for the modifier polymers are denoted in Table II immediately below.
  • a modifier polymer especially one of high molecular weight
  • a modifier polymer can have profound effects on poloxamer dissolution.
  • Cellulose ethers e.g. CMC and HPMC
  • the solution characteristics appear to depend on the average chain length as well as the degree of substitution. As molecular weight increases, the viscosity will increase rapidly.
  • Fractionated poloxamer 407 i.e. poloxamer 407F
  • Pluronic F-127 BASF Co ⁇ oration, Mount Olive, NJ
  • Hydrophilic co-surfactants in the form of fatty acid soaps i.e. sodium oleate, sodium laurate, sodium caprate, and sodium caprylate
  • Nu-Chek Prep. Elysian, MN
  • the buffer materials tromethamine (EM Sciences Inc., Gibbstown, NJ) and maleic acid (Sigma Chemical Co., St. Louis, MO), were used as received, and a hypoosmotic buffer containing 0.1515% w/w tromethamine and 0.1451% w/w maleic acid was prepared.
  • Final formulations contained a constant 20% w/w percentage of poloxamer 407F, and varying levels of fatty acid soaps. For phase behavior studies, samples were loaded into 5 ml Wheaton vacuoles (Fisher
  • FIG. 2 a typical phase diagram obtained for poloxamer 407F/fatty acid soap mixtures is shown in Figure 2. This diagram illustrates the effect of increasing sodium caprate concentrations on the phase behavior of 20% w/w poloxamer 407F solutions in the hypo-osmotic tromethamine/maleate buffer system.
  • the LGT is observed to increase systematically from 19 °C to 87 °C.
  • concentrations between ca. 1.5 and 2.0% caprate the LGT is in the temperature range between room and body temperature. Having a LGT in this temperature range might have some importance for the formulation of antiadhesion products, possibly improving the ease of use by obviating the need to maintain product temperature near 0 °C, and allowing the surgeon greater time to apply compositions in accordance with the methods herein.
  • the gel phase is completely suppressed.
  • the cloud point temperature is above typical steam sterilization temperatures. Being able to maintain a single phase above terminal sterilization temperatures may play a role in reducing post-sterilization syneresis.
  • the addition of fatty acid soaps represents a very efficient way of altering the LGT and cloud point of constitutive polymer gels without varying the rheological characteristics of the gel. This is, of course, in contrast to changing the LGT by varying poloxamer concentration, or the nature of the poloxamer (e.g. poloxamer 338).
  • the chainlength and degree of unsaturation of the hydrophilic co-surfactant may also be used to selectively alter the characteristics of the constitutive polymer gels. These effects are graphically illustrated in Figure 4 where the LGT is plotted as a function of soap concentration for different fatty acid soaps. The value next to the curve refers to the fatty acid portion of the soap. Thus, 8:0 represents an eight carbon fatty acid soap with no double bonds in the alkyl chain. 18:1, on the other hand, represents an alkyl chain containing eighteen carbons and a single double bond. It is apparent from Figure 4 that longer chainlength saturated soaps provide more substantial alterations of the gel characteristics than shorter chainlength analogues which appear to be less efficient at disrupting gel phase formation.
  • Ketoprofen (Sigma Chemical Co., St. Louis, MO)
  • Poloxamer 407 (BASF, Mount Olive, NJ)
  • Drug Concentration Determinations The samples were diluted to a suitable concentration with ethanol (for water insoluble drugs) or DI water. Drug concentrations were measured at the appropriate wavelength for each drug (see Table I) using a UV/Vis spectrophotometer (Beckman model DU-65). Concentrations were determined using Beer's law from the appropriate calibration curve (Table II). Gentamicin sulfate determination was performed by the UCSD Medical Center laboratory.
  • In-vitro release rate of drugs in poloxamer gels were determined using a modified USP dissolution apparatus (Hanson Research model SR6) equipped with enhancer cells. Each of the dissolution vessels was filled with 25 mL of .01 M phosphate buffer (pH 7.4) 1 and left to equilibrate for about 20 minutes to 36.8°C.
  • Membranes 1.2 ⁇ m cellulose ester membranes, 25 mm diameter, type
  • Ketoprofen Conc mg /L 22.9(Abs)-0.2 0.9996
  • Hydrocortisone Conc mg / 27.8(Abs)-0.1 0.9956 solubilization was, for most of the cases, enhanced in the gel state. Solubilization for hydrocortisone was higher in the liquid state. Two reasons could be responsible for the observed reduction in solubilization: 1) hydrocortisone has a relatively higher water solubility than the rest of the hydrophobic drugs tested: 2) samples below the gel transition temperature were equilibrated at room temperature, instead of the lower temperature used for the gels.
  • S and S 0 are the concentration of solubilized drug in the presence and absence of poloxamer, respectively.
  • C is the concentration of poloxamer (weight fraction).
  • Table V shows the apparent distribution coefficients. The higher the value of K, the greater the amount of drug that can be inco ⁇ orated into the system.
  • the modifier polymer is preferably of sufficient molecular weight that it too alters the diffusion path of the solute.
  • the fact that the size of the drug molecules did not seem to affect release rate also supports this hypothesis.

Abstract

The present invention provides a composition comprising one or more constitutive polymers and modifier polymers and/or hydrophilic co-surfactants useful for reducing adhesions or delivering bioactive agents. Methods for preventing and/or reducing post-surgical adhesions or delivering bioactive agents are also provided.

Description

METHODS AND COMPOSITIONS FOR THE DELIVERY OF PHARMACEUTICAL AGENTS AND/OR THE PREVENTION OF ADHESIONS
FIELD OF THE INVENTION The present invention generally relates to pharmaceutical preparations and methods of their use. More particularly, the present invention relates to preparations suitable for the reduction of adhesion formation in mammals or the delivery of pharmaceutically active compounds.
BACKGROUND OF THE INVENTION
Over the years, methods have been developed to achieve the efficient delivery of therapeutic or diagnostic agents to a mammal requiring such treatment. Aqueous liquids which can be applied at room temperature in a free flowing state but which forms a semi- solid gel when warmed to body temperature have been used in such capacities for some time. Such systems combine ease of application with greater retention at the site of application than the use of exclusively free flowing vehicles. For example, in U.S. Pat. No. 4,188,373, incorporated herein by reference, Pluronic® polyols are used in aqueous compositions to provide thermally gelling aqueous systems. Adjusting the concentration provides the desired sol-gel transition temperature. More particularly, the lower the concentration of the incorporated polymer the higher the sol-gel transition temperature. At a critical polymer concentration minimum, the system reaches a point where a gel will not form at any physiologically compatible temperature. While such vehicles are a substantial improvement over prior art systems, it is hard to precisely adjust the sol-gel temperature to the desired value. In U.S. Pat. Nos. 4,474,751; '752; '753; and 4,478,822, each incorporated herein by reference, drug delivery systems are described which utilize thermosetting gels. In these systems both the gel transition temperature and/or rigidity of the gel may be modified by adjustment of the pH and/or the ionic strength as well as by the concentration of the polymer. Although such vehicles may be efficiently used for the delivery of bioactive agents, establishment and maintenance of the desired sol-gel temperature and/ or persistence of the gel may be complicated by several variables including the localized physiology of the mammalian subject. Accordingly, a need still exists for pharmaceutical preparations or drug delivery compositions that allow for the establishment of a precise sol-gel transition temperature and accurate control of the gel dissolution rate in vivo.
Control of such characteristics is also desirable when similar polymeric gels are used for the prevention of adhesion formations in mammals. Adhesions are thought to form following a trauma or injury to the peritoneum. This results in increased vascular permeability, which produces an inflammatory exudate and results in the formation of a fibrin matrix. In normal wound healing, the fibrin matrix is removed by fibrinolysis, and subsequent fibroblast proliferation results in remesothelialization. However under the ischemic conditions present following surgical trauma the fibrinolytic process is suppressed and the fibrin matrix may persist. If it persists until about day three, significant collagen deposition within the fibrin matrix, can lead to adhesion formation.
As will be appreciated by those skilled in the art, prevention of adhesions has been the subject of various efforts since the beginning of this century (see, for example, Surgery, Gynecology and Obstetrics, 133:497-509, 502-503 (1971)). These efforts have included means of preventing the fibrin-coated walls of the intestine from contacting one another by distending the abdomen with oxygen or filling the abdomen with various liquids such as saline solution, paraffin, olive oil, lanolin, concentrated dextrose solution, various macromolecular solutions and silicones.
High molecular weight dextran either alone or in combination with dextrose has also been used (Holtz, et al, Fertility and Sterility, 33:660-662 (1980); 34:394-395 (1980)). One such formulation, HYSKON® (Pharmacia, Piscataway, N.J.), which comprises 32% aqueous solution of dextran 70 containing 10% dextrose, was effective in reducing peritoneal adhesions subsequent to surgery. However, it has been reported that HYSKON has a tendency to support bacterial proliferation. Further concern has been expressed over the anaphylactoid potential of dextran (DiZerega et al., Fertility and Sterility, 40:612-619 (1983)). In addition, the benefit of dextran 70 in preventing post-operative adhesions was shown to be limited to the more dependent regions of the pelvis.
The use of resorbable fibrous barriers to separate injured tissues has also been described (Linsky, J. Reprod. Med., 17-20 (1987)). For example, TC-7 (Johnson and Johnson Products, Inc., New Brunswick, N.J.), an oxidized cellulose fabric barrier, has been used as a treatment to prevent organ adhesion to the peritoneum. Other solid sheet devices include polytetrafluoroethylene (Gore-Tex®, W. L. Gore) and crosslinked hyaluronic acid (SeprafilmΘGenzyme Corp.).
Chondroitin sulfate and sodium carboxymethyl cellulose have also been used to prevent the formation of postoperative adhesions in the rabbit uterus (Oelsner et al., J Reprod. Med. 32:812-814 (1987)). Chondroitin sulfate solutions have also been proposed for intraperitoneal use in the prevention of adhesions in rabbits
More recently, aqueous gel compositions comprising polyalkylene polymers have been shown to successfully reduce adhesions (U.S. Pat. No. 5,366,735, incorporated herein by reference). These compositions can be applied below room temperature as a liquid and form semi-solid gels when warmed to body temperature. However, as with the aforementioned drug delivery compositions, precise control of the sol-gel transition temperature and dissolution rate of the gel within the physiological environment still present problems in many cases. Accordingly, despite these previous efforts, a need exists for improved means to treating and/or preventing post-surgical adhesions. As such, it is an object of the present invention to provide polymeric gel compositions which allow for precise control of the sol-gel transition temperature and/or dissolution rate of the gel once formed.
It is a further objective of the present invention to provide gelling drug delivery preparations, and methods for their use, comprising at least one bioactive agent and exhibiting desired sol-gel transition temperatures and/or gel dissolution rates.
It is yet another objective of the present invention to provide gelling compositions, and methods of their use in preventing or reducing adhesions, which exhibit desired sol-gel transition temperatures and/or gel dissolution rates.
SUMMARY OF THE INVENTION
The present invention accomplishes these and other objectives by providing polymeric compositions that exhibit well defined sol-gel transition temperatures (or defined ranges of temperatures) and/or established dissolution rates. In one embodiment, the disclosed compositions generally comprise at least one constitutive polymer and at least one modifier polymer that may be used to modify or control the dissolution rate of gel once it has been formed. In another preferred embodiment, the compositions of the present invention comprise at least one constitutive polymer and at least one hydrophilic co-surfactant whereby the gelation temperature (or sol-gel transition temperature) of the composition may be controlled or modified. Of course, it will be appreciated that the compositions may comprise at least one constitutive polymer in combination with both at least one modifier polymer and at least one hydrophilic co-surfactant to provide preparations having both selected gelation temperatures and superior dissolution times. Yet other preferred embodiments of the invention will comprise the aforementioned preparations and at least one bioactive agent. In any event, the polyphase preparations of the present invention may be used to retard or prevent the formation of scar tissue or adhesions in a mammal, for the prolonged delivery of a bioactive agent or both. Accordingly, in selected embodiments the present invention comprises methods for the reduction of adhesion or scar tissue formation comprising the administration of the disclosed preparations to a mammal in need thereof. Yet other selected embodiments comprise methods of delivering a bioactive agent to a mammal comprising administering the disclosed polyphase compositions incorporating a pharmaceutically effective amount of at least one bioactive agent to a mammal in need thereof. With respect to each of the aforementioned embodiments the compositions of the present invention will be administered as a relatively free flowing liquid that gels upon contact with the mammalian tissue to provide a viscoelastic semi-solid barrier or mask that may remain in place for an extended period.
In preferred embodiments of the instant invention the constitutive polymer will be a polyoxyalkylene copolymer. More particularly, in selected embodiments the constitutive polymer will be selected from the group consisting of polyoxyalkylene block copolymers, polyoxyalkylene polyethers and combinations thereof. In especially preferred embodiments of the invention, the constitutive polymer will comprise Poloxamer 407. The constitutive polymer or polymers may be present at any concentration that provides the desired gel viscosity and/or viscoelastic properties. Preferably, the constitutive polymers are present in a concentration which, when combined with the other components of the preparation, allows for the administration of the composition as a relatively free flowing liquid which gels upon contact with mammalian tissue.
Besides the constitutive polymer discussed above, selected embodiments of the invention will comprise at least one modifier polymer that may be used to modify the dissolution rate of the composition. Essentially, modifier polymers compatible with the present invention comprise any polymeric entity capable of slowing or retarding the dissolution rate of the constitutive polymer once it has gelled. That is, the modifier polymers of the present invention comprise any polymer that, when added to the constitutive polymer(s), provides for a slower dissolving or diffusing gel when compared with a gel formed from pure constitutive polymer(s) under equivalent conditions. Preferred modifier polymers typically have a relatively high average molecular weight on the order of tens or hundreds of thousands. While a large number of polymeric compounds are suitable for use as modifier polymers, particularly preferred compounds comprise cellulose ethers (carboxymethyl cellulose) and Carbopols (e.g. Carbopol 940-NF). The absolute incorporated concentration of the modifier polymers in the compositions of the present invention is not critical and may be adjusted to provide the desired dissolution rates and/or retention times.
In addition to the aforementioned elements, selected embodiments of the compositions disclosed herein may further comprise one or more hydrophilic co-surfactants which may be used to modify the gelation temperature (sol-gel transition temperature) of the resulting preparation. More particularly, selected hydrophilic co-surfactant(s) may be added to compositions comprising a constitutive copolymer(s) or compositions comprising constitutive copolymer(s) and modifier polymer(s) to alter or modify the gelation temperature of the resulting composition when compared to similar compositions not comprising the hydrophilic co-surfactant. In especially preferred embodiments the hydrophilic may be added at an effective concentration to lower the gelation temperature of the composition so as to provide for more rapid and complete gelation upon contact with the relatively high temperature mammalian tissue. While a number of compounds may be used as hydrophilic co-surfactants in accordance with the teachings herein, particularly preferred embodiments of the present invention incorporate fatty acid soaps such as sodium laureate, sodium caprate or sodium caprylate. Of course it will be appreciated that combinations of hydrophilic co- surfactants may be incorporated in the compositions of the present invention to provide the desired transition temperature or transition temperature range.
As previously alluded to the preparations of the present invention may further include one or more selected bioactive agents. More specifically, pharmaceutically effective amounts of both hydrophilic and lipophilic bioactive agents may be advantageously delivered using the preparations of the present invention. Thus, in accordance with the aforementioned embodiments, bioactive agents compatible with the present invention include, but are not limited to, antibiotics, antivirals, mydriatics, antiglaucomas, anti-inflammatories, -mtihistaminics, antineoplastics, anesthetics, ophthalmic agents, enzymes, cardiovascular agents, polynucleotides, genetic material, viral vectors, immunoactive agents, imaging agents, immunosuppressive agents, peptides, proteins, physiological gases, gastrointestinal agents and combinations thereof. Particularly preferred compositions may comprise one or more humectants, bactericides, bacteriostatic agents, fibrinolytic agents or agents effective in preventing leukocyte migration into the area of surgical injury. Pharmaceutically effective amounts of the selected bioactive agents may be determined using techniques well known in the art. It will further be appreciated that the bioactive agents may be incorporated in the form of relatively insoluble solid particulates or associated with insoluble polymeric particulates. It will be appreciated that, in accordance with the teachings herein, the preparations of the present invention, with or without an incorporated bioactive agent, may be administered to a patient using a route of administration selected from the group consisting of topical, subcutaneous, pulmonary, synovial, intramuscular, intraperitoneal, nasal, vaginal, rectal, aural, oral and ocular routes. The administered composition preferably gels upon contact with the relatively warm mammalian tissue and may act as a depot for the prolonged delivery of one or more incorporated bioactive agents. In other embodiments the gelled compositions may act as a barrier or film which prevents or retards the formation of adhesion or scar tissue. Since the present invention provides particularly effective methods for the prevention of post-surgical and other adhesion formation, administration of pharmaceutically effective amounts to the peritoneal, pelvic or pleural cavity is especially preferred. As such adhesions are often associated with injury to mammalian organs, those skilled in the art will appreciate that the compositions are particularly useful when applied to the selected area during or immediately following surgery.
Besides the components mentioned above, the compositions of the present invention may further comprise pharmaceutically acceptable stabilizers, preservatives and buffers, preferably in an amount sufficient to maintain the pH of the composition at about pH 7.4±0.2.
Other objects, features and advantages of the present invention will be apparent to those skilled in the art from a consideration of the following detailed description of preferred exemplary embodiments thereof taken in conjunction with the associated Figures which will first be described briefly. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a graphical representation of an exemplary phase diagram for a prior art composition comprising a constitutive polymer;
Figure 2 illustrates an equilibrium phase diagram of 20% w/w poloxamer 407 (407F) in tromethamine/maleate buffer with added sodium caprate with arrows indicating that the cloud point temperature is greater than the highest temperature measured, i.e. 140 °C;
Figure 3 is a graphical representation of a gelation profile of 1%, 3% and 5% (w/w) sodium caprylate in 20% w/w poloxamer 407 (407F) in hypotonic tromethamine/maleic acid buffer; Figure 4 is a graphical representation illustrating the effect of fatty acid soap concentration on the lower gelation temperature (LGT) of 20% w/w poloxamer 407F solutions for soaps of varying alkyl chain lengths and degrees of saturation.
DETAILED DESCRIPTION OF THE INVENTION Compositions and methods are disclosed herein for delivering bioactive agents and/or reducing post-surgical adhesion formation reformation in mammals following injury to the organs or tissues, particularly those of the peritoneal, pelvic or pleural cavity. The compositions of the invention are also useful in reducing adhesion formation/reformation in other body spaces such as the subdural, extraocular, intraocular, otic, synovial, tendon sheath, or those body spaces created either surgically or accidentally. In selected embodiments of the invention, the concentration of the constitutive polymer in the disclosed compositions may be adjusted to take advantage of the gelation properties of certain polyoxyalkylene polymers. For instance, at certain concentrations aqueous solutions of said polymers form clear gels at mammalian body temperatures but are liquids at ambient temperatures or below. Of course, it is a major advantage of the present invention that selected hydrophilic co-surfactants may also be used to modify the gelation temperature of the disclosed compositions. This advantageously allows the selected compositions of the present invention to be administered as a relatively free flowing liquid which gels or thickens at the body temperature of the mammalian subject. However, it should be appreciated that compositions may be formed in accordance with the teachings herein that do not gel or thicken following application to the selected area or tissue. Preferably, the osmolality and pH of the compositions are adjusted to match the pH and osmotic pressure of mammalian bodily fluids, i.e. approximately pH 7.4. Subsequent to deposition of the compositions of the invention in the peritoneal, pelvic, or pleural cavity of a mammal, or other body spaces the constitutive polymer (i.e., a polyoxyalkylene block copolymer) is eventually excreted in a non-metabolized form, mainly through the kidney.
The present compositions may also be used as a distending medium during diagnostic or operative endoscopic procedures, such as, for example, for intrauterine procedures. In addition to the anti-adhesive properties, since certain aqueous concentrations of the preferred polyoxyalkylene block copolymers form a clear gel, their use is well suited for visualization of interior cavities.
In a further advantage, the disclosed formulations provide a barrier between tissues for hours or days. Because they are applied as liquids, they are easier to use, particularly for laparoscopic surgical procedures. That is, the compositions of the instant invention may be administered though a relatively small incision using a cannula or catheter assembly. Those skilled in the art will appreciate that the disclosed compositions of the present invention are preferably aqueous based preparations. Thus, the compositions typically comprise water in an amount of from about 60% to about 90%, by weight, preferably, about 70% to about 85%, by weight, and most preferably, about 75% to about 82% by weight, based upon the total weight of the composition. As used herein, the terms "peritoneal" and "abdominal" cavity are used as synonyms, as are the terms "pleural" and "thoracic" cavity.
As used herein, the term "polyalkylene block polymers" include those polymers which form clear gels at mammalian body temperatures but are liquids at ambient temperatures or below. As used herein, the term "gel" is defined as a solid or semisolid colloid containing a certain quantity of water. The colloidal solution with water is often called a "hydrosol".
A. Constitutive Polymers:
As set forth above, he present invention comprises at least one constitutive polymer dispersed in a aqueous medium. In preferred embodiments of the instant invention the constitutive polymer will be a polyoxyalkylene polymer. More particularly, in selected embodiments the constitutive polymer will be selected from the group consisting of polyoxyalkylene block copolymers, polyoxyalkylene polyethers and combinations thereof. In especially preferred embodiments of the invention, the constitutive polymer will comprise poloxamer 407. The constitutive polymer or polymers may be present at any concentration that provides the desired gel viscosity and/or viscoelastic properties. Preferably, the constitutive polymers are present in a concentration which, when combined with the other components of the preparation, allows for the administration of the composition as a relatively free flowing liquid which gels upon contact with mammalian tissue.
Thus, according to a preferred embodiment, the compositions comprise one or more polyoxyalkylene block copolymers of the formula
Y[(A)n-E-H]x (I)
wherein A is a polyoxyalkylene moiety; x is at least 2; Y is derived from water or an organic compound containing x reactive hydrogen atoms;
E is a polyoxyethylene moiety; n has a value such that the average molecular weight of A is at least about 500; and the total average molecular weight of the copolymer is at least about 5000. Preferably, the polyoxyalkylene moiety A has an oxygen/carbon atom ratio of less than 0.5. According to one embodiment of the invention, A is derived from an alkylene oxide selected from the group consisting of butylene oxide, propylene oxide or a mixture thereof. Preferably, A is a polyoxypropylene moiety, and preferably has an average molecular weight of from about 3,000 to about 4,000 g mol"1. The polyoxyethylene moiety E preferably constitutes from about 60 to about 85% by weight of the copolymer, more preferably at least about 70%.
In one embodiment, Y is derived from a water soluble organic compound having 1 to about 6 carbon atoms. In another embodiment, Y is derived from an organic compound selected from the group consisting of propylene glycol, glycerin, pentaerythritol trimethylolpropane, ethylenediamine and mixtures thereof.
According to one embodiment, the copolymer has the formula: HO(C2H4O)£(C4H8O)fl(C2H4O)6H (H)
wherein a and b are integers such that (C4H8O)α has a molecular weight of at least about 500. Useful polyoxyalkylene block copolymers which will form gels in aqueous solutions can be prepared using a hydrophobe base (such as A in Formulas (I) and (II)) derived from propylene oxide, butylene oxide or mixtures thereof. These block copolymers and representative methods of preparation are further generally described in U.S. Pat. Nos. 2,677,700; 2,674,619; and U.S. Pat. No. 2,979,528, incoφorated herein by reference.
Generally, the polyoxybutylene-based block copolymers useful in the compositions of the invention are prepared by first condensing 1,2 butylene oxide with a water soluble organic compound initiator containing 1 to about 6 carbon atoms such as 1,4 butylene glycol or propylene glycol and at least 2 reactive hydrogen atoms to prepare a polyoxyalkylene polymer hydrophobe of at least about 500, preferably at least about 1000, most preferably at least about 1500 average molecular weight. Subsequently, the hydrophobe is capped with an ethylene oxide residue. Specific methods for preparing these compounds are described in U.S. Pat. No. 2,828,345 and British Patent No. 722,746, both of which are herein incorporated by reference.
In a further preferred embodiment, the compositions comprise polyoxyethylene- polyoxypropylene block copolymers of the formula (III):
HO(C2H4O)b(C3H6O)a(C2H4O)bH (III)
wherein a is an integer such that the hydrophobe base represented by (C3H6O)a has a molecular weight of at least about 900, preferably at least about 2500, most preferably at least about 4000 average molecular weight, as determined by hydroxyl number. In a particularly preferred embodiment, the compositions comprise a polyxyethylene-polyoxyproplyene block copolymer of formula (III), having a polyoxyproplyene hydrophobe base average molecular weight of about 4000, a total average molecular weight of about 12,000 and containing oxyethylene groups in the amount of about 70% by weight of the total weight of the copolymer. This copolymer is sold under the trademark PLURONIC® F-127 (also known as poloxamer 407)(BASF Corp, Parsippany, N.J.). More specifically, poloxamer 407 is a tri-block copolymer containing two polyoxyethylene blocks flanking a central polyoxypropylene block. The USP material has an average molecular formula of (EO)101-(PO)56-(EO)101, and average molecular weight of ca. 12,000 g mol"1. When placed in an aqueous solution in accordance with the present invention, poloxamer 407 self-assembles so as to remove contact between the polyoxypropylene groups and water (i.e. self-assembly is driven by the hydrophobic effect). The self-assembled units are termed micelles. The structure of the micelles and the interactions between them is strongly dependent on temperature. Interestingly, a large increase in solution viscosity (i.e. gel-phase formation) is noted with increasing temperature. Gel phase formation occurs as a result of organization of the micelles into a three- dimensional cubic array.
In another embodiment, the copolymer has the formula:
(R)2N-(CH2)2-N(R)2 (IN)
wherein R is H(OC2H4)b(OC3H6)a-; and a and b are integers such that the hydrophobe base represented by (C3H6O)a has a sum average molecular weight of at least about 2000, about 3 to about 5%. The hydrophobe base is prepared by adding propylene oxide for reaction at the site of the four reactive hydrogen atoms on the amine groups of ethylenediamine. An ethylene oxide residue is used to cap the hydrophobe base.
In all permutations of copolymers of formula (I), it is preferred that the polyoxyethylene chain constitute from about 60 to about 85% by weight of the colpolymer, preferably at least about 10%. It is further preferred that the copolymer have a total average molecular weight of at least about 5000, preferably from about 9,000 to about 15,000 as specified in the USP).
The procedure used to prepare aqueous solutions which form gels of the polyoxyalkylene block copolymers is well known. Either a hot or cold process for forming the solutions can be used. A cold technique involves the steps of dissolving the polyoxyalkylene block copolymer at a temperature of about 5° to about 10° C in water. When solution is complete the system is brought to room temperature whereupon it forms a gel. If the hot process of forming the gel is used the polymer is added to water heated to a temperature of about 75 °C to about 85 °C, with slow stirring until a clear homogeneous solution is obtained. Upon cooling, a clear gel is formed. Block copolymer gels containing polyoxybutylene hydrophobes must be prepared by the above hot process, since these will not liquefy at low temperatures.
The organic compound initiator which is utilized in the preparation of the polyoxyalkylene block copolymers generally is water or an organic compound, and can contain a plurality of reactive hydrogen atoms. Preferably, Y in formulas (I) and (II) above is defined as derived from a water soluble organic compound having 1 to about 6 carbon atoms and containing x reactive hydrogen atoms where x has a value generally, of at least 1, preferably, a value of at least 2. Falling within the scope of the compounds from which Y is derived from water soluble organic compounds having at least two reactive hydrogen atoms are water soluble organic compounds such as propylene glycol, glycerin, pentaerythritol, trimethylolpropane, ethylenediamine, and mixtures thereof and the like.
The oxypropylene chains can optionally contain small amounts of at least one of oxy ethylene or oxy butylene groups. Oxy ethylene chains can optionally contain small amounts of at least one of oxypropylene or oxybutylene groups. Oxy butylene chains can optionally contain small amounts of at least one of oxy ethylene or oxypropylene groups. The physical form of the polyoxyalkylene block copolymers can be a viscous liquid, a paste or a solid granular material depending upon the molecular weight of the polymer.
In addition to those polyoxyalkylene polymers described above, the present compositions may comprise other polyoxyalkylene polymers which form gels at low concentrations in water. Examples of such polymers are described in U.S. Pat. No. 4,810,503, incorporated herein by reference. These polymers are prepared by capping conventional polyoxyalkylene polyether polyols with an alphaolefin epoxide having an average of about 20 to about 45 carbon atoms, or mixtures thereof. Aqueous solutions of these polymers gel in combination with surfactants, which can be ionic or nonionic. The combination of the capped polyether polymers and the surfactants provide aqueous gels at low concentrations of the capped polymer and surfactant which generally do not exceed 10% by weight total.
Conventional copolymer polyether polyols are prepared by preparing block or heteric intermediate polymers of ethylene oxide and at least one lower alkylene oxide having 3 to 4 carbon atoms as intermediates. These are then capped with the alpha-olefin epoxide. Ethylene oxide homopolymers capped with the alpha-olefin oxides are also useful as intermediates.
The heteric copolymer intermediate is prepared by mixing ethylene oxide and at least one lower alkylene oxide having 3 to 4 carbon atoms with a low molecular weight active hydrogen-containing compound initiator having at least two active hydrogens and preferably,
2 to 6 active hydrogen atoms such as a polyhydric alcohol, containing from 2 to 10 carbon atoms and from 2 to 6 hydroxyl groups, heating said mixture to a temperature in the range of about 50° C to 150° C, preferably, from 80° C to 130°, under an inert gas pressure, preferably, from about 30 psig to 90 psig. A block copolymer intermediate is prepared by reacting either the ethylene oxide or the alkylene oxide having 3 to 4 carbon atoms with the active hydrogen-containing compound followed by reaction with the other alkylene oxide.
The ethylene oxide and the alkylene oxides having from 3 to 4 carbon atoms are used in the intermediates in amounts so that the resulting polyether product will contain at least 10 percent by weight, preferably about 70 percent to about 90 percent by weight, ethylene oxide residue. The ethylene oxide homopolymer intermediate is prepared by reacting ethylene oxide with the active hydrogen-containing compound. The reaction conditions for preparing the block copolymer and ethylene oxide homopolymer intermediates are similar to those for the heteric copolymer intermediate. The temperature and pressure are maintained in the above ranges for a period of about one hour to ten hours, preferably one to three hours.
The alpha-olefin oxides which are utilized to modify the conventional polyether intermediates are those oxides, and commercially available mixtures thereof, generally containing an average of about 20 to 45, preferably about 20 to 30, carbon atoms. The amount of alpha-olefin required to obtain the more efficient capped polyethers is generally about 0.3 to 10 percent, preferably about 4 to 8 percent, of the total weight of the polyethers. Further description regarding the preparation of heteric and block copolymers of alkylene oxides and ethylene oxide homopolymers is described in the art (U.S. Pat. Nos.
3,829,506, 3,535,307; 3,036,118; 2,979,578; 2,677,700; and 2,675,619, incorporated herein by reference.) Whatever constitutive polymer is selected the absolute concentration present in the compositions of the present invention is determined by the gelation characteristics desired.
One major advantage of the present invention is that the desired gelation temperatures and viscosity of the resulting gels may be adjusted through the addition of modifier polymers and hydrophilic co-surfactants. This allows the use of lower concentrations of constitutive polymer without markedly reducing the ultimate gel characteristics of the composition. However, for the purposes of the present invention, exemplary concentrations of constitutive polymer may range from approximately 2% w/w to 50% w/w and more preferably from 4% to 30% w/w and even more preferably from 16% to 28% w/w.
B. Modifier Polymers:
As set forth above any biocompatible polymeric entity that modifies the dissolution time of the gel resulting from the administration of the compositions of the present invention may be used in accordance with the teachings herein. In general terms, preferred modifier polymers to alter the dissolution time should preferably have the following characteristics: (a) high molecular weight; (b) effective swelling in water but poor dissolution: (c) compatibility with the constitutive polymer and, in particular, poloxamers; and (d) stability to extremes in heat and pH. Those skilled in the art will appreciate that the phrase "alter the dissolution time" is held to mean the alteration of the gel dissolution time in vitro or in vivo with respect to a gel comprising constitutive polymer without the modifier polymer under similar conditions. It will further be appreciated that the alteration of dissolution times or release rates of the constitutive polymer from the gel matrix may be used to optimize formulations for antiadhesion applications as well as for other applications including controlled drug delivery.
Without wishing to be bound by any one particular theory, it is believed that release (and subsequent gel dissolution) is a function of several physicochemical characteristics within the gel, and can be modified by the addition of high molecular weight polymers such as sodium carboxymethyl cellulose, polyacrylates (i.e. Carbopols) or other polyester based polymers. It appears that the dissolution rate is modified by the formation of a strong polymeric matrix (i.e. the modifier polymeric matrix) that controls the release of the constitutive polymer via diffusion through the formed modifier polymer interstices. One possible reason for this effect may be that the constitutive polymer has to diffuse around the long linear molecules of the incorporated modifier polymer. In general, this effect appears to be most pronounced when the selected modifier polymer(s) have a molecular weight greater than or equal to approximately 500,000 although modifier polymers of much lower molecular weight (i.e. on the order of 50,000). In particularly preferred embodiments the selected modifier polymers will combine a relatively high molecular weight with a biodegradable moiety in their structure to speed excretion. High molecular weight polylactic-glycolide copolymers which are broken down by hydrolytic decomposition are one example of such a polymer. It should be emphasized that these modifier polymers may also be used to slow the dissolution (and hence prolong delivery time) of any incorporated bioactive agent.
While any polymeric entity possessing the appropriate characteristics may be incorporated in the compositions of the present invention, exemplary polymers compatible with the teachings herein include, but are not limited to: poly(acrylic acid), poly(styrene sulfonate), carboxymethylcellulose, poly(vinyl alcohol), poly(ethylene oxide), poly(vinylpyrrolidone), shellac, cellulose acetate phthalate, cellulose acetate succinate, polyvinyl acetate phthalate, hydroxypropylmethylcellulose acetate, poly(methacrylic acid-co- methylmethacrylate), poly(methyl acrylate), poly(methyl methacrylate), poly(glutamic acid), poly(lactic acid), poly(lactic-glycolide), poly(glycolic acid), poly(ε-caprolactone), poly(β- hydroxybutyric acid), poly(β-hydroxy valeric acid), polydioxanone, poly(ethylene terephthalate), poly (malic acid), poly(tartronic acid), poly(ortho esters), polyanhydrides, polycyanoacrylate, poly(phosphoesters), polyphosphazenes, poly(lysine), polysaccharides, chitosan, polyelectrolytes, gelatin, gum arabic, poly(amino acids), agar, furcelleran, alginate, carageenan, starch, pectin, celluloses, exudate gums, tragacanth, karaya, ghatti seed gums, guar gum, locust bean gum, xanthan, pullulan, scleroglucan, curdlan, dextran, gellan, chitin, chondroitin sulfate, water soluble collagen, dermantan sulfate, heparin, keratan sulfate, hyaluronic acid and combinations thereof. It will further be appreciated that any pharmaceutically acceptable salt of the foregoing compounds may be used in the disclosed compositions with compromising the effectiveness thereof. As will be seen in the Examples below the modifier polymers of the present invention may be used in suprisingly low concentrations to provide extended dissolution times or release times. In this regard, the selected modifier polymers are preferably incoφorated in a range between about 0.05% and about 25% by weight and more preferably in a range of from approximately 0.5% to approximately 5% by weight. Of course the absolute amount of modifier polymer included in the composition will depend on factors such as the constitutive polymer selected, the molecular weight of the modifier polymer and the physiochemical properties of the various composition components. These determinations are well within the purview of the skilled artisan and may easily be determined without undue experimentation.
C. Hydrophilic Co-Surfactants: Yet another aspect of the present invention comprises the addition of a hydrophilic co- surfactant to the disclosed compositions (i.e. constitutive polymer preparations and constitutive polymer + modifier polymer preparations) to alter the physiochemical properties thereof. That is, the incoφoration of a hydrophilic co-surfactant in accordance with the teachings herein may provide several advantages over prior art formulations. These advantages are most easily understood in conjunction with a graphical representation of a polyphase system of the instant invention and examples set forth below.
Accordingly, turning to Fig. 1 a phase diagram for a constitutive polymer solution (poloxamer 407) is shown. The lower gelation temperature (LGT) refers to the temperature at which the poloxamer micelles (sol phase) self-assemble into the cubic array (i.e. the gel phase). At temperatures above the upper gelation temperature (UGT) the micelles change their shape from spheres to prolates, thereby negating their ability to assemble in a cubic packing. This leads to the reformation of the low viscosity sol phase. Above another critical temperature, termed the cloud point (CP), the micelles separate into their own coacervate phase in excess water. The solution clouds due to mixing of the two insoluble phases. The lower gelation temperature (LGT) of the constitutive polymer solutions in water is largely dependent upon the total constitutive polymer concentration, such that increases in concentration lead to decreases in the LGT. Fractionation of the constitutive polymer (fractionated using organic phase separation or other means known in the art, such as described, for example, in Textbook of Polymer Science. F. Billmeyer, Wiley-Interscience, pp. 45-56 (1971)) and the addition of high viscosity carboxymethylcellulose (CMC) does little to alter the LGT. FloGel 28 (28% w/w poloxamer 407) has an LGT of 13 °C, and is currently applied surgically at a temperature of 0°C. Therefore, application of the product will have to be done in a timely fashion to avoid gelation in the application catheter. Additionally, it has been hypothesized that increases in the LGT to a temperature close to or above room temperature may be advantageous.
As previously discussed, the equilibrium phase behavior of solutions comprising a constitutive polymer can be dramatically altered by the addition of hydrophilic co-surfactants. The changes in phase behavior are typically manifested by significant increases in the lower gelation temperature and cloud point temperature. While any hydrophilic co-surfactant may be used to modify the equilibrium phase behavior of the disclosed compositions in accordance with the teachings herein, hydrophilic co-surfactants comprising fatty acid soaps are particularly compatible with the present invention. In this regard long chain, saturated soaps appear to be particularly efficient at altering the phase behavior to provide the desired composition characteristics. Significantly, the rheological properties of the gelled compositions of the present invention are unaltered by the presence of the fatty acid soaps, indicating that, as long as the critical packing volume of the cubic phase is exceeded, the rheology will remain virtually unchanged. Thus, in accordance with the instant invention, the addition of hydrophilic co-surfactants to the disclosed polyphase systems provides an efficient method for modifying the gelation temperature and cloud point temperature. These changes in phase behavior are particularly advantageous for a drug delivery vehicle or antiadhesion product as they allow for storage and application at temperatures near room temperature. Moreover, these characteristics reduce the potential for significant syneresis during terminal sterilization.
Accordingly, preferred embodiments of the present invention may comprise effective amounts of at least one hydrophilic co-surfactant. In particularly preferred embodiments the incoφorated hydrophilic co-surfactant will comprise a fatty acid soap. Those skilled in the art will appreciate that fatty acid soaps are GRAS (generally regarded as safe) materials, present naturally in the human body, and included in many pharmaceutical products including large volume parenterals (e.g. Fluosol®). Their toxicological profile is well understood and, at the concentrations compatible with the present invention, they pose no toxicological risk. While several compounds comprising fatty acids are useful in the present invention, especially compatible fatty acid soaps comprise sodium oleate, sodium laurate, sodium caprate, sodium caprylate and combinations thereof.
In any event, as illustrated by the Examples below, the hydrophilic co-surfactants of the present invention may be incoφorated in relatively low concentrations to provide the desired gelation properties. It will be appreciated that the selected hydrophilic co-surfactant or surfactants may comprise any concentration that provides for the preferred gelation temperatures. However, exemplary concentrations of hydrophilic co-surfactants compatible with the instant invention are typically in a range between about 0.05% and about 25% by weight and more preferably in a range of from approximately 0.5% to approximately 5% by weight.
D. Bioactive Agents: In addition to the antiadhesion characteristics of the compositions of the present invention the preparations also provide for the efficient delivery of bioactive agents. Along with the prolonged deposition time supplied by the disclosed compositions, they may increase the solubilization and bioavailability of incoφorated pharmaceutical compounds. More particularly, the micelle core of the gelled compositions of the present invention may serve as a reservoir for solubilizing nonpolar solutes such as hydrophobic drugs. Interestingly the micelles may also self-assemble to form stiff gels above a critical temperature. As previously discussed, gel formation appears to occur when the micelles behave as hard spheres in a close-packed simple cubic array. This thermal gelation property provides interesting formulation alternatives for pharmaceutical applications, whereby the poloxamer micelles are applied in the fluid sol state and allowed to gel in place on tissue surfaces. Thus, in addition to their ability to act as a barrier to prevent surgical adhesions, poloxamer gels may also be an ideal drug delivery vehicle, owing to their low toxicity and ability to impede drug diffusion.
Accordingly, the compositions disclosed herein may further optionally comprise one or more pharmaceutically acceptable adjuvants such as a humectant, a bactericide, a bacteriostatic agent, an antihistamine, or a decongestant, an agent to prevent leucocyte migration into the area of surgical injury, or a fibrinolytic agent. Useful humectants include, but are not limited to, glycerin, propylene glycol and sorbitol. Useful bactericides include, by way of example, antibacterial substances such as β-lactam antibiotics, such as cefoxitin, n- formamidoyl thienamycin and other thienamycin derivatives, tetracyclines, chloramphenicol, neomycin, gramicidin, bacitracin, sulfonamides; aminoglycoside antibiotics such as gentamycin, kanamycin, amikacin, sisomicin and tobramycin; nalidixic acids and analogs such as norfloxacin and the antimicrobial combination of fluoroalanine/pentizidone; nitrofurazones, and the like. Antihistamines and decongestants such as pyrilamine, chloφheniramine, tetrahydrozoline, antazoline, and the like, can also be used in admixtures as well as anti-inflammatories such as cortisone, hydrocortisone, beta-methasone, dexamethasone, fluocortolone, prednisolone, triamcinolone, indomethacin, sulindac, its salts and its corresponding sulfide, and the like. Both steroidal and nonsteroidal compounds are particularly compatible with the compositions and methods of the present invention. With regard to the latter, ketoprofen, indomethacin and tolmetin sodium are particularly preferred. Nitric oxide donors such as nononates and nitrosylated compounds may also be incoφorated. Useful leucocyte migration preventing agents which can be used in admixtures include but are not limited to silver sulfadiazine, acetylsalicylic acid, indomethacin and Nafazatrom. Useful fibrinolytic agents include urokinase, streptokinase, tissue plasminogen activator (TPA) and acylated plasmin.
In a more general sense, compatible bioactive agents comprise both hydrophilic and lipophilic compounds including antibiotics, antivirals, mydriatics, antiglaucomas, anti- inflammatories, antihistaminics, antineoplastics, anesthetics, ophthalmic agents including anti- glaucomics, enzymes, cardiovascular agents, polynucleotides, genetic material, viral vectors, immunoactive agents, imaging agents, immunosuppressive agents, peptides, proteins, physiological gases, gastrointestinal agents and combinations thereof.
Because the preparations of the present invention are uniquely suited for various administrative techniques such as ocular, oral, pulmonary, rectal, synovial, subcutaneous, intramuscular, intraperitoneal, nasal, vaginal, or aural administration of medicaments or diagnostic compounds, they are compatible for use with a wide variety of bioactive agents. For example, ophthalmic applications involving topical administration of the disclosed preparations are particularly preferred. Accordingly, the foregoing list of compounds is exemplary only and not intended to be limiting. It will also be appreciated by those skilled in the art that the proper amount of bioactive agent and the timing of the dosages may be determined for the formulations in accordance with already-existing information and without undue experimentation.
Preferably, the compositions are applied to surgically injured tissue as an aqueous solution which upon contact with living mammalian tissue forms a firm, adherent gel. Where the composition is a viscous liquid or paste, these compositions can be applied without dilution to areas of surgical injury in the abdominal or thoracic cavities. The formulations adhere to the site of tissue injury and reduce or prevent the formation of postsurgical adhesions during the healing process.
In addition to the aforementioned applications, the preparations of the invention may also be used to deliver therapeutic and diagnostic agents to the gastrointestinal tract by, for example, the oral or direct routes of administration. A contemplated example would be the delivery of antibiotics to the lining of the gastrointestinal tract in the treatment of Heliobacter pylori infections. H. pylori has been implicated in the cause of gastric ulcers and stomach cancer. Antibiotics effective in the treatment of H pylori infections could be administered in the form of a free flowing liquid that gels and adheres to the sites of infection.
It will be appreciated that the compositions of the present invention may further contain preservatives, cosolvents, suspending agents, viscosity enhancing agents, ionic- strength and osmolality adjustors and other excipients in addition to buffering agents. Suitable water soluble preservatives which may be employed are sodium bisulfite, sodium thiosulfate, ascorbate, benzalkonium chloride, chlorabutanol, thimerosal, phenylmercuric borate, parabens, benzylalcohol phenylethanol and others. These agents may be present, generally, in amounts of about 0.001% to about 5% by weight and, preferably, in the amount of about 0.01 to about 2% by weight.
Suitable buffering agents or salts useful in maintaining pΗ include alkali or alkaline earth metal carbonates, chlorides, sulfates, phosphates, bicarbonates, citrates, borates, acetates and succinates such as sodium phosphate, citrate, borate, acetate, bicarbonate, carbonate and tromethamine (TRIS). Preferably, these agents are present in amounts sufficient to maintain the pΗ of the system at 7.4+ 0.2 and preferably, 7.4. As such, the buffering agent can be as much as 5% by weight.
It will also be appreciated by those skilled in the art that the preparations of the present invention may be sterilized, for example, by heat, irradiation, ultrafiltration or combinations of any of these or equivalent techniques. Specifically, the preparations of the invention may be sterilized, for example, by autoclaving at 121 °C for 15 minutes or by filtration through a 0.22 mm filter.
The high bioavailability bioactive preparations of the present invention may advantageously be supplied to the physician in a sterile prepackaged form. More particularly, the formulations may be supplied as stable, preformed preparations, ready for administration or as separate, ready to mix components. When supplied as components the final preparation of the polyphase material could easily be performed in the pharmacy just prior to administration.
The following examples illustrate the various aspects of the invention but are not intended to limit its scope. Where not otherwise specified throughout this specification and claims, temperatures are given in degrees centigrade, and parts, percentages, and proportions are by weight. EXAMPLE 1 Synthesis of Compositions Comprising Modifier Polymers
Compositions comprising a constitutive polymer (poloxamer 407) and a modifier polymer (sodium carboxymethylcellulose) were prepared by dissolving the poloxamer in distilled water (4°C) to give a concentration of 28% by weight in accordance with the cold process described above for forming aqueous solutions.
Formulation 1 : FloGel 28B (Control)
Ingredients Source Lot %w/w grams
Poloxamer 407, NF, Prill BASF WPDP-586B 28.0000
280.00
Tromethamine (TRIS), USP Spectrum ID 289 0.1091
1.09
Maleic Acid Spectrum IK 051 0.1045
1.05
Sodium Hydroxide Pellets, USP Spectrum IG 043 0.0420
0.42
Sterile Water for Irrigation, USP Baxter G876094 71.7444
717.44
Total
1000
Formulation 2 : FloGel 25B/0.5
Ingredients Source Lot %w/w grams
Poloxamer 407, NF, Prill BASF WPDP-586B 25.0000 250.00 Sodium Carboxymethylcellulose Spectrum JA 156 0.5000
5.00
Tromethamine (TRIS), USP Spectrum ID 289 0.1091
1.09
Maleic Acid Spectrum IK 051 0.1045
1.05
Sodium Hydroxide Pellets, USP Spectrum IG 043 0.0420
0.42
Sterile Water for Irrigation, USP Baxter G876094 74.2444
742.44
Total
1000
Formulation 3 : FloGel 20F/0.8
Ingredients Source Lot %w/w grams
Poloxamer 407, Fractionated MDN 1145-107 20.0000
200.00
Sodium Carboxymethylcellulose Spectrum JA 156 0.8000
8.00
Tromethamine (TRIS), USP Spectrum ID 289 0.1091
1.09
Maleic Acid Spectrum IK 051 0.1045 1.05
Sodium Hydroxide Pellets, USP Spectrum IG 043 0.0420
0.42
Sterile Water for Irrigation, USP Baxter G876094 78.9444
789.44
Total
1000 Formulation 4 : FloGel 16B/1.5
Ingredients Source Lot _%w/w grams
Poloxamer 407, NF, Prill BASF WPDP-586B 16.0000
160.00 Sodium Carboxymethylcellulose Spectrum JA 156 1.5000
15.00 Tromethamine (TRIS), USP Spectrum ID 289 0.1091
1.09 Maleic Acid Spectrum IK 051 0.1045
1.05 Sodium Hydroxide Pellets, USP Spectrum IG 043 0.0420
0.42 Sterile Water for Irrigation, USP Baxter G876094 82.2444
822.44
Total
1000
EXAMPLE 2 Anti-adhesion Characteristics of Compositions Comprising Modifier Polymers
The following test procedure was utilized to determine the effect of the formulations of Example 1 on surgically injured rats. Female Sprague-Dawley rats having a 300-400 gram body weight were anesthetized with pentobarbital sodium (30 milligrams per kilogram of body weight) by application intrapertoneally through the left lumbar region of the ventral abdominal wall. Surgical defects (2) were created in directly opposed proximity by excising the peritoneal membrane and thereby exposing sidewall muscle tissue (2x1 cm). The outer membrane of the cecum was removed by surgical peeling, thus exposing blood vessel loops (2xlcm). Both exposed defects were abraded to cause petechial bleeding, and then exposed to direct radiant heat source for 15 minutes to accelerate desiccation. One ml of the compositions of Example 1 (application temperature of 0 °C) was applied to one injured site. The other injured site was left untreated.
Results of this experiment indicate that a formulation containing only poloxamer 407 (Formulation 1) reduced adhesions by approximately 50%, while formulations containing poloxamer 407 and carboxymethylcellulose (Formulations 2-4) reduced adhesions by 70 to 99%. The increased efficacy of formulations containing both polymers may be due to a reduced rate of erosion in vivo analogous to that observed in vitro. All of the formulations exhibited maximal efficacy when applied to the injured tissue at approximately 0 °C.
EXAMPLE 3 Dissolution Rates of Compositions Comprising Modifier Polymers
The following example is directed to the determination of dissolution rates for various formulations prepared in accordance with the teachings herein. Several of the formulations incoφorate at least one modifier polymer.
Materials:
Chemicals utilized in the study and their sources are listed below, All chemicals were used without further purification.
Sodium phosphate dibasic, Na2HPO4, '7^0 (Sigma Chemical Co.. St. Louis, MO)
Maleic acid sodium salt (Sigma Chemical Co., St. Louis, MO)
1 N hydrochloric acid solution (Fisher Scientific, Fair Lawn, NJ)
Potassium nitrate (Fisher Scientific. Fair Lawn. NJ) 0.1 N potassium hydroxide solution (Fisher Scientific, Fair Lawn, NJ)
Methylene Chloride stabilized with amylene (Fisher Scientific. Fair Lawn, NJ) Picric Acid with 35% water (Aldrich Chemical Co., St. Louis. MO)
Tris(hydroxymethyl)-aminomethane (EM Science, Wakefield, RI)
Poloxamer 407 (BASF. Mount Olive, NJ) Fractionated Poloxamer 407 (APC Lot #9630201)
Carbopol 940 NF (BF-Goodrich. Cleveland, OH) Hydroxypropylmethylcellulose K100M, HPMC-K100M, (Dow Chemical Company, Midland, MI)
Carboxymethylcellulose high viscosity, CMC (Spectrum Chemical Co., Gardena, CA) Carboxymethylcellulose medium viscosity CMC-MV (Penta Manufacturing Co., Livingston, NJ)
Preparation. Polymer solutions were prepared by first dispersing the modifier polymer (i.e., CMC, hydropropylmethylcellulose (HPMC) or Carbopol) in the Tris/maleate buffer solution (0.1515 g of tris(hydroxymethyl)-aminomethane and 0.1726 g of sodium maleate were dissolved and brought up to 100 g with DI water) until fully hydrated. Poloxamer 407 (the constitutive polymer) was then added to the sample in an ice bath (T = 3- 5°C), and mixed until the poloxamer dissolved. The sample was kept under refrigeration until usage.
In-vitro dissolution rate of poloxamer gels. The in-vitro dissolution rates of poloxamer-based gels were determined using a modified USP dissolution apparatus (Hanson Research model SR6,) equipped with enhancer cells. Each of the dissolution vessels were filled with 25 mL of 0.1 M phosphate buffer (pH 7.4) (26.78 g of sodium phosphate dibasic (Na2HPO4»7H20) was brought to a volume of 1L with DI water) and left to equilibrate for about 20 minutes to 36.8°C. Membranes (1.2 μm cellulose ester membranes, 25 mm diameter, type RAWP) were presoaked in phosphate buffer and placed on the cell.
Approximately 0.6 mL of sample in the fluid sol phase was then loaded into each of the enhancer cells. The cells were subsequently closed and the fluid sol phase allowed to gel at room temperature prior to introduction into the dissolution vessels. The dissolution paddles were rotated at a speed of 100 φm (approximating the hydrodynamic stress found in the peritoneal cavity) and were adjusted to remain at approximately 1 cm from the top of the cells throughout all experiments. The in-vitro release of poloxamer from the gels was monitored over a period of 4 hr, with 1 mL samples collected every 0.5 hr. The vessel was replaced with fresh buffer each time a sample aliquot was removed. Each sample was run in triplicate. The average standard error of the measurements was of 0.013. Quantitation of poloxamer. The determination of poloxamer concentration in the aqueous phase was carried out using the potassium picrate spectrophotometric method. This method is based on the extraction of picrate ion from water into an organic solvent in association with potassium ion complexes of polyoxyethylene chains. The procedure consists of mixing 250 μL of potassium picrate solution (0.23g of picric acid (wet-based) dissolved in 10 mL of potassium hydroxide solution and brought to a volume of 50 mL with DI water) with 1 mL of 2.5 M potassium nitrate solution (50.55g of potassium nitrate was brought up to a volume of 200 mL with DI water; the pH was then adjusted to 12 with 0.1 N KOH) and 0.1 mL of the sample containing the poloxamer solution in a 16x250 mm test tube. The mixture is then vortexed and extracted with 3 mL of CH2C12. The absorbance of the organic phase was measured at 378 run vs. a reagent blank, with the concentration determined from a calibration curve (Table 1) prepared by adding aliquots of surfactant standard solution (1024 ug/mL). A Beckman UN/Vis spectrophotometer model DU-65 was used to measure poloxamer concentration. The pH was adjusted to 7.4 with IN HCl using a Sentron pH meter.
Table I
Poloxamer 407 Standard Solutions
Std ID Poloxamer 407 (μg) Absorbance (378 nm)
1 64 0.245
2 128 0.497
3 256 0.982
Release Rates of Test Formulations: The results obtained for various poloxamer- based formulations, including those comprising modifier polymers are detailed in Table II. The nomenclature of the various compositions is detailed below. FloGel 25 refers to a 25% w/w formulation of poloxamer 407 in the Tris/maleate buffer system. Should the letter F follow the number, the poloxamer 407 has been fractionated to remove low molecular weight impurities. For the puφoses of this example, poloxamer 407 is the constitutive polymer. Should the formulation contain a modifier polymer, it follows after a slash. Thus, FloGel 20F/0.5C, contains 20% w/w fractionated poloxamer 407 and 0.5% w/w high viscosity grade CMC. The acronyms for the modifier polymers are denoted in Table II immediately below.
Table II In-Nitro Release Profiles of FloGels: Sample k (hr"n) n MDT (hr)
Flogel 25 0.15 0.66 0.011 7
Flogel 28 0.15 0.66 0.0026 7
Flogel 28a 0.97 1.0 0.009 1
Flogel 28F 0.22 0.5 0.099 7
Flogel 25/0.5 Ca 0.78 0.8 -0.041 1
Flogel 0.22 0.5 -0.040 7
25/0.5CMN
Flogel 25/1 CMV 0.23 0.5 -0.032 6
Flogel 25/0.5 0.17 0.5 -0.023 11
HPMC
Flogel 25/0.5C 0.14 0.5 -0.0064 17
Flogel 25/0.5 0.12 0.5 -0.028 22
940
Flogel 20F/0.5C 0.10 0.5 0.001 33
Flogel 20F/0.8C 0.074 0.5 0.0053 60
Flogel 16/1.5C 0.044 0.6 0.018 67
Flogel 14F/1C 0.037 0.6 0.019 90
a No membrane used in dissolution study
F Fractionated Poloxamer 407 940 Carbopol 940-NF
C High Viscosity CMC
CMV Medium Viscosity CMC
HPMC Hydroxypropylmethylcellulose
Discussion: The data reported in Table II are fits to the Korsmeyer-Peppas equation
V viz.
Q = k - + b (V)
where Q is the amount released at the time t, Qα is the overall released amount, k is a release rate constant of the n* order, n is a dimensionless number related to the dissolution mechanism and b is the y axis intercept, characterizing the initial burst effect. A value of n=0.5 characterizes a release mechanism controlled by polymer diffusion, while a value of n=1.0 characterizes an erosion controlled mechanism. Erosion and diffusion control the process in equal parts if n=0.66. Since the release rate constant k has the dimension hr"n, values for different mechanisms cannot be compared directly. To overcome this problem it is possible to define another quantity termed the mean dissolution time (MDT). The MDT is the sum of the different periods of time the poloxamer molecules stay in the matrix before release, divided by the total number of molecules, and is calculated according to equation VI: MDT=n_k"1 n (VI) n
Poloxamer 407 gels, in the absence of a membrane in the dissolution apparatus, exhibit erosion controlled kinetics (n=1.0) with an MDT of 1 hr. Placement of the cellulose ester membrane introduces a diffisional barrier to the release, and is characterized by equal contributions of erosion and diffusion control (n=0.66), with an MDT of 7 hr. Changes in gel viscosity (i.e. comparison of FloGel 25 vs. FloGel 28) and poloxamer fractionation do not appreciably alter the dissolution mechanism or the MDT.
The addition of a modifier polymer, especially one of high molecular weight, can have profound effects on poloxamer dissolution. Cellulose ethers (e.g. CMC and HPMC) are long chain polymers. The solution characteristics appear to depend on the average chain length as well as the degree of substitution. As molecular weight increases, the viscosity will increase rapidly.
Addition of 0.5% w/w of the high viscosity CMC to a 25% w/w poloxamer 407 solution dramatically increases the MDT to 17 hr. It also changes the mechanism of release to one of pure diffusion control (i.e. n=0.50). Alternatively, the medium viscosity grade of CMC does not appear to have a dramatic effect on the MDT at the concentrations of modifier polymer employed. Other high molecular weight polymers (e.g. Carbopol 940-NF) also alter the dissolution mechanism and dramatically increase the MDT. In short it was suprisingly found that increases in the MDT by nearly an order of magnitude can be achieved by the addition of a modifier polymer. It is believed that the magnitude of the dissolution times measured in this in-vitro test are indicative of release rates found in-vivo in the peritoneal cavity.
EXAMPLE 4
Modification of the Gelation Temperature of Poloxamer Preparations
Through the Incorporation of Hydrophilic Co-Surfactants
In order to demonstrate the advantages associated with the addition of a hydrophilic co-surfactant to polymeric compositions in accordance with the present invention, several different preparations were formulated. Methods: Fractionated poloxamer 407 (i.e. poloxamer 407F) was prepared from NF grade Pluronic F-127 (BASF Coφoration, Mount Olive, NJ) as described herein. Hydrophilic co-surfactants in the form of fatty acid soaps (i.e. sodium oleate, sodium laurate, sodium caprate, and sodium caprylate) were obtained from Nu-Chek Prep. (Elysian, MN). The buffer materials, tromethamine (EM Sciences Inc., Gibbstown, NJ) and maleic acid (Sigma Chemical Co., St. Louis, MO), were used as received, and a hypoosmotic buffer containing 0.1515% w/w tromethamine and 0.1451% w/w maleic acid was prepared. Final formulations contained a constant 20% w/w percentage of poloxamer 407F, and varying levels of fatty acid soaps. For phase behavior studies, samples were loaded into 5 ml Wheaton vacuoles (Fisher
Scientific, Pittsburgh, PA), and flame-sealed. The vacuoles were then immersed in a constant temperature bath. For temperatures less than 60 °C, phase behavior was determined in a water bath (Koehler, Bohemia, NY). At higher temperatures an oil bath (Haake, model DC3, Germany) was utilized. Temperature was raised in two degree increments from ca. 1 °C to 120 °C. Samples were allowed to equilibrate for at least 1 hour at constant temperature prior to examination. Since the cubic liquid crystalline phase is isotropic (i.e. not birefringent), the determination of the gel boundary is somewhat subjective. Once equilibrium was reached, the vials were simply inverted and gravity was allowed to determine if the sample was in the sol or gel state. Rheological studies were performed on a Rheometric Scientific Inc. (Piscataway,
NJ.) model SR 5000 constant stress rheometer. A 25 mm parallel plate geometry with a gap of 1.0 mm was employed. In dynamic temperature ramp studies, a sinusoidal stress (ω = 1 s"1) was applied at a stress less than the yield stress of the material (ca. 1-10 Pa). This ensured that the sample was in the linear viscoelastic region. Temperature was ramped at a rate of 2 °C min"1. Rapid temperature equilibration was ensured with a peltier/water bath system. Samples were loaded at 0-5 °C (i.e. in the sol phase), and allowed to gel on the plate. This was done to avoid applying an unknown shear history to the sample. Plots of the complex viscosity (η*) vs. temperature were recorded.
Discussion: Details regarding the equilibrium phase behavior of 20% poloxamer 407F solutions with added fatty acid soaps are shown in Table III below. As may be seen from the data, fatty acid soaps have a substantial effect on the phase behavior of the poloxamer 407F solutions. The LGT for a 20% poloxamer 407F solution in the absence of added fatty acid soaps is 19 °C. With added fatty acid soaps, the LGT can be increased to temperatures as high as 87 °C (observed with 4.86% added sodium caprate). In addition, the cloud point temperature, which is 108 °C for the 20% poloxamer 407F solution, may be easily increased to temperatures above 140 °C, i.e. significantly above the typical temperatures "used during terminal steam sterilization.
Table III
Phase behavior of 20% poloxamer 407F solutions in hypoosmotic tromethamine/maleate buffer with added fatty acid soaps.
Figure imgf000033_0001
In accordance with the results reported above a typical phase diagram obtained for poloxamer 407F/fatty acid soap mixtures is shown in Figure 2. This diagram illustrates the effect of increasing sodium caprate concentrations on the phase behavior of 20% w/w poloxamer 407F solutions in the hypo-osmotic tromethamine/maleate buffer system.
Above a sodium caprate concentration of ca. 1% w/w, the LGT is observed to increase systematically from 19 °C to 87 °C. At concentrations between ca. 1.5 and 2.0% caprate, the LGT is in the temperature range between room and body temperature. Having a LGT in this temperature range might have some importance for the formulation of antiadhesion products, possibly improving the ease of use by obviating the need to maintain product temperature near 0 °C, and allowing the surgeon greater time to apply compositions in accordance with the methods herein. Above ca. 5% caprate, the gel phase is completely suppressed. Also of note in Figure 2 is the fact that above ca. 2% sodium caparate, the cloud point temperature is above typical steam sterilization temperatures. Being able to maintain a single phase above terminal sterilization temperatures may play a role in reducing post-sterilization syneresis.
Dynamic temperature ramp studies for formulations with varying levels of added sodium caprylate are shown in Figure 3. At low temperatures (below gel phase formation), the dynamic rheological method is not an efficient method for measuring low viscosities. This leads to a significant degree of noise in the data. Once the sol-gel phase transition is encountered, a shaφ increase by ca. 4 orders of magnitude in the complex viscosity is noted. Above the phase transition, the dynamic rheological method is very sensitive and little noise is apparent in the data. Interestingly, the complex viscosity of the gel phase remains virtually constant as the LGT is varied by the addition of the hydrophilic co-surfactants. This is consistent with the model that the gel phase formation is due to the formation of a cubic array of micelles above their critical packing volume fraction. Accordingly, as long as the critical volume fraction is exceeded, the rheological properties of the gel do not appear to be significantly altered. Thus, the addition of fatty acid soaps represents a very efficient way of altering the LGT and cloud point of constitutive polymer gels without varying the rheological characteristics of the gel. This is, of course, in contrast to changing the LGT by varying poloxamer concentration, or the nature of the poloxamer (e.g. poloxamer 338).
The chainlength and degree of unsaturation of the hydrophilic co-surfactant may also be used to selectively alter the characteristics of the constitutive polymer gels. These effects are graphically illustrated in Figure 4 where the LGT is plotted as a function of soap concentration for different fatty acid soaps. The value next to the curve refers to the fatty acid portion of the soap. Thus, 8:0 represents an eight carbon fatty acid soap with no double bonds in the alkyl chain. 18:1, on the other hand, represents an alkyl chain containing eighteen carbons and a single double bond. It is apparent from Figure 4 that longer chainlength saturated soaps provide more substantial alterations of the gel characteristics than shorter chainlength analogues which appear to be less efficient at disrupting gel phase formation. Thus, while 2.5% of 12:0 soap is required to melt the 20% poloxamer 407F gel, nearly 11.5% of the 8:0 soap is used to provide the same characteristics. Unsaturated soaps also appear to be less active than their saturated analogues at increasing both the LGT and cloud point of poloxamer 407 gels. In any case, with a 20% w/w concentration of poloxamer 407F, it is possible to achieve gelation between room and body temperature, and a cloud point greater than 121 °C for the caprate (10:0) and caprylate (8:0) soaps. Moreover, as the total poloxamer concentration is increased, higher concentrations of hydrophilic co-surfactant may be used to achieve the same degree of shift in the LGT. Thus, the laurate soap (12:0) may be preferred under these conditions.
EXAMPLE 5
Drug Solubility and in-vitro Drug Release Rates in Poloxamer 407-Based Thermoreversible Gels
In order to demonstrate the advantages of the present invention with respect to drug delivery, selected compounds were incoφorated in various preparations formed in conjunction with the present invention.
Materials The chemicals utilized in this study and their sources are listed below. All chemicals were used without further purification.
Ketoprofen (Sigma Chemical Co., St. Louis, MO)
Prednisone (Sigma Chemical Co., St. Louis, MO)
Indomethacin (Sigma Chemical Co., St. Louis, MO) Tolmetin Sodium (Sigma Chemical Co., St. Louis, MO)
Hydrocortisone (Sigma Chemical Co., St. Louis, MO)
Poloxamer 407 (BASF, Mount Olive, NJ)
Ethyl Alcohol 200 proof (Spectrum Chemical Co.. Gardena, CA)
FloGel 28 (MDV Technologies Inc., Dearborn MI) FloGel 25B/0.5C (MDV Technologies Inc., Dearborn MI)
FloGel 25 B/ 1 C (Alliance Pharmaceutical lot # 587-29b).
FloGel 25 (Alliance Pharmaceutical lot #587-29a).
FloGel 28F (Alliance Pharmaceutical lot #534-73).
Gentamicin Sulfate (Amresco, Solon, OH) Methods
Preparation of Drug solutions in Poloxamer 407 -water systems. Poloxamer 407 was added to deionized (DI) water in an ice bath (T = 3-5°C), and mixed until the poloxamer dissolved. Excess amounts of drug were then added to the aqueous poloxamer solutions and allowed to equilibrate overnight (T = 3-5°C). The next day the sample was warmed momentarily to 40° C to hasten solubilization. This process was repeated two or three times to ensure saturation. Samples containing less than 20% poloxamer 407 were then equilibrated overnight at room temperature while the other samples were equilibrated at 5°. Prior to analysis, the samples were filtered through a 0.2μm nylon filter syringe to remove unsolubilized drug. Samples were assayed for solubilized drug concentrations by absorbance spectroscopy (see below).
Preparation of drug solutions in FloGel. Two mg of drug was added to 1 mL of the FloGel material and equilibrated overnight (T = 3-5°C). As before, the sample was warned two to three times to 40°to hasten solubilization. Samples were stored at 5°C until use.
Drug Concentration Determinations: The samples were diluted to a suitable concentration with ethanol (for water insoluble drugs) or DI water. Drug concentrations were measured at the appropriate wavelength for each drug (see Table I) using a UV/Vis spectrophotometer (Beckman model DU-65). Concentrations were determined using Beer's law from the appropriate calibration curve (Table II). Gentamicin sulfate determination was performed by the UCSD Medical Center laboratory.
In-vitro release rate of drugs in poloxamer gels. The in-vitro release rates of drugs in poloxamer-based gels were determined using a modified USP dissolution apparatus (Hanson Research model SR6) equipped with enhancer cells. Each of the dissolution vessels was filled with 25 mL of .01 M phosphate buffer (pH 7.4)1 and left to equilibrate for about 20 minutes to 36.8°C. Membranes (1.2 μm cellulose ester membranes, 25 mm diameter, type
1 26.78 g of sodium phosphate dibasic (Na-HP04 7H20) was brought to a volume of 1 L with DI water. The pH was adjusted to a pH of 7.4 with IN HCl using a Sentron pH meter. RAWP) were presoaked in phosphate buffer and placed on the cell. Approximately 0.6 mL of sample in the fluid sol phase was then loaded into each of the enhancer cells. The cells were subsequently closed and the gel phase was allowed to form at room temperature prior to introduction into the dissolution vessels. The dissolution paddles were rotated at a speed of 100 φm (approximating the hydrodynamic stress found at the peritoneal cavity) and were positioned approximately 1 cm from the top of the cells for all experiments. The in-vitro release of drug from the gels was monitored over a period of 4 hr. One mL samples were collected every 0.5 hr. The vessel was refilled with fresh buffer every time a sample aliquot was removed, and each sample was run in duplicate. For additional details regarding the dissolution apparatus the reader is referred to: (Dellamary L:In-vitro dissolution rates of poloxamer-based thermoreversible gels. Research & Development Technical Report No. EPR-32-97-4).
Table IV
Calibration Curve Equations for the Different Drugs Selected
Drug Calibration Curve Equation R.2
Ketoprofen Concmg/L=22.9(Abs)-0.2 0.9996
Prednisone Concmg/ =33.3(Abs)-0.06 0.9999
Indomethacin Concmg/L=232.4(Abs)-2 0.991
Tolmetin Sodium Concmg/ =22.9(Abs)-0.7 0.9919
Hydrocortisone Concmg/ =27.8(Abs)-0.1 0.9956 solubilization was, for most of the cases, enhanced in the gel state. Solubilization for hydrocortisone was higher in the liquid state. Two reasons could be responsible for the observed reduction in solubilization: 1) hydrocortisone has a relatively higher water solubility than the rest of the hydrophobic drugs tested: 2) samples below the gel transition temperature were equilibrated at room temperature, instead of the lower temperature used for the gels.
Apparent distribution coefficients (Km) of the hydrophobic drugs between a micellar phase and an aqueous phase were determined according to equation VII.
S=KmC+l (VII) S0
Where S and S0 are the concentration of solubilized drug in the presence and absence of poloxamer, respectively. C is the concentration of poloxamer (weight fraction). Table V, shows the apparent distribution coefficients. The higher the value of K, the greater the amount of drug that can be incoφorated into the system.
Table V
Distribution Coefficients of Hydrophobic Drugs Between a Micellar Phase and an Aqueous Phase
Drug Evaluation Distribution Coeffϊecient (log Km)
Liquid State | Gel State
Hydrocortisone 1 0.55 Prednisone 1.1 1.5 Ketoprofen 2.9 3.1 Indomethacin 3 3.4
Positive distribution coefficients (log K,-,) indicate that the drug preferably partitions into the micelle rather than into the water phase. Sodium tolmetin, in contrast, would rather partition into the water phase. It is not possible to calculate a partition coefficient for tolmetin using equation VII. The decreasing solubility with increasing poloxamer concentration gives a negative slope and an undefined value of log K,-.. It is clear that tolmetin partitions almost entirely into the bulk aqueous phase and very little is solubilized in the poloxamer micelle. The above results confirm that hydrophobic drugs are actually solubilizing into the core of poloxamer 407 micelles.
There were no appreciable differences in diffusion coefficients or mean dissolution times (MDT) between different drugs within the same poloxamer formulations. Thus, no differences were observed between sodium tolmetin and ketoprofen despite the fact that the tolmetin is simply dissolved in the continuous aqueous phase of the gel while the ketoprofen is solubilized in the micelle core. These results imply that the micelles present little impediment to diffusion (i.e. there is no diffusional resistance). Significant reductions in the rate of diffusion are observed, however, when a modifier polymer (i.e. carboxymethylcellulose, CMC) is added to the poloxamer. The slow diffusion observed in poloxamer gets results (at least in part) from the longer diffusion path that a drug must take in order to pass around the micelles. In the case of polymer mixtures, the modifier polymer is preferably of sufficient molecular weight that it too alters the diffusion path of the solute. The fact that the size of the drug molecules did not seem to affect release rate also supports this hypothesis. While this invention has been described with reference to certain specific embodiments, it will be recognized by those skilled in the art that many variations are possible without departing from the scope and spirit of the invention, and it will be understood that it is intended to cover all changes and modifications of the invention, disclosed herein for the puφoses of illustration, which do not constitute departures from the spirit and scope of the invention.

Claims

CLAIMSWe claim:
1. A composition for reducing post-surgical adhesion, said composition comprising one or more constitutive polymers and one or more modifier polymers.
2. A composition of claim 1 wherein the constitutive polymer is a polyoxyalkylene copolymer.
3. A composition of claim 2 wherein the polyoxyalkylene copolymer is selected from polyoxyalkylene block copolymers, polyoxyalkylene polyethers and combinations thereof.
4. A composition of claim 3 wherein the polyoxyalkylene block polymer is of the formula
Y[(A)n-E-H]x (I)
wherein A is a polyoxyalkylene moiety; x is at least 2;
Y is derived from water or an organic compound containing x reactive hydrogen atoms;
E is a polyoxyethylene moiety; n has a value such that the average molecular weight of A is at least about 500; and the total average molecular weight of the copolymer is at least about 5000.
5. The composition of claim 4 wherein said polyoxyalkylene moiety is derived from an alkylene oxide selected from the group consisting of butylene oxide, propylene oxide and mixtures thereof.
6. The composition of claim 4 wherein said copolymer has the formula: HO(C2H4O)6(C3H6O)╬▒(C2H4O)╬┤H (IV)
wherein a and b are integers such that (C3H6O)╬▒ has a molecular weight of at least about 900.
7. The composition of claim 6 wherein a is 101 and b is 56.
8. The composition of claim 1 wherein the constitutive polymer is present in an amount of about 2 to about 50 percent by weight of the total weight of said composition.
9. The composition of claim 8 wherein said copolymer is present in an amount of about 16 to about 28 percent by weight.
10. The composition of claim 1 wherein the modifier polymer is selected from cellulose ethers and Carbopols.
11. The composition of claim 10 wherein the cellulose ether is carboxymethlycellulose or a pharmaceutically acceptable salt thereof.
12. The composition of claim 1 comprising from about 0.05 to about 25 percent by weight of the modifier polymer.
13. The composition of claim 12 comprising from about 0.5 to about 5 percent by weight of the modified polymer.
14. A composition of claim 1 further comprising one or more hydrophobic co-surfactants.
15. A composition of claim 14 wherein the co-surfactant is a fatty acid soap.
16. A composition of claim 15 wherein said fatty acid soap is sodium oleate, sodium laureate, sodium caprate or sodium caprylate.
17. The composition of claim 14 comprising from about 0.05 to about 25 percent by weight of hydrophobic co-surfactant.
18. The composition of claim 17 comprising from about 0.5 to about 5 percent by weight of hydrophobic co-surfactant.
19. A composition for reducing post-surgical adhesion, said composition comprising one or more constitutive polymers and one or hydrophilic co-surfactants.
20. A composition of claim 19 wherein the constitutive polymer is a polyoxyalkylene copolymer.
21. A composition of claim 20 wherein the polyoxyalkylene copolymer is selected from polyoxyalkylene block copolymers, polyoxyalkylene polyethers and combinations thereof.
22. A composition of claim 21 wherein the polyoxyalkylene block polymer is of the formula
Y[(A)n-E-H]x (I)
wherein A is a polyoxyalkylene moiety; x is at least 2;
Y is derived from water or an organic compound containing x reactive hydrogen atoms; E is a polyoxyethylene moiety; n has a value such that the average molecular weight of A is at least about 500; and the total average molecular weight of the copolymer is at least about 5000.
23. The composition of claim 22 wherein said polyoxyalkylene moiety is derived from an alkylene oxide selected from the group consisting of butylene oxide, propylene oxide, and mixtures thereof.
24. The composition of claim 23, wherein said copolymer has the formula:
HO(C2H4O)£(C3H6O)Ω(C2H4O)&H (III)
wherein a and b are integers such that (C3H6O)fl has a molecular weight of at least about 900.
25. The composition of claim 24 wherein a is 56 and b is 101.
26. The composition of claim 19 wherein the constitutive polymer is present in an amount of about 2 to about 50 percent by weight of the total weight of said composition.
27. The composition of claim 26 wherein said copolymer is present in an amount of about 16 to about 28 percent by weight.
28. A composition of claim 19 wherein the co-surfactant is a fatty acid soap.
29. A composition of claim 28 wherein said fatty acid soap is sodium oleate, sodium laureate, sodium caprate or sodium caprylate.
30. The composition of claim 1 or 19 further comprising one or more bioactive agents.
31. The composition of claim 30 wherein the bioactive agents is a humectant, bactericide, bacteriostatic agent or fibrinolytic agent.
32. The composition of claim 31 wherein the biactive agent is a humectant present in the amount of about 0.5 to about 10 percent by weight and is selected from the group consisting of propylene glycol, glycerin, sorbitol and mixtures thereof.
33. The composition of claim 1 or 19 further comprising a pharmaceutically acceptable buffer.
34. The composition of claim 33 wherein the amount of said buffer is sufficient to maintain the pH of said composition at about pH 7.4.
35. A method for reducing post-surgical adhesion comprising administering an effective amount of the composition of claim 1 or 19.
36. The method of claim 35 wherein said adhesion is associated with injury to mammalian organs.
37. The method of claim 36 wherein said organs are located in the peritoneal, pelvic or pleural cavity.
PCT/US1997/023865 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions WO1999032151A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP97951781A EP1039932A1 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
US09/582,508 US6280745B1 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
CA002316248A CA2316248A1 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
AU55335/98A AU753206B2 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
JP2000525141A JP2001526246A (en) 1997-12-23 1997-12-23 Methods and compositions for drug delivery and / or adhesion prevention
PCT/US1997/023865 WO1999032151A1 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
AU19438/99A AU1943899A (en) 1997-12-23 1998-12-23 Methods for delivering pharmaceutical agents to mucosal surfaces
PCT/US1998/027410 WO1999032152A2 (en) 1997-12-23 1998-12-23 Methods for delivering pharmaceutical agents to mucosal surfaces

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US1997/023865 WO1999032151A1 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09/582,508 A-371-Of-International US6280745B1 (en) 1996-02-09 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
US09/930,760 Continuation US20020048602A1 (en) 1996-02-09 2001-08-15 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions

Publications (1)

Publication Number Publication Date
WO1999032151A1 true WO1999032151A1 (en) 1999-07-01

Family

ID=22262364

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1997/023865 WO1999032151A1 (en) 1997-12-23 1997-12-23 Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
PCT/US1998/027410 WO1999032152A2 (en) 1997-12-23 1998-12-23 Methods for delivering pharmaceutical agents to mucosal surfaces

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US1998/027410 WO1999032152A2 (en) 1997-12-23 1998-12-23 Methods for delivering pharmaceutical agents to mucosal surfaces

Country Status (5)

Country Link
EP (1) EP1039932A1 (en)
JP (1) JP2001526246A (en)
AU (2) AU753206B2 (en)
CA (1) CA2316248A1 (en)
WO (2) WO1999032151A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002070021A1 (en) * 2001-03-05 2002-09-12 Teva Pharmaceutical Industries Ltd. Gastric delivery of antineoplastic agents and methods of treatment via inhibition of cancer cell using them
EP1414497A1 (en) * 2001-08-08 2004-05-06 University Of Florida Novel microemulsion and micelle systems for solubilizing drugs
US6881420B2 (en) 2000-06-23 2005-04-19 Teva Pharmaceutical Industries Ltd. Compositions and dosage forms for gastric delivery of irinotecan and methods of treatment that use it to inhibit cancer cell proliferation
WO2005079856A1 (en) * 2004-02-23 2005-09-01 The University Of British Columbia Drug delivery compositions comprising hydrophobic polymers and amphipathic molecules
US7073512B2 (en) * 1997-05-07 2006-07-11 Saturnus Ag Adhesion prevention and an endoscopic insufflation system therefor
US7875677B2 (en) 2001-04-20 2011-01-25 The University Of British Columbia Micellar drug delivery systems for hydrophobic drugs
US8546363B2 (en) 2008-05-14 2013-10-01 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US8575122B2 (en) 2008-07-21 2013-11-05 Otonomy, Inc. Controlled release auris sensory cell modulator compositions and methods for the treatment of otic disorders
US8784870B2 (en) 2008-07-21 2014-07-22 Otonomy, Inc. Controlled release compositions for modulating free-radical induced damage and methods of use thereof
US8802116B2 (en) 2003-02-27 2014-08-12 Novasel Australia Pty. Ltd. Poloxamer emulsion preparations
US8852626B2 (en) 2008-06-27 2014-10-07 Otonomy, Inc. Controlled-release CNS modulating compositions and methods for the treatment of otic disorders
WO2015175899A1 (en) * 2014-05-16 2015-11-19 Ultrast, Inc. Phase-shifting formulations
US9205048B2 (en) 2008-07-21 2015-12-08 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
US9486405B2 (en) 2013-08-27 2016-11-08 Otonomy, Inc. Methods for the treatment of pediatric otic disorders
US11040004B2 (en) 2016-09-16 2021-06-22 Otonomy, Inc. Otic gel formulations for treating otitis externa

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7138133B2 (en) 2001-10-10 2006-11-21 The Procter & Gamble Company Orally administered liquid compositions
JP2005534430A (en) * 2002-08-06 2005-11-17 ジェンベック, インコーポレイテッド Improved injection system
US7223826B2 (en) 2003-01-30 2007-05-29 3M Innovative Properties Company Amide-functional polymers, compositions, and methods
US20040151691A1 (en) 2003-01-30 2004-08-05 Oxman Joel D. Hardenable thermally responsive compositions
JP4473539B2 (en) * 2003-08-12 2010-06-02 エスエス製薬株式会社 Pharmaceutical composition
US20090196844A1 (en) 2006-02-01 2009-08-06 Samyang Corporation Composition for inhibiting adhesion
US8062282B2 (en) 2006-02-13 2011-11-22 Fossa Medical, Inc. Methods and apparatus for temporarily occluding body openings
CA2731766C (en) * 2008-07-21 2014-01-28 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
EP3508197A1 (en) * 2009-10-21 2019-07-10 Otonomy, Inc. Modulation of gel temperature of poloxamer-containing formulations
JP2013508381A (en) * 2009-10-21 2013-03-07 オトノミ―,インク. Control of the gelation temperature of formulations containing poloxamers
JP6143286B2 (en) * 2013-05-13 2017-06-07 学校法人 関西大学 Anti-adhesion material and method for producing the same
WO2016013700A1 (en) 2014-07-24 2016-01-28 (주)메디톡스 Temperature sensitive adhesion prevention composition and use thereof
JP6403297B2 (en) * 2017-05-02 2018-10-10 川澄化学工業株式会社 Method for producing anti-adhesion material

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4810503A (en) * 1987-03-31 1989-03-07 Basf Corporation Polymers which form gels at low concentrations in water
WO1990004971A1 (en) * 1988-11-03 1990-05-17 M.D.R. Group, Inc. Improved viscoelastic fluid for use in surgery and other therapies and method of using same
WO1991019481A1 (en) * 1990-06-15 1991-12-26 Allergan, Inc. Reversible gelation compositions and methods of use
EP0470251A1 (en) * 1989-04-25 1992-02-12 The Green Cross Corporation Liquid prostaglandin composition
US5126141A (en) * 1988-11-16 1992-06-30 Mediventures Incorporated Composition and method for post-surgical adhesion reduction with thermo-irreversible gels of polyoxyalkylene polymers and ionic polysaccharides
EP0551626A1 (en) * 1991-12-19 1993-07-21 LEK, tovarna farmacevtskih in kemicnih izdelkov, d.d. Thermoreversible gel as a liquid pharmaceutical carrier for a galenic formulation
US5366735A (en) * 1988-11-16 1994-11-22 Mediventures, Inc. Method and composition for post-surgical adhesion reduction

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4963367A (en) * 1984-04-27 1990-10-16 Medaphore, Inc. Drug delivery compositions and methods
KR0151704B1 (en) * 1989-11-27 1998-10-15 아만 히데아키 Fat emulsion
CA2040460C (en) * 1990-05-01 1997-06-10 Tacey X. Viegas Drug delivery with thermoreversible gels
US5484610A (en) * 1991-01-02 1996-01-16 Macromed, Inc. pH and temperature sensitive terpolymers for oral drug delivery
IT1247529B (en) * 1991-04-24 1994-12-17 Poli Ind Chimica Spa PHARMACEUTICAL COMPOSITIONS IN FOAM FORM FOR INTRAVAGINAL, SKIN AND ORAL ADMINISTRATION
IT1255460B (en) * 1992-07-28 1995-11-02 Poli Ind Chimica Spa PHARMACEUTICAL COMPOSITIONS IN THE FORM OF MICROEMULSIONS OR BIOADHESIVE LIPOSOMIAL DISPERSIONS FOR THE TRANSMUCOSAL ADMINISTRATION OF PEPTIDAL SUBSTANCES AND PHARMACOLOGICALLY ACTIVE PROTEINS
WO1994014415A1 (en) * 1992-12-24 1994-07-07 Hemagen/Pfc Fluorocarbon emulsions
ATE189772T1 (en) * 1993-04-16 2000-03-15 Wakamoto Pharma Co Ltd WATER-BASED MEDICAL COMPOSITION THAT GELS REVERSIBLY UNDER HEAT
BR9407728A (en) * 1993-10-01 1997-02-12 Syntex Inc Oral suspensions of high doses of microfenolate mofetil
MX9603858A (en) * 1994-03-04 1997-12-31 Univ Washington Block and graft copolymers and methods relating thereto.
US5585108A (en) * 1994-12-30 1996-12-17 Nanosystems L.L.C. Formulations of oral gastrointestinal therapeutic agents in combination with pharmaceutically acceptable clays

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4810503A (en) * 1987-03-31 1989-03-07 Basf Corporation Polymers which form gels at low concentrations in water
WO1990004971A1 (en) * 1988-11-03 1990-05-17 M.D.R. Group, Inc. Improved viscoelastic fluid for use in surgery and other therapies and method of using same
US5126141A (en) * 1988-11-16 1992-06-30 Mediventures Incorporated Composition and method for post-surgical adhesion reduction with thermo-irreversible gels of polyoxyalkylene polymers and ionic polysaccharides
US5366735A (en) * 1988-11-16 1994-11-22 Mediventures, Inc. Method and composition for post-surgical adhesion reduction
EP0470251A1 (en) * 1989-04-25 1992-02-12 The Green Cross Corporation Liquid prostaglandin composition
WO1991019481A1 (en) * 1990-06-15 1991-12-26 Allergan, Inc. Reversible gelation compositions and methods of use
EP0551626A1 (en) * 1991-12-19 1993-07-21 LEK, tovarna farmacevtskih in kemicnih izdelkov, d.d. Thermoreversible gel as a liquid pharmaceutical carrier for a galenic formulation

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7073512B2 (en) * 1997-05-07 2006-07-11 Saturnus Ag Adhesion prevention and an endoscopic insufflation system therefor
US6881420B2 (en) 2000-06-23 2005-04-19 Teva Pharmaceutical Industries Ltd. Compositions and dosage forms for gastric delivery of irinotecan and methods of treatment that use it to inhibit cancer cell proliferation
US7674480B2 (en) 2000-06-23 2010-03-09 Teva Pharmaceutical Industries Ltd. Rapidly expanding composition for gastric retention and controlled release of therapeutic agents, and dosage forms including the composition
WO2002070021A1 (en) * 2001-03-05 2002-09-12 Teva Pharmaceutical Industries Ltd. Gastric delivery of antineoplastic agents and methods of treatment via inhibition of cancer cell using them
US7875677B2 (en) 2001-04-20 2011-01-25 The University Of British Columbia Micellar drug delivery systems for hydrophobic drugs
EP1414497A1 (en) * 2001-08-08 2004-05-06 University Of Florida Novel microemulsion and micelle systems for solubilizing drugs
EP1414497A4 (en) * 2001-08-08 2007-11-21 Univ Florida Novel microemulsion and micelle systems for solubilizing drugs
US8802116B2 (en) 2003-02-27 2014-08-12 Novasel Australia Pty. Ltd. Poloxamer emulsion preparations
WO2005079856A1 (en) * 2004-02-23 2005-09-01 The University Of British Columbia Drug delivery compositions comprising hydrophobic polymers and amphipathic molecules
US9511020B2 (en) 2008-05-14 2016-12-06 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US8680082B2 (en) 2008-05-14 2014-03-25 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US8680083B2 (en) 2008-05-14 2014-03-25 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US9744126B2 (en) 2008-05-14 2017-08-29 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US8828980B2 (en) 2008-05-14 2014-09-09 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US8546363B2 (en) 2008-05-14 2013-10-01 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US8658626B2 (en) 2008-05-14 2014-02-25 Otonomy, Inc. Controlled release corticosteroid compositions and methods for the treatment of otic disorders
US9333171B2 (en) 2008-06-27 2016-05-10 Otonomy, Inc. Controlled-release CNS modulating compositions and methods for the treatment of otic disorders
US10918594B2 (en) 2008-06-27 2021-02-16 Otonomy, Inc. Controlled-release CNS modulating compositions and methods for the treatment of otic disorders
US8852626B2 (en) 2008-06-27 2014-10-07 Otonomy, Inc. Controlled-release CNS modulating compositions and methods for the treatment of otic disorders
US9427472B2 (en) 2008-07-21 2016-08-30 Otonomy, Inc. Controlled release compositions for modulating free-radical induced damage and methods of use thereof
US10772828B2 (en) 2008-07-21 2020-09-15 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
US9205048B2 (en) 2008-07-21 2015-12-08 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
US11369566B2 (en) 2008-07-21 2022-06-28 Alk-Abelló, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
US9233068B2 (en) 2008-07-21 2016-01-12 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of OTIC disorders
US9066855B2 (en) 2008-07-21 2015-06-30 Otonomy, Inc. Controlled release auris sensory cell modulator compositions and methods for the treatment of otic disorders
US9603796B2 (en) 2008-07-21 2017-03-28 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
US8784870B2 (en) 2008-07-21 2014-07-22 Otonomy, Inc. Controlled release compositions for modulating free-radical induced damage and methods of use thereof
US8575122B2 (en) 2008-07-21 2013-11-05 Otonomy, Inc. Controlled release auris sensory cell modulator compositions and methods for the treatment of otic disorders
US9808460B2 (en) 2008-07-21 2017-11-07 Otonomy, Inc. Controlled release auris sensory cell modulator compositions and methods for the treatment of otic disorders
US9867778B2 (en) 2008-07-21 2018-01-16 Otonomy, Inc. Controlled release antimicrobial compositions and methods for the treatment of otic disorders
US9486405B2 (en) 2013-08-27 2016-11-08 Otonomy, Inc. Methods for the treatment of pediatric otic disorders
AU2015258997B2 (en) * 2014-05-16 2019-04-18 Ultrast, Inc. Phase-shifting formulations
CN106573089A (en) * 2014-05-16 2017-04-19 奥创斯特公司 Phase-shifting formulations
WO2015175899A1 (en) * 2014-05-16 2015-11-19 Ultrast, Inc. Phase-shifting formulations
US11040004B2 (en) 2016-09-16 2021-06-22 Otonomy, Inc. Otic gel formulations for treating otitis externa

Also Published As

Publication number Publication date
JP2001526246A (en) 2001-12-18
CA2316248A1 (en) 1999-07-01
WO1999032152A2 (en) 1999-07-01
AU5533598A (en) 1999-07-12
EP1039932A1 (en) 2000-10-04
WO1999032152A3 (en) 1999-12-09
AU1943899A (en) 1999-07-12
AU753206B2 (en) 2002-10-10

Similar Documents

Publication Publication Date Title
US6280745B1 (en) Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
AU753206B2 (en) Methods and compositions for the delivery of pharmaceutical agents and/or the prevention of adhesions
US5366735A (en) Method and composition for post-surgical adhesion reduction
US5681576A (en) Method and composition for post surgical adhesion reduction
US5126141A (en) Composition and method for post-surgical adhesion reduction with thermo-irreversible gels of polyoxyalkylene polymers and ionic polysaccharides
US5292516A (en) Body cavity drug delivery with thermoreversible gels containing polyoxyalkylene copolymers
US5298260A (en) Topical drug delivery with polyoxyalkylene polymer thermoreversible gels adjustable for pH and osmolality
US5306501A (en) Drug delivery by injection with thermoreversible gels containing polyoxyalkylene copolymers
FI103714B (en) Gel-forming liquid medium composition
JP3181912B2 (en) Reversible gelling composition and method of use
AU762677B2 (en) Prolonged release bioadhesive vaginal gel dosage form
JPH02300114A (en) Remedy transmitting composite and trauma bandage composite
ES2311455T3 (en) COMPOSITIONS OF POLYACYDES, OF NON-IONIC POLYMERS OF ALKYLENE OXIDE MONOMERS AND MULTIVALENT CATIONS AND ITS USE FOR REDUCTION OF ADHERENCES.
PT2552487T (en) Thermogelling anaesthetic compositions
PT1392254E (en) Micellar drug delivery systems for hydrophobic drugs
JPH04225914A (en) Drug releasing composition using heat reversible gel
US6436425B1 (en) Method and non-gelling composition for inhibiting post-surgical adhesions
US6034088A (en) Method and composition for inhibiting post-surgical adhesions
JP4268227B2 (en) Solid particle forming composition
JP2000516614A (en) Compositions for pharmaceutical applications
KR20070122519A (en) Peg-polyacetal diblock and triblock copolymers and pharmaceutical compositions
PT87886B (en) METHOD FOR PREVENTING FIBRIN DEPOSITION OR ADHESION FORMATION AND PROCESS FOR THE PREPARATION OF THERAPEUTIC COMPOSITIONS FOR THAT END
WO1998029127A1 (en) Method and non-gelling composition for inhibiting post-surgical adhesions

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2316248

Country of ref document: CA

Ref country code: CA

Ref document number: 2316248

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 09582508

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 55335/98

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1997951781

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997951781

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1997951781

Country of ref document: EP