US20160310573A1 - Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines - Google Patents

Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines Download PDF

Info

Publication number
US20160310573A1
US20160310573A1 US15/201,032 US201615201032A US2016310573A1 US 20160310573 A1 US20160310573 A1 US 20160310573A1 US 201615201032 A US201615201032 A US 201615201032A US 2016310573 A1 US2016310573 A1 US 2016310573A1
Authority
US
United States
Prior art keywords
disc
tnf
pro
hsca
level
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US15/201,032
Inventor
Mohamed Attawia
Thomas M. DiMauro
Hassan Serhan
Sudhakar Kadiyala
David Urbahns
Scott Bruder
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DePuy Spine LLC
DePuy Synthes Products Inc
Original Assignee
DePuy Spine LLC
DePuy Synthes Products Inc
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 DePuy Spine LLC, DePuy Synthes Products Inc filed Critical DePuy Spine LLC
Priority to US15/201,032 priority Critical patent/US20160310573A1/en
Publication of US20160310573A1 publication Critical patent/US20160310573A1/en
Assigned to DEPUY SPINE, INC. reassignment DEPUY SPINE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DIMAURO, THOMAS M., KADIYALA, SUDHAKAR, SERHAN, HASSAN, URBAHNS, DAVID, ATTAWIA, MOHAMED, BRUDER, SCOTT
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/56Surgical instruments or methods for treatment of bones or joints; Devices specially adapted therefor
    • A61B17/58Surgical instruments or methods for treatment of bones or joints; Devices specially adapted therefor for osteosynthesis, e.g. bone plates, screws, setting implements or the like
    • A61B17/68Internal fixation devices, including fasteners and spinal fixators, even if a part thereof projects from the skin
    • A61B17/70Spinal positioners or stabilisers ; Bone stabilisers comprising fluid filler in an implant
    • A61B17/7061Spinal positioners or stabilisers ; Bone stabilisers comprising fluid filler in an implant for stabilising vertebrae or discs by improving the condition of their tissues, e.g. using implanted medication or fluid exchange
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B5/00Measuring for diagnostic purposes; Identification of persons
    • A61B5/05Detecting, measuring or recording for diagnosis by means of electric currents or magnetic fields; Measuring using microwaves or radio waves 
    • A61B5/055Detecting, measuring or recording for diagnosis by means of electric currents or magnetic fields; Measuring using microwaves or radio waves  involving electronic [EMR] or nuclear [NMR] magnetic resonance, e.g. magnetic resonance imaging
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/32Bones; Osteocytes; Osteoblasts; Tendons; Tenocytes; Teeth; Odontoblasts; Cartilage; Chondrocytes; Synovial membrane
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • A61K38/363Fibrinogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4833Thrombin (3.4.21.5)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • 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
    • 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
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • A61K9/0085Brain, e.g. brain implants; Spinal cord
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21005Thrombin (3.4.21.5)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]

Definitions

  • the natural intervertebral disc contains a jelly-like nucleus pulposus surrounded by a fibrous annulus fibrosus. Under an axial load, the nucleus pulposus compresses and radially transfers that load to the annulus fibrosus.
  • the laminated nature of the annulus fibrosus provides it with a high tensile strength and so allows it to expand radially in response to this transferred load.
  • ECM extracellular matrix
  • proteoglycans contained sulfated functional groups that retain water, thereby providing the nucleus pulposus within its cushioning qualities.
  • These nucleus pulposus cells may also secrete small amounts of cytokines as well as matrix metalloproteinases (“MMPs”). These cytokines and MMPs help regulate the metabolism of the nucleus pulposus cells.
  • DDD disc degeneration disease
  • mechanical instabilities in other portions of the spine In these instances, increased loads and pressures on the nucleus pulposus cause the cells to emit larger than normal amounts of the above-mentioned cytokines.
  • genetic factors such as programmed cell death, or apoptosis can also cause the cells within the nucleus pulposus to emit toxic amounts of these cytokines and MMPs.
  • the pumping action of the disc may malfunction (due to, for example, a decrease in the proteoglycan concentration within the nucleus pulposus), thereby retarding the flow of nutrients into the disc as well as the flow of waste products out of the disc. This reduced capacity to eliminate waste may result in the accumulation of high levels of toxins.
  • the toxic levels of the cytokines present in the nucleus pulposus begin to degrade the extracellular matrix (in particular, the MMPs (under mediation by the cytokines) begin cleaving the water-retaining portions of the proteoglycans, thereby reducing its water-retaining capabilities).
  • This degradation leads to a less flexible nucleus pulposus, and so changes the load pattern within the disc, thereby possibly causing delamination of the annulus fibrosus. These changes cause more mechanical instability, thereby causing the cells to emit even more cytokines, thereby upregulating MMPs.
  • the disc begins to bulge (“a herniated disc”), and then ultimately ruptures, causing the nucleus pulposus to contact the spinal cord and produce pain.
  • Olmarker, Spine 26(8), 2001, pp. 863-9 (“Olmarker I”) and Aoki, Spine 27(15), 2002, pp. 1614-17 teach that TNF- ⁇ appears to play a role in producing the pain associated with the nucleus pulposus contacting nerve roots of the spinal cord.
  • a therapeutic treatment of nerve disorders comprising administration of a therapeutically effective dosage of at least two substances selected from the group consisting of TNF inhibitors (both specific and non-specific), IL-1 inhibitors, IL-6 inhibitors, IL-8 inhibitors, FAS inhibitors, FAS ligand inhibitors, and IFN-gamma inhibitors.
  • TNF inhibitors both specific and non-specific
  • IL-1 inhibitors IL-6 inhibitors
  • IL-8 inhibitors IL-8 inhibitors
  • FAS inhibitors FAS ligand inhibitors
  • IFN-gamma inhibitors IFN-gamma inhibitors
  • Olmarker II further teaches that these substances are to be administered through systemic pathways.
  • Olmarker II teaches that “the major contribution of TNF-alpha may be derived from recruited, aggregated and maybe even extravasated leukocytes, and that successful pharmacologic block may be achieved only by systemic treatment. [0133].
  • Olmarker II appears to discourage the local addition of one therapeutic agent (doxycycline) to a transplanted nucleus pulposus. [0128].
  • Olmarker III teaches the prevention of neovasculariation and/or neo-innervation of intervertebral discs by the administration of anti-angiogenic substances. Again, however, Olmarker III teaches systemic administration of these therapeutic agents.
  • Tobinick discloses treating herniated discs with cytokine antagonists, including infliximab.
  • cytokine antagonists including infliximab.
  • Tobinick teaches that local administration involves a subcutaneous injection near the spinal cord. Accordingly, Tobinick does not teach a procedure involving a sustained delivery of a drug for the treatment of DDD, nor directly administering a specific cytokine antagonist (such as infliximab) into the disc.
  • TNF antagonists may be administered by interspinous injection in the human and that the dosage level is in the range of 1 mg to 300 mg per dose, with dosage intervals as short as two days.
  • Interleukin-1 antagonists are administered in a therapeutically effective dose, which will generally be 10 mg to 200 mg per dose, and their dosage interval will be as short as once daily.
  • Tronick III Tobinick, Swiss Med. Weekly, 2003, 133, 170-77 (“Tobinick III”) teaches both perispinal and epidural administration of TNF inhibitors for spine related therapies.
  • Karppinen, Spine, 28(8), 203, pp. 750-4 teaches intravenously injecting or orally administering infliximab into patients suffering from sciatica.
  • Karppinen does not teach a procedure involving a sustained delivery of a drug for the treatment of DDD, nor directly administering a specific cytokine antagonist (such as infliximab) into the disc.
  • anti-inflammatory agents are non-specific and therefore may produce unwanted side effects upon other cells, proteins and tissue.
  • pain-reducing effect of these agents is typically only temporary.
  • these agents typically only relieve pain, and are neither curative nor restorative.
  • Alini Eur. Spine J. 11(Supp.2), 2002, pp. S215-220, teaches therapies for early stage DDD, including injection of inhibitors of proteolytic enzymes or biological factors that stimulate cell metabolic activity (i.e., growth factors) in order to slow down the degenerative process.
  • Alini I does not disclose inhibiting growth factors.
  • Trieu discloses an intervertebral disc nucleus comprising a hydrogel that may deliver desired pharmacological agents.
  • these pharmacological agents may include growth factors such as TGF-B and anti-inflammatory drugs, including steroids.
  • these pharmacological agents may be dispersed within the hydrogel having an appropriate level of porosity to release the pharmacological agent at a desired rate.
  • Trieu teaches that these agents may be released upon cyclic loading or upon resorption.
  • Le discloses the use of anti-TNF antibodies for therapy of TNF-mediated pathologies. Le teaches parental administration of the antibodies.
  • the present inventors have developed a number of procedures for efficaciously treating degenerative disc disease by drug therapy.
  • the present inventors have developed a method of treating an intervertebral disc in which a high specificity inhibitor of a pro-inflammatory cytokine is administered transdiscally (i.e., the target tissue is a degenerating disc).
  • the present invention seeks to treat the degenerative disc at a much earlier stage of DDD than Tobinick and Karppinenen and thereby prevents degradation of the ECM.
  • nerve ending nociceptors are present within the annulus fibrosus, and that cytokines such as TNF irritate nerves. It is believed that injecting an anti-TNF antagonist into the disc space also prevents the TNF from causing nerve irritation within the disc. Thus, the pain attributed to irritation of these nerves can be efficiently eliminated.
  • this outer component of the disc may provide a suitable depot for the high specificity cytokine antagonist (HSCA), thereby increasing its half-life in the disc.
  • HSCA high specificity cytokine antagonist
  • the HSCA may be combined with other therapeutic agents (such as TGF-B, or mesenchymal stem cells) that can also be injected into the disc without reducing the effectiveness of those agents.
  • other therapeutic agents such as TGF-B, or mesenchymal stem cells
  • a method of treating an intervertebral disc having a nucleus pulposus comprising the steps of:
  • HSCA high specificity cytokine antagonist
  • a protein may be inhibited at the synthesis level, at the translation level, by shedding, by antibodies, or by soluble receptors.
  • the term “patient” refers to a human having a degenerating disc.
  • Transdiscal administration includes, but is not limited to:
  • trans-endplate administration providing the formulation in a depot at a location outside but closely adjacent the endplates of the adjacent vertebral bodies
  • the degenerating disc to which the therapeutic drug is administered may be in any one of a number of degenerative states. Accordingly, the degenerating disc may be an intact disc.
  • the degenerating disc may be a herniated disc (wherein a portion of the annulus fibrosus has a bulge).
  • the degenerating disc may be a ruptured disc (i.e., wherein the annulus fibrosus has ruptured and bulk nucleus pulposus has exuded).
  • the degenerating disc may be delaminated (wherein adjacent layers of the annulus fibrosus have separated).
  • the degenerating disc may have fissures (wherein the annulus fibrosus has fine cracks or tears through which selected molecules from the nucleus pulposus can leak).
  • the present invention is directed to providing directly through a diseased intervertebral disc at least one highly specific cytokine antagonist capable of specifically inhibiting a cytokine (preferably, a pro-inflammatory cytokine) present in the disc.
  • a cytokine preferably, a pro-inflammatory cytokine
  • the HSCA inhibits the action of a specific pro-inflammatory cytokine released by disc cells or by invading macrophages during the degenerative process.
  • the antagonist is capable of specifically inhibiting a pro-inflammatory cytokine selected from the group consisting of TNF- ⁇ , an interleukin (preferably, IL-1, 11-6 and IL-8), phospholipase A2 (PLA2), FAS, an FAS ligand, and IFN-gamma.
  • a pro-inflammatory cytokine selected from the group consisting of TNF- ⁇ , an interleukin (preferably, IL-1, 11-6 and IL-8), phospholipase A2 (PLA2), FAS, an FAS ligand, and IFN-gamma.
  • a pro-inflammatory cytokine selected from the group consisting of TNF- ⁇ , an interleukin (preferably, IL-1, 11-6 and IL-8), phospholipase A2 (PLA2), FAS, an FAS ligand, and IFN-gamma.
  • PKA2 phospholipase A2
  • FAS an FAS ligand
  • the HSCA inhibits the cytokine by preventing its production. In some embodiments, the HSCA inhibits the cytokine by binding to a membrane-bound cytokine. In others, the HSCA inhibits the cytokine by binding to a solubilized cytokine. In some embodiments, the HSCA inhibitor inhibits the cytokine by both binding to membrane bound cytokines and to solubilized cytokine. In some embodiments, the HSCA is a monoclonal antibody (“mAb”). The use of mAbs is highly desirable since they bind specifically to a certain target protein and to no other proteins. In some embodiments, the HSCA inhibits the cytokine by binding to a natural receptor of the target cytokine.
  • mAb monoclonal antibody
  • the HSCA inhibits the cytokine by preventing its production.
  • One example thereof is an inhibitor of p38 MAP kinase.
  • the TNF inhibitor inhibits the TNF by binding to membrane bound TNF in order to prevent its release from membrane.
  • the TNF inhibitor inhibits the TNF by binding to solubilized TNF.
  • One example thereof is etanercept.
  • the TNF inhibitor inhibits the TNF by both binding to membrane bound TNF and to solubilized TNF.
  • the HSCA inhibits the cytokine by binding to a natural receptor of the target cytokine.
  • Preferred TNF antagonists include, but are not limited to the following: etanercept (Enbrel.®.-Amgen); infliximab (Remicade.®.-Johnson and Johnson); D2E7, a human anti-TNF monoclonal antibody (Knoll Pharmaceuticals, Abbott Laboratories); CDP 571 (a humanized anti-TNF IgG4 antibody); CDP 870 (an anti-TNF alpha humanized monoclonal antibody fragment), both from Celltech; soluble TNF receptor Type I (Amgen); pegylated soluble TNF receptor Type I (PEGs TNF-R1) (Amgen); and onercept, a recombinant TNF binding protein (r-TBP-1) (Serono).
  • etanercept Enbrel.®.-Amgen
  • infliximab Resmicade.®.-Johnson and Johnson
  • D2E7 a human anti-TNF monoclonal antibody
  • CDP 571
  • TNF antagonists suitable for compositions, combination therapy, co-administration, devices and/or methods of the present invention include, but are not limited to, anti-TNF antibodies (e.g., at least one TNF antagonist (e.g., but not limited to a TNF chemical or protein antagonist, TNF monoclonal or polyclonal antibody or fragment, a soluble TNF receptor (e.g., p55, p70 or p85) or fragment, fusion polypeptides thereof, or a small molecule TNF antagonist, e.g., TNF binding protein I or II (TBP-1 or TBP-II), nerelimonmab, infliximab, enteracept (EnbrelTM), adalimulab (HumiraTM), CDP-571, CDP-870, afelimomab, lenercept, and the like), antigen-binding fragments thereof, and receptor molecules which
  • a “tumor necrosis factor antibody,” “TNF antibody,” “TNF ⁇ antibody,” or fragment and the like decreases, blocks, inhibits, abrogates or interferes with TNF ⁇ activity in vitro, in situ and/or preferably in vivo.
  • a suitable TNF human antibody of the present invention can bind TNF ⁇ and includes anti-TNF antibodies, antigen-binding fragments thereof, and specified mutants or domains thereof that bind specifically to TNF ⁇ .
  • a suitable TNF antibody or fragment can also decrease block, abrogate, interfere, prevent and/or inhibit TNF RNA, DNA or protein synthesis, TNF release, TNF receptor signaling, membrane TNF cleavage, TNF activity, TNF production and/or synthesis.
  • Chimeric antibody cA2 consists of the antigen binding variable region of the high-specificity neutralizing mouse anti-human TNF ⁇ IgG1 antibody, designated A2, and the constant regions of a human IgG1, kappa immunoglobulin.
  • the human IgG1 Fc region improves allogeneic antibody effector function, increases the circulating serum half-life and decreases the immunogenicity of the antibody.
  • the avidity and epitope specificity of the chimeric antibody cA2 is derived from the variable region of the murine antibody A2.
  • a preferred source for nucleic acids encoding the variable region of the murine antibody A2 is the A2 hybridoma cell line.
  • Chimeric A2 (cA2) neutralizes the cytotoxic effect of both natural and recombinant human TNF ⁇ in a dose dependent manner. From binding assays of chimeric antibody cA2 and recombinant human TNF ⁇ , the specificity constant of chimeric antibody cA2 was calculated to be 1.04 ⁇ 10 10 M ⁇ 1 . Preferred methods for determining monoclonal antibody specificity and specificity by competitive inhibition can be found in Harlow, et al., antibodies: A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988; Colligan et al., eds., Current Protocols in Immunology , Greene Publishing Assoc.
  • murine monoclonal antibody A2 is produced by a cell line designated c134A.
  • Chimeric antibody cA2 is produced by a cell line designated c168A.
  • Preferred TNF receptor molecules useful in the present invention are those that bind TNFa with high specificity (see, e.g., Feldmann et al., International Publication No. WO 92/07076 (published Apr. 30, 1992); Schall et al., Cell 61:361-370 (1990); and Loetscher et al., Cell 61:351-359 (1990), which references are entirely incorporated herein by reference) and optionally possess low immunogenicity.
  • the 55 kDa (p55 TNF-R) and the 75 kDa (p75 TNF-R) TNF cell surface receptors are useful in the present invention.
  • Truncated forms of these receptors comprising the extracellular domains (ECD) of the receptors or functional portions thereof (see, e.g., Corcoran et al., Eur. J. Biochem. 223:831-840 (1994)), are also useful in the present invention.
  • Truncated forms of the TNF receptors, comprising the ECD have been detected in urine and serum as 30 kDa and 40 kDa TNFa inhibitory binding proteins (Engelmann, H. et al., J. Biol. Chem. 265:1531-1536 (1990)).
  • TNF receptor multimeric molecules and TNF immunoreceptor fusion molecules, and derivatives and fragments or portions thereof, are additional examples of TNF receptor molecules which are useful in the methods and compositions of the present invention.
  • the TNF receptor molecules which can be used in the invention are characterized by their ability to treat patients for extended periods with good to excellent alleviation of symptoms and low toxicity. Low immunogenicity and/or high specificity, as well as other undefined properties, can contribute to the therapeutic results achieved.
  • TNF receptor multimeric molecules useful in the present invention comprise all or a functional portion of the ECD of two or more TNF receptors linked via one or more polypeptide linkers or other nonpeptide linkers, such as polyethylene glycol (PEG).
  • the multimeric molecules can further comprise a signal peptide of a secreted protein to direct expression of the multimeric molecule.
  • TNF immunoreceptor fusion molecules useful in the methods and compositions of the present invention comprise at least one portion of one or more immunoglobulin molecules and all or a functional portion of one or more TNF receptors. These immunoreceptor fusion molecules can be assembled as monomers, or hetero- or homo-multimers. The immunoreceptor fusion molecules can also be monovalent or multivalent. An example of such a TNF immunoreceptor fusion molecule is TNF receptor/IgG fusion protein. TNF immunoreceptor fusion molecules and methods for their production have been described in the art (Lesslauer et al., Eur. J. Immunol. 21:2883-2886 (1991); Ashkenazi et al., Proc. Natl. Acad. Sci.
  • a functional equivalent, derivative, fragment or region of TNF receptor molecule refers to the portion of the TNF receptor molecule, or the portion of the TNF receptor molecule sequence which encodes TNF receptor molecule, that is of sufficient size and sequences to functionally resemble TNF receptor molecules that can be used in the present invention (e.g., bind TNF ⁇ with high specificity and possess low immunogenicity).
  • a functional equivalent of TNF receptor molecule also includes modified TNF receptor molecules that functionally resemble TNF receptor molecules that can be used in the present invention (e.g., bind TNF ⁇ with high specificity and possess low immunogenicity).
  • a functional equivalent of TNF receptor molecule can contain a “SILENT” codon or one or more amino acid substitutions, deletions or additions (e.g., substitution of one acidic amino acid for another acidic amino acid; or substitution of one codon encoding the same or different hydrophobic amino acid for another codon encoding a hydrophobic amino acid).
  • SILENT substitution of one acidic amino acid for another acidic amino acid
  • substitution of one codon encoding the same or different hydrophobic amino acid for another codon encoding a hydrophobic amino acid See Ausubel et al., Current Protocols in Molecular Biology , Greene Publishing Assoc. and Wiley-Interscience, New York (1987-2003).
  • the monoclonal antibody that inhibits TNF-a is selected from the group consisting of monoclonal rodent-human antibodies, rodent antibodies, human antibodies or any portions thereof, having at least one antigen binding region of an immunoglobulin variable region, which antibody binds TNF.
  • this monoclonal antibody is selected from the group of compounds disclosed in U.S. Pat. No. 6,277,969, the specification of which is incorporated by reference.
  • the infliximab is delivered in a formulation having an infliximab concentration of between about 30 mg/ml and about 60 mg/ml.
  • the specific inhibitor of TNF-a is an inhibitor of p38 MAP kinase, preferably, a small molecule inhibitor of p38 MAP kinase.
  • the inhibition of p38 MAP kinase is believed to block production of both TNF-a and 11-2, both of which are pro-inflammatory cytokines.
  • the small molecule inhibitors of p38 MAP kinase are very specific & potent ( ⁇ nM). Without wishing to be tied to a theory, it is believed that inhibition of p38 should not block TGF signaling nor TGF activity. It is further believed that p38 inhibitors may also block induction of some metalloproteinases, COX 2 and NO synthetase. It is further believed that P38 inhibitors do not inhibit interleukins involved in immune cell proliferation such as IL-2.
  • the HSCA is a specific antagonist of an interleukin.
  • the target interleukin is selected from the group consisting IL-1, IL-2, IL-6 and IL-8, and IL-12.
  • Preferred antagonists include but are not limited to Kineretg (recombinant IL 1-RA, Amgen), IL1-Receptor Type 2 (Amgen) and IL-1 Trap (Regeneron).
  • DDD involves the progressive degeneration of a disc in which many factors are involved. In many of these instances, simply providing a single dose or even a regimen over the space of a few days may not be sufficient to resolve the DDD. For example, if DDD were caused in part by mechanical instability in a functional spinal unit, then simply providing a one-time therapy for the disc cells will likely only delay the onset of the DDD. Therefore, there is a need to provide a long-term drug therapy treatment of DDD that does not require multiple injections.
  • the HSCA Because it is believed that the cytokines of interest both produce pain and degrade the ECM when present within the nucleus pulposus, it is desirable for the HSCA to remain within the nucleus pulposus as long as possible in a pharmaceutically effective amount.
  • the half-life of the HSCA within the nucleus pulposus will depend upon many factors, including the size of the HSCA and its charge. In general, the larger the molecular weight of the HSCA, the more likely it is to remain contained by the annulus fibrosus portion of the disc.
  • the half-life of the HSCA is relatively short, then it would be desirable for a relatively large dose of the HSCA to be administered into the disc. In this condition, quick depletion of the HSCA would not cause the HSCA to fall below therapeutically effective levels until an extended period.
  • a large dose of the HSCA would be desirable in such instances, it is also known that nociceptors present on the inner wall of the annulus fibrosus react to increased pressure and produce pain, and that one avenue for increasing the pressure in the nucleus pulposus is to inject a critical volume of water. In some cases, the added amount could be as little as one cc by volume to produce pain. Accordingly, if a dilute concentration of an HSCA is added to the nucleus pulposus to provide a large dose, the resulting pressure increase caused by this added volume could be sufficient to cause acute pain.
  • the volume of drug delivered be no more than 1 ml, preferably no more than 0.5 ml, more preferably between 0.1 and 0.3 ml. When injected in these smaller quantities, it is believed the added volume will not cause an appreciable pressure increase in the nucleus pulposus.
  • Olmarker mixed 100 ⁇ l of a formulation comprising only 1.11 mg/ml of a monoclonal antibody into 40 mg of an extracted nucleus pulposus.
  • the concentration of the TNF-a antagonist in the administered drug is at least 100 mg/ml.
  • 100 mg of the HSCA is needed to produce the desired therapeutic result, no more than 1 ml of the drug need be injected.
  • the concentration of the TNF-a antagonist in the administered drug is at least 200 mg/ml. In this condition, no more than 0.5 ml of the drug need be injected.
  • the concentration of the TNF-a antagonist in the administered drug is at least 500 mg/ml. In this condition, between 0.03 and 0.3 ml of the drug need be injected.
  • the HSCA is provided in a sustained release device.
  • the sustained release device is adapted to remain within the disc for a prolonged period and slowly release the HSCA contained therein to the surrounding environment. This mode of delivery allows an HSCA to remain in therapeutically effective amounts within the disc for a prolonged period.
  • the HSCA is predominantly released from the sustained delivery device by its diffusion through the sustained delivery device (preferably, though a polymer). In others, the HSCA is predominantly released from the sustained delivery device by the biodegradation of the sustained delivery device (preferably, biodegradation of a polymer).
  • the sustained release device comprises a bioresorbable material whose gradual erosion causes the gradual release of the HSCA to the disc environment.
  • the sustained release device comprises a bioresorbable polymer.
  • the bioresorbable polymer has a half-life of at least one month, more preferably at least two months, more preferably at least 6 months.
  • the sustained release device provides controlled release. In others, it provides continuous release. In others, it provides intermittent release. In others, the sustained release device comprises a biosensor.
  • the sustained delivery device comprises bioerodable macrospheres.
  • the HSCA is preferably contained in a gelatin (or water or other solvent) within the capsule, and is released to the disc environment when the outer shell has been eroded.
  • the device can include a plurality of capsules having outer shells of varying thickness, so that the sequential breakdown of the outer shells provides periodic release of the HSCA.
  • the sustained delivery device comprises an inflammatory-responsive delivery system, preferably comprising bioerodable microspheres that are eroded by invading macrophages.
  • an inflammatory-responsive delivery system preferably comprising bioerodable microspheres that are eroded by invading macrophages.
  • This technology provides a high correspondence between physiologic inflammation of disc environment and the release of the HSCAs into that environment.
  • the technology disclosed in Brown et al., Arthritis. Rheum. 1998 December; 41(12) pp., 2185-95 is selected.
  • the sustained delivery device comprises the devices disclosed in U.S. Pat. No. 5,728,396 (“Peery”), the specification of which is incorporated by reference in its entirety.
  • the sustained delivery device comprises a plurality (preferably at least one hundred) of water-containing chambers, each chamber containing the HSCA. Each chamber is defined by bilayer lipid membranes comprising synthetic duplicates of naturally occurring lipids.
  • the release of the drug can be controlled by varying at least one of the aqueous excipients, the lipid components, and the manufacturing parameters.
  • the formulation comprises no more than 10% lipid.
  • the DepofoamTM technology of Skyepharma PLC located in London, United Kingdom is selected.
  • the sustained delivery device comprises a delivery system disclosed in U.S. Pat. No. 5,270,300 (“Hunziker”), the specification of which is incorporated by reference in its entirety.
  • the sustained delivery device comprises the co-polymer poly-DL-lactide-co-glycolide (PLG).
  • the formulation is manufactured by combining the HSCA, the co-polymer and a solvent to form a droplet, and then evaporating the solvent to form a microsphere. The plurality of microspheres are then combined in a biocompatible diluent.
  • the HSCA is released from the co-polymer by its diffusion therethrough and by the biodegradation of the co-polymer.
  • the ProLeaseTM technology of Alkermes located in Cambridge, Mass.
  • Hydrogels can also be used to deliver the HSCA is a time-release manner to the disc environment.
  • a “hydrogel” is a substance formed when an organic polymer (natural or synthetic) is set or solidified to create a three-dimensional open-lattice structure that entraps molecules of water or other solution to form a gel. The solidification can occur, e.g., by aggregation, coagulation, hydrophobic interactions, or cross-linking.
  • the hydrogels employed in this invention rapidly solidify to keep the HSCA at the application site, thereby eliminating undesired migration from the disc.
  • the hydrogels are also biocompatible, e.g., not toxic, to cells suspended in the hydrogel.
  • a “hydrogel-HSCA composition” is a suspension of a hydrogel containing desired HSCA.
  • the hydrogel-HSCA composition forms a uniform distribution of HSCA with a well-defined and precisely controllable density.
  • the hydrogel can support very large densities of HSCA.
  • the hydrogel allows diffusion of nutrients and waste products to, and away from, the HSCA, which promotes tissue growth.
  • Hydrogels suitable for use in the present invention include water-containing gels, i.e., polymers characterized by hydrophilicity and insolubility in water. See, for instance, “Hydrogels”, pages 458-459 in Concise Encyclopedia of Polymer Science and Engineering, Eds. Mark et al., Wiley and Sons, 1990, the disclosure of which is incorporated herein by reference. Although their use is optional in the present invention, the inclusion of hydrogels is highly preferred since they tend to contribute a number of desirable qualities. By virtue of their hydrophilic, water-containing nature, hydrogels can:
  • a) house viable cells, such as mesenchymal stems cells, and
  • the hydrogel is a fine, powdery synthetic hydrogel. Suitable hydrogels exhibit an optimal combination of such properties as compatibility with the matrix polymer of choice, and biocompatability.
  • the hydrogel can include any of the following: polysaccharides, proteins, polyphosphazenes, poly(oxyethylene)-poly(oxypropylene) block polymers, poly(oxyethylene)-poly(oxypropylene) block polymers of ethylene diamine, poly(acrylic acids), poly(methacrylic acids), copolymers of acrylic acid and methacrylic acid, poly(vinyl acetate), and sulfonated polymers.
  • these polymers are at least partially soluble in aqueous solutions, e.g., water, or aqueous alcohol solutions that have charged side groups, or a monovalent ionic salt thereof.
  • aqueous solutions e.g., water, or aqueous alcohol solutions that have charged side groups, or a monovalent ionic salt thereof.
  • polymers with acidic side groups that can be reacted with cations e.g., poly(phosphazenes), poly(acrylic acids), and poly(methacrylic acids).
  • acidic groups include carboxylic acid groups, sulfonic acid groups, and halogenated (preferably fluorinated) alcohol groups.
  • polymers with basic side groups that can react with anions are poly(vinyl amines), poly(vinyl pyridine), and poly(vinyl imidazole).
  • the sustained delivery device includes a polymer selected from the group consisting of PLA, PGA, PCL, and mixtures thereof.
  • the half-life of the HSCA within the disc is relatively long, then it may be assumed that a relatively small dose of the HSCA can be administered into the disc. In this condition, the slow depletion of the HSCA would not cause the HSCA to fall below therapeutically effective levels in the disc until an extended period of time has elapsed.
  • the dose administered can be very small.
  • HSCA HSCA is effective when present in the range of 1-10 mg/kg or 1-10 ppm (as is believed to be the case for the TNF antagonist RemicadeTM), and since a typical nucleus pulposus of a disc has a volume of about 3 ml (or 3 cc, or 3g), then only about 3-30 ug of the HSCA need be administered to the disc in order to provide a long lasting effective amount of the drug.
  • Tobinick discloses that at least 1 mg of cytokine antagonist should be administered perispinally in order to cure back pain.
  • Olmarker mixed 100 ml of a formulation comprising 1.11 mg/ml of a monoclonal antibody into 40 mg of an extracted nucleus pulposus, thereby producing a monoclonal antibody concentration of about 3 parts per thousand.
  • the smaller amounts available by this route reduce the chances of deleterious side effects of the HSCA.
  • infliximab concentration in the disc of about 6 mg/ml, or 6 parts per thousand.
  • infliximab has the same half-life within a nucleus pulposus as it does when administered systemically (i.e., about 1 week)
  • the concentration of infliximab would remain above about 10 ppm for about 9 weeks. Therefore, if another dose were needed, the clinician would only need to provide the second dose after about two months.
  • the HSCA is provided in a dose of less than 1 mg, preferably, less than 0.5 mg, more preferably, less than 0.1 mg, more preferably less than 0.01 mg.
  • the HSCA provided in these smaller amounts is a TNF antagonist, more preferably is infliximab.
  • the formulation of the present invention is administered directly into the disc through the outer wall of the annulus fibrosus. More preferably, the direct administration includes depositing the HSCA in the nucleus pulposus portion of the disc. In this condition, the fibrous nature of the annulus fibrosus that surrounds the nucleus pulposus will help keep the HSCA contained within the disc.
  • the formulation of the present invention is injected into the disc through a small bore needle.
  • the needle has a bore of 22 gauge or less, so that the possibilities of producing a herniation are mitigated. More preferably, the needle has a bore of 24 gauge or less, so that the possibilities of producing a herniation are even further mitigated.
  • the volume of the direction injection of the formulation is sufficiently high so as to cause a concern of overpressurizing the nucleus pulposus, then it is preferred that at least a portion of the nucleus pulposus be removed prior to direct injection.
  • the volume of removed nucleus pulposus is substantially similar to the volume of the formulation to be injected. More preferably, the volume of removed nucleus pulposus is within 80-120% of the volume of the formulation to be injected. In addition, this procedure has the added benefit of at least partially removing some degenerated disc from the patient.
  • the formulation is delivered into the disc space through the endplate of an opposing vertebral body. This avenue eliminates the need to puncture the annulus fibrosus, and so eliminates the possibility of herniation.
  • the cytokine antagonists may therapeutically treat the disc by binding the target cytokine, and thereby reducing pain and arresting degradation of the ECM, it is believed that at least some of these antagonists do not help repair the damage done by the cytokine to the ECM.
  • a method of treating degenerative disc disease in an intervertebral disc having a nucleus pulposus comprising the steps of:
  • both the HSCA and second therapeutic agent are locally administered into the disc. Because the HSCA is specific, it does not interfere with the locally administered second therapeutic agent, and so each agent may independently work to provide therapy to the diseased disc.
  • the HSCA and second therapeutic agent are administered simultaneously. In others, the HSCA is administered first. In still others, the second therapeutic agent is administered first.
  • Other compounds which may be added to the disc include, but are not limited to: vitamins and other nutritional supplements; hormones; glycoproteins; fibronectin; peptides and proteins; carbohydrates (both simple and/or complex); proteoglycans; oligonucleotides (sense and/or antisense DNA and/or RNA); BMPs; antibodies (for example, to infectious agents, tumors, drugs or hormones); and gene therapy reagents. Genetically altered cells and/or other cells may also be included in the matrix of this invention. If desired, substances such as pain killers and narcotics may also be admixed with a polymer for delivery and release to the disc space.
  • healthy cells are introduced into the disc that have the capability of at least partially repairing any damage done to the disc during the degenerative process.
  • these cells are introduced into the nucleus pulposus and ultimately produce new extracellular matrix for the nucleus pulposus.
  • these cells are introduced into the annulus fibrosus and produce new extracellular matrix for the annulus fibrosus.
  • these cells are obtained from another human individual (allograft), while in others, the cells are obtained from the same individual (autograft).
  • the cells are taken from an intervertebral disc (and can be either nucleus pulposus cells or annulus fibrosus cells), while in others, the cells are taken from a non-disc tissue (and may be mesenchymal stem cells).
  • autograft chondrocytes such as from the knee, hip, shoulder, finger or ear may be used.
  • the viable cells comprise mesenchymal stem cells (MSCs).
  • MSCs provide a special advantage for administration into a degenerating disc because it is believed that they can more readily survive the relatively harsh environment present in the degenerating disc; that they have a desirable level of plasticity; and that they have the ability to proliferate and differentiate into the desired cells.
  • the mesenchymal stems cells are obtained from bone marrow, preferably autologous bone marrow. In others, the mesenchymal stems cells are obtained from adipose tissue, preferably autologous adipose tissue.
  • the mesenchymal stem cells injected into the disc are provided in an unconcentrated form. In others, they are provided in a concentrated form. When provided in concentrated form, they are preferably uncultured. Uncultured, concentrated MSCs can be readily obtained by centrifugation, filtration, or immuno-absorption. When filtration is selected, the methods disclosed in U.S. Pat. No. 6,049,026 (“Muschler”), the specification of which is incorporated by reference in its entirety, are preferably used. In some preferred embodiments, the matrix used to filter and concentrate the MSCs is also administered into the nucleus pulposus. If this matrix has suitable mechanical properties, it can be used to restore the height of the disc space that was lost during the degradation process.
  • growth factors encompasses any cellular product that modulates the growth or differentiation of other cells, particularly connective tissue progenitor cells.
  • the growth factors that may be used in accordance with the present invention include, but are not limited to, members of the fibroblast growth factor family, including acidic and basic fibroblast growth factor (FGF-1 and -2) and FGF-4, members of the platelet-derived growth factor (PDGF) family, including PDGF-AB, PDGF-BB and PDGF-AA; EGFs, members of the insulin-like growth factor (IGF) family, including IGF-I and -II; the TGF- ⁇ superfamily, including TGF- ⁇ 1, 2 and 3 (including MP-52), osteoid-inducing factor (OIF), angiogenin(s), endothelins, hepatocyte growth factor and keratinocyte growth factor; members of the bone morphogenetic proteins (BMP's) BMP-1, (BMP-3); BMP-2; OP-1; BMP
  • BMP's
  • the growth factor is selected from the group consisting of TGF-B, bFGF, and IGF-1. These growth factors are believed to promote regeneration of the nucleus pulposus.
  • the growth factor is TGF-B. More preferably, TGF-B is administered in an amount of between 10 ng/ml and 5000 ng/ml, more preferably between 50 ng/ml and 500 ng/ml, more preferably between 100 ng/ml and 300 ng/ml.
  • platelet concentrate is provided as the second therapeutic agent.
  • the growth factors released by the platelets are present in an amount at least two-fold (more preferably, four-fold) greater than the amount found in the blood from which the platelets were taken. More preferably, the platelet concentrate is autologous.
  • the platelet concentrate is platelet rich plasma (PRP).
  • PRP platelet rich plasma
  • PRP is advantageous because it contains growth factors that can restimulate the growth of the ECM, and because its fibrin matrix provides a suitable scaffold for new tissue growth.
  • a method of treating degenerative disc disease in an intervertebral disc having a nucleus pulposus comprising the steps of:
  • pro-inflammatory cytokines selected from the group consisting of TNF- ⁇ , an interleukin (preferably, IL-1, 11-6 and IL-8), FAS, an FAS ligand, and IFN-gamma.
  • At least one of the substances is an antagonist of TNF- ⁇ .
  • the other substance is an antagonist of an interleukin.
  • the formulation comprises a suitable biocompatible carrier such as saline.
  • the carrier is selected from the carriers disclosed in U.S. Pat. No. 6,277,969 (“Le”), the specification of which is incorporated by reference in its entirety.
  • a formulation for treating degenerative disc disease comprising:
  • a second therapeutic agent selected from the group consisting of:
  • the high specificity cytokine antagonist is selected from the group consisting of antagonists of TNF and antagonists of an interleukin.
  • the diagnostic test comprises a non-invasive diagnostic test comprising using an MRI.
  • the clinician would first perform a discogram in order to identify which disc or discs are responsible for the patient's low back pain. Next, the clinician would perform an invasive or non-invasive test upon the targeted disc in order to confirm the presence of or quantify the level of the pro-inflammatory cytokine.
  • the diagnostic test comprises an invasive test in which a portion of the disc is removed and analyzed.
  • the clinician removes a portion of the nucleus pulposus.
  • the clinician removes a portion of the annulus fibrosus.
  • the removed material is a portion of the nucleus pulposus.
  • the presence of pro-inflammatory cytokines in the removed material may detected by procedures including but not limited to electrophoresis, or an enzyme-linked immunoabsorbent assay (as per Burke, Br. JBJS, 84-B(2), 2002).
  • the diagnostic methods disclosed in U.S. Pat. No. 6,277,969 (“Le”) are selected.
  • high specificity anti-TNF- ⁇ compounds are used as diagnostic tools for detecting TNF-alpha in the patient known or suspected to have a high level of TNF-alpha.
  • the clinician After determining the levels of the different pro-inflammatory cytokines in the degenerating disc, the clinician will preferably proceed to compare these diagnosed levels against pre-determined levels of the pro-inflammatory cytokines. If the diagnosed level of the pro-inflammatory cytokine exceeds the pre-determined level, then the clinician may conclude that these higher levels are causing unwanted inflammatory action and proceed to directly inject a specific HSCA into the disc capable of inhibiting the targeted protein.
  • the predetermined level for an interleukin is at least 100 pg/ml. In some embodiments, the predetermined level for IL-6 is at least 250 pg/ml. In some embodiments, the predetermined level for IL-8 is at least 500 pg/ml. In some embodiments, the predetermined level for PGE2 is at least 1000 pg/ml. In some embodiments, the predetermined level for TNF-a is at least 500 pg/ml. In others, the predetermined level for TNF- ⁇ is at least 20 pg/ml, more preferably at least 30 pg/ml, more preferably at least 50 pg/ml, more preferably at least 1 ng/ml. In others, the predetermined level for TNF- ⁇ is at least 1 ng/disc.
  • the present invention can also be used to prevent degeneration of an intervertebral disc in a human individual, namely, by following a procedure comprising the steps of:
  • This non-limiting prophetic example describes how to transdiscally administer a formulation comprising a HSCA and saline into a nucleus pulposus of a degenerating disc.
  • the clinician uses a diagnostic test to verify that a particular disc within a patient has high levels of a particular pro-inflammatory cytokine.
  • the clinician provides a local anesthetic (such as 5 ml lidocaine) to the region dorsal of the disc of concern to reduce subcutaneous pain.
  • a local anesthetic such as 5 ml lidocaine
  • the clinician punctures the skin of the patient dorsal the disc of concern with a relatively large (e.g., 18-19 gauge) needle having a stylet therein, and advances the needle through subcutaneous fat and dorsal sacrolumbar ligament and muscles to the outer edge of the intervertebral disc.
  • a relatively large (e.g., 18-19 gauge) needle having a stylet therein, and advances the needle through subcutaneous fat and dorsal sacrolumbar ligament and muscles to the outer edge of the intervertebral disc.
  • the stylet is removed from the needle.
  • the clinician receives a syringe having a smaller gauge needle adapted to fit within the larger gauge needle.
  • This needle is typically a 22 or 24 gauge needle.
  • the barrel of the syringe contains the formulation of the present invention.
  • the formulation contains infliximab, and has an infliximab concentration of between about 30 mg/ml and about 60 mg/ml.
  • the physician advances the smaller needle co-axially through the larger needle and past the distal end of the larger needle, thereby puncturing the annulus fibrosus.
  • the smaller needle is then further advanced into the center of the nucleus pulposus.
  • the clinician depresses the plunger of the syringe, thereby injecting between about 0.1 and 1 ml of the formulation into the nucleus pulposus.
  • This non-limiting prophetic example is substantially similar to that of Example I, except that the formulation comprises a sustained release device comprising the co-polymer poly-DL-lactide-co-glycolide (PLG).
  • the formulation contains infliximab as the antagonist, and has an infliximab concentration of between about 30 mg/ml and about 60 mg/ml.

Abstract

The present invention relates to injecting a high specificity cytokine antagonist into a diseased intervertebral disc.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 14/220,273, filed Mar. 20, 2014, which is a continuation of U.S. application Ser. No. 12/291,378, filed Nov. 7, 2008, now U.S. Pat. No. 8,728,523, issued May 20, 2014, which is a continuation of U.S. application Ser. No. 11/881,926, filed Jul. 30, 2007, now Abandoned, which is a Divisional of U.S. application Ser. No. 10/456,948, filed Jun. 6, 2003, now U.S. Pat. No. 7,344,716, issued Mar. 18, 2008, which claims the benefit of U.S. Provisional Application No. 60/470,098, filed May 13, 2003. The entire teachings of the above applications are incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • The natural intervertebral disc contains a jelly-like nucleus pulposus surrounded by a fibrous annulus fibrosus. Under an axial load, the nucleus pulposus compresses and radially transfers that load to the annulus fibrosus. The laminated nature of the annulus fibrosus provides it with a high tensile strength and so allows it to expand radially in response to this transferred load.
  • In a healthy intervertebral disc, cells within the nucleus pulposus produce an extracellular matrix (ECM) containing a high percentage of proteoglycans. These proteoglycans contained sulfated functional groups that retain water, thereby providing the nucleus pulposus within its cushioning qualities. These nucleus pulposus cells may also secrete small amounts of cytokines as well as matrix metalloproteinases (“MMPs”). These cytokines and MMPs help regulate the metabolism of the nucleus pulposus cells.
  • In some instances of disc degeneration disease (DDD), gradual degeneration of the intervetebral disc is caused by mechanical instabilities in other portions of the spine. In these instances, increased loads and pressures on the nucleus pulposus cause the cells to emit larger than normal amounts of the above-mentioned cytokines. In other instances of DDD, genetic factors, such as programmed cell death, or apoptosis can also cause the cells within the nucleus pulposus to emit toxic amounts of these cytokines and MMPs. In some instances, the pumping action of the disc may malfunction (due to, for example, a decrease in the proteoglycan concentration within the nucleus pulposus), thereby retarding the flow of nutrients into the disc as well as the flow of waste products out of the disc. This reduced capacity to eliminate waste may result in the accumulation of high levels of toxins.
  • As DDD progresses, the toxic levels of the cytokines present in the nucleus pulposus begin to degrade the extracellular matrix (in particular, the MMPs (under mediation by the cytokines) begin cleaving the water-retaining portions of the proteoglycans, thereby reducing its water-retaining capabilities). This degradation leads to a less flexible nucleus pulposus, and so changes the load pattern within the disc, thereby possibly causing delamination of the annulus fibrosus. These changes cause more mechanical instability, thereby causing the cells to emit even more cytokines, thereby upregulating MMPs. As this destructive cascade continues and DDD further progresses, the disc begins to bulge (“a herniated disc”), and then ultimately ruptures, causing the nucleus pulposus to contact the spinal cord and produce pain.
  • Olmarker, Spine 26(8), 2001, pp. 863-9 (“Olmarker I”) and Aoki, Spine 27(15), 2002, pp. 1614-17 teach that TNF-α appears to play a role in producing the pain associated with the nucleus pulposus contacting nerve roots of the spinal cord.
  • US Published Patent Application No. US 2003/0039651 (“Olmarker II”) teaches a therapeutic treatment of nerve disorders comprising administration of a therapeutically effective dosage of at least two substances selected from the group consisting of TNF inhibitors (both specific and non-specific), IL-1 inhibitors, IL-6 inhibitors, IL-8 inhibitors, FAS inhibitors, FAS ligand inhibitors, and IFN-gamma inhibitors.
  • In the examples of Olmarker II, Olmarker II further teaches that these substances are to be administered through systemic pathways. In particular, Olmarker II teaches that “the major contribution of TNF-alpha may be derived from recruited, aggregated and maybe even extravasated leukocytes, and that successful pharmacologic block may be achieved only by systemic treatment. [0133]. Of note, Olmarker II appears to discourage the local addition of one therapeutic agent (doxycycline) to a transplanted nucleus pulposus. [0128].
  • PCT Published Patent Application No. WO 02/100387 (“Olmarker III”) teaches the prevention of neovasculariation and/or neo-innervation of intervertebral discs by the administration of anti-angiogenic substances. Again, however, Olmarker III teaches systemic administration of these therapeutic agents.
  • U.S. Pat. No. 6,419,944 (“Tobinick”) discloses treating herniated discs with cytokine antagonists, including infliximab. However, Tobinick teaches that local administration involves a subcutaneous injection near the spinal cord. Accordingly, Tobinick does not teach a procedure involving a sustained delivery of a drug for the treatment of DDD, nor directly administering a specific cytokine antagonist (such as infliximab) into the disc.
  • US Published Patent Application No. 2003/0049256 (Tobinick II) discloses that injection of such therapeutic molecules to the anatomic area adjacent to the spine is accomplished by interspinous injection, and preferably is accomplished by injection through the skin in the anatomic area between two adjacent spinous processes of the vertebral column.
  • Tobinick II further teaches that TNF antagonists may be administered by interspinous injection in the human and that the dosage level is in the range of 1 mg to 300 mg per dose, with dosage intervals as short as two days. Tobinick II further discloses that Interleukin-1 antagonists are administered in a therapeutically effective dose, which will generally be 10 mg to 200 mg per dose, and their dosage interval will be as short as once daily.
  • Tobinick, Swiss Med. Weekly, 2003, 133, 170-77 (“Tobinick III”) teaches both perispinal and epidural administration of TNF inhibitors for spine related therapies.
  • Karppinen, Spine, 28(8), 203, pp. 750-4, teaches intravenously injecting or orally administering infliximab into patients suffering from sciatica.
  • As with Tobinick I and II, Karppinen does not teach a procedure involving a sustained delivery of a drug for the treatment of DDD, nor directly administering a specific cytokine antagonist (such as infliximab) into the disc.
  • U.S. Pat. No. 6,352,557 (Ferree) teaches adding therapeutic substances such as anti-inflammatory medications to morselized extra-cellular matrix, and injecting that combination into an interverterbral disc.
  • However many anti-inflammatory agents are non-specific and therefore may produce unwanted side effects upon other cells, proteins and tissue. In addition, the pain-reducing effect of these agents is typically only temporary. Lastly, these agents typically only relieve pain, and are neither curative nor restorative.
  • Alini, Eur. Spine J. 11(Supp.2), 2002, pp. S215-220, teaches therapies for early stage DDD, including injection of inhibitors of proteolytic enzymes or biological factors that stimulate cell metabolic activity (i.e., growth factors) in order to slow down the degenerative process. Alini I does not disclose inhibiting growth factors.
  • US Published Patent Application US 2002/0026244 (“Trieu”) discloses an intervertebral disc nucleus comprising a hydrogel that may deliver desired pharmacological agents. Trieu teaches that these pharmacological agents may include growth factors such as TGF-B and anti-inflammatory drugs, including steroids. Trieu further teaches that these pharmacological agents may be dispersed within the hydrogel having an appropriate level of porosity to release the pharmacological agent at a desired rate. Trieu teaches that these agents may be released upon cyclic loading or upon resorption.
  • Takegami, Spine, 27(12), 2002, 1318-25 teaches that injecting TGF-B into the disc space results in enhanced replenishment of the extracellular matrix damaged by cytokines. Takegami further teaches that the half-life of a growth factor injected into the interveterbal disc can be expected to be longer than that injected into a synovial joint because the nucleus pulposus is surrounded by the fibrous structure of the annulus fibrosus, thus providing a confined environment. Diwan, Tissue Engineering in Orthopedic Surgery, 31(3) July 2000, pp. 453-464, reports on another Takegami paper that concluded that a delivery system allowing prolonged delivery (>3 days) would have to be used to obtain the observed effect of the growth factor.
  • Alini, Spine 2003 28(5), pp. 446-54, discloses a cell seeded collagen-hyaluronan scaffold supplemented with growth factors such as TGF-B, bFGF, and IGF-1 for use in regenerating a nucleus pulposus.
  • Maeda et al. Spine 2000, 25(20 pp. 166-169, 2000 reports on the in vitro response to interleukin-1 receptor antagonist protein (IRAP) of rabbit annulus fibrosus exposed to IL-1. Maeda suggests that TRAP could be useful in inhibiting the degradation of the disc.
  • Yabuki, Spine, 2001, 26(8), 870-5, teaches the use of an anti-TNF drug for the treatment of sciatica.
  • U.S. Pat. No. 6,277,969 (“Le”) discloses the use of anti-TNF antibodies for therapy of TNF-mediated pathologies. Le teaches parental administration of the antibodies.
  • In sum, when investigators suggest the administration of specific TNF-a inhibitors or specific interleukin inhibitors, the investigators appear not only to teach only the administration of those therapeutics to tissue outside the disc, but it also appears to discourage the trans-discal administration of therapeutic substances.
  • SUMMARY OF THE INVENTION
  • The present inventors have developed a number of procedures for efficaciously treating degenerative disc disease by drug therapy.
  • The present inventors have noted that although Tobinick, Olmarker and Karppinenen taught the therapeutic use of pro-inflammatory cytokine-antagonist monoclonal antibodies in treating sciatica, each of these investigators targeted tissue outside of the disc.
  • In accordance with the present invention, the present inventors have developed a method of treating an intervertebral disc in which a high specificity inhibitor of a pro-inflammatory cytokine is administered transdiscally (i.e., the target tissue is a degenerating disc).
  • There are believed to be several advantages to directly administering these therapeutic inhibitors to a targeted disc over the treatments disclosed by Tobinick and Karppinenen:
  • First, since it is known that many cytokines (such as interleukins and TNF-α) also play roles in mediating the degradation of the extracellular matrix (ECM) of the nucleus pulposus, injecting an antagonist or inhibitor of these proteins directly into the disc prevents the target cytokine from inducing any further ECM degradation. In effect, the transdiscal administration of the cytokine antagonist arrests the aging process of the degenerating disc. Accordingly, the present invention seeks to treat the degenerative disc at a much earlier stage of DDD than Tobinick and Karppinenen and thereby prevents degradation of the ECM.
  • Second, it is further known that nerve ending nociceptors are present within the annulus fibrosus, and that cytokines such as TNF irritate nerves. It is believed that injecting an anti-TNF antagonist into the disc space also prevents the TNF from causing nerve irritation within the disc. Thus, the pain attributed to irritation of these nerves can be efficiently eliminated.
  • Third, since the annulus fibrosus portion of the disc comprises a relatively dense fibrosus structure, this outer component of the disc may provide a suitable depot for the high specificity cytokine antagonist (HSCA), thereby increasing its half-life in the disc.
  • Fourth, since the high specificity antagonist inhibits only the specific cytokine of interest, the HSCA may be combined with other therapeutic agents (such as TGF-B, or mesenchymal stem cells) that can also be injected into the disc without reducing the effectiveness of those agents.
  • Fifth, since it is believed that many of the problematic cytokines are actually secreted by either nucleus pulposus or annulus fibrosus cells, transdiscal injection of the high specificity antagonists will advantageously attack the problematic cytokines at their source of origination.
  • Accordingly, in a first aspect of the present invention, there is provided a method of treating an intervertebral disc having a nucleus pulposus, comprising the steps of:
  • a) transdiscally administering a formulation comprising a high specificity cytokine antagonist (HSCA) into an intervertebral disc.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A description of preferred embodiments of the invention follows.
  • For the purposes of the present invention, the terms “inhibitor” and antagonist” are used interchangeably. A protein may be inhibited at the synthesis level, at the translation level, by shedding, by antibodies, or by soluble receptors. The term “patient” refers to a human having a degenerating disc.
  • For the purposes of the present invention “Transdiscal administration” includes, but is not limited to:
  • a) injecting a formulation into the nucleus pulposus of a degenerating disc, preferably a relatively intact degenerating disc,
  • b) injecting a formulation into the annulus fibrosus of a degenerating disc, preferably relatively intact degenerating disc.
  • c) providing the formulation in a patch attached to the outer wall of the annulus fibrosus,
  • d) providing the formulation in a depot at a location outside but closely closely adjacent the outer wall of the annulus fibrosus (hereinafter, “trans-annular administration”.
  • e) providing the formulation in a depot at a location outside but closely adjacent the endplates of the adjacent vertebral bodies (hereinafter, “trans-endplate administration”.
  • Because DDD is a continuous process, the degenerating disc to which the therapeutic drug is administered may be in any one of a number of degenerative states. Accordingly, the degenerating disc may be an intact disc. The degenerating disc may be a herniated disc (wherein a portion of the annulus fibrosus has a bulge). The degenerating disc may be a ruptured disc (i.e., wherein the annulus fibrosus has ruptured and bulk nucleus pulposus has exuded). The degenerating disc may be delaminated (wherein adjacent layers of the annulus fibrosus have separated). The degenerating disc may have fissures (wherein the annulus fibrosus has fine cracks or tears through which selected molecules from the nucleus pulposus can leak).
  • The present invention is directed to providing directly through a diseased intervertebral disc at least one highly specific cytokine antagonist capable of specifically inhibiting a cytokine (preferably, a pro-inflammatory cytokine) present in the disc. Preferably, the HSCA inhibits the action of a specific pro-inflammatory cytokine released by disc cells or by invading macrophages during the degenerative process.
  • In some embodiments, the antagonist is capable of specifically inhibiting a pro-inflammatory cytokine selected from the group consisting of TNF-α, an interleukin (preferably, IL-1, 11-6 and IL-8), phospholipase A2 (PLA2), FAS, an FAS ligand, and IFN-gamma. Such specific inhibitors include those identified on pages 5-18 of Olmarker II, the specification of which is incorporated by reference in its entirety.
  • In some embodiments, the HSCA inhibits the cytokine by preventing its production. In some embodiments, the HSCA inhibits the cytokine by binding to a membrane-bound cytokine. In others, the HSCA inhibits the cytokine by binding to a solubilized cytokine. In some embodiments, the HSCA inhibitor inhibits the cytokine by both binding to membrane bound cytokines and to solubilized cytokine. In some embodiments, the HSCA is a monoclonal antibody (“mAb”). The use of mAbs is highly desirable since they bind specifically to a certain target protein and to no other proteins. In some embodiments, the HSCA inhibits the cytokine by binding to a natural receptor of the target cytokine.
  • In some embodiments, the HSCA inhibits the cytokine by preventing its production. One example thereof is an inhibitor of p38 MAP kinase. In some embodiments, the TNF inhibitor inhibits the TNF by binding to membrane bound TNF in order to prevent its release from membrane. In others, the TNF inhibitor inhibits the TNF by binding to solubilized TNF. One example thereof is etanercept. In some embodiments, the TNF inhibitor inhibits the TNF by both binding to membrane bound TNF and to solubilized TNF. One example thereof is infliximab. In some embodiments, the HSCA inhibits the cytokine by binding to a natural receptor of the target cytokine.
  • Preferred TNF antagonists include, but are not limited to the following: etanercept (Enbrel.®.-Amgen); infliximab (Remicade.®.-Johnson and Johnson); D2E7, a human anti-TNF monoclonal antibody (Knoll Pharmaceuticals, Abbott Laboratories); CDP 571 (a humanized anti-TNF IgG4 antibody); CDP 870 (an anti-TNF alpha humanized monoclonal antibody fragment), both from Celltech; soluble TNF receptor Type I (Amgen); pegylated soluble TNF receptor Type I (PEGs TNF-R1) (Amgen); and onercept, a recombinant TNF binding protein (r-TBP-1) (Serono).
  • TNF antagonists suitable for compositions, combination therapy, co-administration, devices and/or methods of the present invention (further comprising at least one anti body, specified portion and variant thereof, of the present invention), include, but are not limited to, anti-TNF antibodies (e.g., at least one TNF antagonist (e.g., but not limited to a TNF chemical or protein antagonist, TNF monoclonal or polyclonal antibody or fragment, a soluble TNF receptor (e.g., p55, p70 or p85) or fragment, fusion polypeptides thereof, or a small molecule TNF antagonist, e.g., TNF binding protein I or II (TBP-1 or TBP-II), nerelimonmab, infliximab, enteracept (Enbrel™), adalimulab (Humira™), CDP-571, CDP-870, afelimomab, lenercept, and the like), antigen-binding fragments thereof, and receptor molecules which bind specifically to TNF; compounds which prevent and/or inhibit TNF synthesis, TNF release or its action on target cells, such as thalidomide, tenidap, phosphodiesterase inhibitors (e.g, pentoxifylline and rolipram), A2b adenosine receptor agonists and A2b adenosine receptor enhancers; compounds which prevent and/or inhibit TNF receptor signaling, such as mitogen activated protein (MAP) kinase inhibitors; compounds which block and/or inhibit membrane TNF cleavage, such as metalloproteinase inhibitors; compounds which block and/or inhibit TNF activity, such as angiotensin converting enzyme (ACE) inhibitors (e.g., captopril); and compounds which block and/or inhibit TNF production and/or synthesis, such as MAP kinase inhibitors.
  • As used herein, a “tumor necrosis factor antibody,” “TNF antibody,” “TNFα antibody,” or fragment and the like decreases, blocks, inhibits, abrogates or interferes with TNFα activity in vitro, in situ and/or preferably in vivo. For example, a suitable TNF human antibody of the present invention can bind TNFα and includes anti-TNF antibodies, antigen-binding fragments thereof, and specified mutants or domains thereof that bind specifically to TNFα. A suitable TNF antibody or fragment can also decrease block, abrogate, interfere, prevent and/or inhibit TNF RNA, DNA or protein synthesis, TNF release, TNF receptor signaling, membrane TNF cleavage, TNF activity, TNF production and/or synthesis.
  • Chimeric antibody cA2 consists of the antigen binding variable region of the high-specificity neutralizing mouse anti-human TNFα IgG1 antibody, designated A2, and the constant regions of a human IgG1, kappa immunoglobulin. The human IgG1 Fc region improves allogeneic antibody effector function, increases the circulating serum half-life and decreases the immunogenicity of the antibody. The avidity and epitope specificity of the chimeric antibody cA2 is derived from the variable region of the murine antibody A2. In a particular embodiment, a preferred source for nucleic acids encoding the variable region of the murine antibody A2 is the A2 hybridoma cell line.
  • Chimeric A2 (cA2) neutralizes the cytotoxic effect of both natural and recombinant human TNFα in a dose dependent manner. From binding assays of chimeric antibody cA2 and recombinant human TNFα, the specificity constant of chimeric antibody cA2 was calculated to be 1.04×1010M−1. Preferred methods for determining monoclonal antibody specificity and specificity by competitive inhibition can be found in Harlow, et al., antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988; Colligan et al., eds., Current Protocols in Immunology, Greene Publishing Assoc. and Wiley Interscience, New York, (1992-2000); Kozbor et al., Immunol. Today, 4:72-79 (1983); Ausubel et al., eds. Current Protocols in Molecular Biology, Wiley Interscience, New York (1987-2000); and Muller, Meth. Enzymol., 92:589-601 (1983), which references are entirely incorporated herein by reference.
  • In a particular embodiment, murine monoclonal antibody A2 is produced by a cell line designated c134A. Chimeric antibody cA2 is produced by a cell line designated c168A.
  • Additional examples of monoclonal anti-TNF antibodies that can be used in the present invention are described in the art (see, e.g., U.S. Pat. No. 5,231,024; Moller, A. et al., Cytokine 2(3):162-169 (1990); U.S. application Ser. No. 07/943,852 (filed Sep. 11, 1992); Rathj en et al., International Publication No. WO 91/02078 (published Feb. 21, 1991); Rubin et al., EPO Patent Publication No. 0 218 868 (published Apr. 22, 1987); Yone et al., EPO Patent Publication No. 0 288 088 (Oct. 26, 1988); Liang, et al., Biochem. Biophys. Res. Comm. 137:847-854 (1986); Meager, et al., Hybridoma 6:305-311 (1987); Fendly et al., Hybridoma 6:359-369 (1987); Bringman, et al., Hybridoma 6:489-507 (1987); and Hirai, et al., J. Immunol. Meth. 96:57-62 (1987), which references are entirely incorporated herein by reference).
  • Preferred TNF receptor molecules useful in the present invention are those that bind TNFa with high specificity (see, e.g., Feldmann et al., International Publication No. WO 92/07076 (published Apr. 30, 1992); Schall et al., Cell 61:361-370 (1990); and Loetscher et al., Cell 61:351-359 (1990), which references are entirely incorporated herein by reference) and optionally possess low immunogenicity. In particular, the 55 kDa (p55 TNF-R) and the 75 kDa (p75 TNF-R) TNF cell surface receptors are useful in the present invention. Truncated forms of these receptors, comprising the extracellular domains (ECD) of the receptors or functional portions thereof (see, e.g., Corcoran et al., Eur. J. Biochem. 223:831-840 (1994)), are also useful in the present invention. Truncated forms of the TNF receptors, comprising the ECD, have been detected in urine and serum as 30 kDa and 40 kDa TNFa inhibitory binding proteins (Engelmann, H. et al., J. Biol. Chem. 265:1531-1536 (1990)). TNF receptor multimeric molecules and TNF immunoreceptor fusion molecules, and derivatives and fragments or portions thereof, are additional examples of TNF receptor molecules which are useful in the methods and compositions of the present invention. The TNF receptor molecules which can be used in the invention are characterized by their ability to treat patients for extended periods with good to excellent alleviation of symptoms and low toxicity. Low immunogenicity and/or high specificity, as well as other undefined properties, can contribute to the therapeutic results achieved.
  • TNF receptor multimeric molecules useful in the present invention comprise all or a functional portion of the ECD of two or more TNF receptors linked via one or more polypeptide linkers or other nonpeptide linkers, such as polyethylene glycol (PEG). The multimeric molecules can further comprise a signal peptide of a secreted protein to direct expression of the multimeric molecule. These multimeric molecules and methods for their production have been described in U.S. application Ser. No. 08/437,533 (filed May 9, 1995), the content of which is entirely incorporated herein by reference.
  • TNF immunoreceptor fusion molecules useful in the methods and compositions of the present invention comprise at least one portion of one or more immunoglobulin molecules and all or a functional portion of one or more TNF receptors. These immunoreceptor fusion molecules can be assembled as monomers, or hetero- or homo-multimers. The immunoreceptor fusion molecules can also be monovalent or multivalent. An example of such a TNF immunoreceptor fusion molecule is TNF receptor/IgG fusion protein. TNF immunoreceptor fusion molecules and methods for their production have been described in the art (Lesslauer et al., Eur. J. Immunol. 21:2883-2886 (1991); Ashkenazi et al., Proc. Natl. Acad. Sci. USA 88:10535-10539 (1991); Peppel et al., J. Exp. Med. 174:1483-1489 (1991); Kolls et al., Proc. Natl. Acad. Sci. USA 91:215-219 (1994); Butler et al., Cytokine 6(6):616-623 (1994); Baker et al., Eur. J. Immunol. 24:2040-2048 (1994); Beutler et al., U.S. Pat. No. 5,447,851; and U.S. application Ser. No. 08/442,133 (filed May 16, 1995), each of which references are entirely incorporated herein by reference). Methods for producing immunoreceptor fusion molecules can also be found in Capon et al., U.S. Pat. No. 5,116,964; Capon et al., U.S. Pat. No. 5,225,538; and Capon et al., Nature 337:525-531 (1989), which references are entirely incorporated herein by reference.
  • A functional equivalent, derivative, fragment or region of TNF receptor molecule refers to the portion of the TNF receptor molecule, or the portion of the TNF receptor molecule sequence which encodes TNF receptor molecule, that is of sufficient size and sequences to functionally resemble TNF receptor molecules that can be used in the present invention (e.g., bind TNFα with high specificity and possess low immunogenicity). A functional equivalent of TNF receptor molecule also includes modified TNF receptor molecules that functionally resemble TNF receptor molecules that can be used in the present invention (e.g., bind TNFα with high specificity and possess low immunogenicity). For example, a functional equivalent of TNF receptor molecule can contain a “SILENT” codon or one or more amino acid substitutions, deletions or additions (e.g., substitution of one acidic amino acid for another acidic amino acid; or substitution of one codon encoding the same or different hydrophobic amino acid for another codon encoding a hydrophobic amino acid). See Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Assoc. and Wiley-Interscience, New York (1987-2003).
  • In some embodiments, the monoclonal antibody that inhibits TNF-a is selected from the group consisting of monoclonal rodent-human antibodies, rodent antibodies, human antibodies or any portions thereof, having at least one antigen binding region of an immunoglobulin variable region, which antibody binds TNF. Preferably, this monoclonal antibody is selected from the group of compounds disclosed in U.S. Pat. No. 6,277,969, the specification of which is incorporated by reference. In some embodiments, the infliximab is delivered in a formulation having an infliximab concentration of between about 30 mg/ml and about 60 mg/ml.
  • In some embodiments, the specific inhibitor of TNF-a is an inhibitor of p38 MAP kinase, preferably, a small molecule inhibitor of p38 MAP kinase. The inhibition of p38 MAP kinase is believed to block production of both TNF-a and 11-2, both of which are pro-inflammatory cytokines. The small molecule inhibitors of p38 MAP kinase are very specific & potent (˜nM). Without wishing to be tied to a theory, it is believed that inhibition of p38 should not block TGF signaling nor TGF activity. It is further believed that p38 inhibitors may also block induction of some metalloproteinases, COX 2 and NO synthetase. It is further believed that P38 inhibitors do not inhibit interleukins involved in immune cell proliferation such as IL-2.
  • In some embodiments, the HSCA is a specific antagonist of an interleukin. Preferably, the target interleukin is selected from the group consisting IL-1, IL-2, IL-6 and IL-8, and IL-12. Preferred antagonists include but are not limited to Kineretg (recombinant IL 1-RA, Amgen), IL1-Receptor Type 2 (Amgen) and IL-1 Trap (Regeneron).
  • The present inventors note that DDD involves the progressive degeneration of a disc in which many factors are involved. In many of these instances, simply providing a single dose or even a regimen over the space of a few days may not be sufficient to resolve the DDD. For example, if DDD were caused in part by mechanical instability in a functional spinal unit, then simply providing a one-time therapy for the disc cells will likely only delay the onset of the DDD. Therefore, there is a need to provide a long-term drug therapy treatment of DDD that does not require multiple injections.
  • Because it is believed that the cytokines of interest both produce pain and degrade the ECM when present within the nucleus pulposus, it is desirable for the HSCA to remain within the nucleus pulposus as long as possible in a pharmaceutically effective amount. The half-life of the HSCA within the nucleus pulposus will depend upon many factors, including the size of the HSCA and its charge. In general, the larger the molecular weight of the HSCA, the more likely it is to remain contained by the annulus fibrosus portion of the disc.
  • If the half-life of the HSCA is relatively short, then it would be desirable for a relatively large dose of the HSCA to be administered into the disc. In this condition, quick depletion of the HSCA would not cause the HSCA to fall below therapeutically effective levels until an extended period.
  • Although a large dose of the HSCA would be desirable in such instances, it is also known that nociceptors present on the inner wall of the annulus fibrosus react to increased pressure and produce pain, and that one avenue for increasing the pressure in the nucleus pulposus is to inject a critical volume of water. In some cases, the added amount could be as little as one cc by volume to produce pain. Accordingly, if a dilute concentration of an HSCA is added to the nucleus pulposus to provide a large dose, the resulting pressure increase caused by this added volume could be sufficient to cause acute pain.
  • For example, if it were determined that 100 mg of an HSCA was needed to therapeutically effect a nucleus pulposus, and that HSCA was provided in concentrations of 30-60 mg/ml, then at least 1.5 ml of the HSCA would need to be injected into the nucleus pulposus in order to provide the desired therapeutic effect. However, when injecting volumes into the nucleus pulposus, it is desirable that the volume of drug delivered be no more than 1 ml, preferably no more than 0.5 ml, more preferably between 0.1 and 0.3 ml. When injected in these smaller quantities, it is believed the added volume will not cause an appreciable pressure increase in the nucleus pulposus.
  • In contrast, Olmarker mixed 100 μl of a formulation comprising only 1.11 mg/ml of a monoclonal antibody into 40 mg of an extracted nucleus pulposus.
  • Accordingly, in some embodiments, the concentration of the TNF-a antagonist in the administered drug is at least 100 mg/ml. When 100 mg of the HSCA is needed to produce the desired therapeutic result, no more than 1 ml of the drug need be injected. Preferably, the concentration of the TNF-a antagonist in the administered drug is at least 200 mg/ml. In this condition, no more than 0.5 ml of the drug need be injected. Preferably, the concentration of the TNF-a antagonist in the administered drug is at least 500 mg/ml. In this condition, between 0.03 and 0.3 ml of the drug need be injected.
  • In some embodiments, the HSCA is provided in a sustained release device. The sustained release device is adapted to remain within the disc for a prolonged period and slowly release the HSCA contained therein to the surrounding environment. This mode of delivery allows an HSCA to remain in therapeutically effective amounts within the disc for a prolonged period.
  • In some embodiments, the HSCA is predominantly released from the sustained delivery device by its diffusion through the sustained delivery device (preferably, though a polymer). In others, the HSCA is predominantly released from the sustained delivery device by the biodegradation of the sustained delivery device (preferably, biodegradation of a polymer).
  • Preferably, the sustained release device comprises a bioresorbable material whose gradual erosion causes the gradual release of the HSCA to the disc environment. In some embodiments, the sustained release device comprises a bioresorbable polymer. Preferably, the bioresorbable polymer has a half-life of at least one month, more preferably at least two months, more preferably at least 6 months.
  • In some embodiments, the sustained release device provides controlled release. In others, it provides continuous release. In others, it provides intermittent release. In others, the sustained release device comprises a biosensor.
  • In some embodiments, the sustained delivery device comprises bioerodable macrospheres. The HSCA is preferably contained in a gelatin (or water or other solvent) within the capsule, and is released to the disc environment when the outer shell has been eroded. The device can include a plurality of capsules having outer shells of varying thickness, so that the sequential breakdown of the outer shells provides periodic release of the HSCA.
  • In some embodiments, the sustained delivery device comprises an inflammatory-responsive delivery system, preferably comprising bioerodable microspheres that are eroded by invading macrophages. This technology provides a high correspondence between physiologic inflammation of disc environment and the release of the HSCAs into that environment. Preferably, the technology disclosed in Brown et al., Arthritis. Rheum. 1998 December; 41(12) pp., 2185-95 is selected.
  • In some embodiments, the sustained delivery device comprises the devices disclosed in U.S. Pat. No. 5,728,396 (“Peery”), the specification of which is incorporated by reference in its entirety.
  • In some embodiments, the sustained delivery device comprises a plurality (preferably at least one hundred) of water-containing chambers, each chamber containing the HSCA. Each chamber is defined by bilayer lipid membranes comprising synthetic duplicates of naturally occurring lipids. The release of the drug can be controlled by varying at least one of the aqueous excipients, the lipid components, and the manufacturing parameters. Preferably, the formulation comprises no more than 10% lipid. In some embodiments, the Depofoam™ technology of Skyepharma PLC (located in London, United Kingdom) is selected.
  • In some embodiments, the sustained delivery device comprises a delivery system disclosed in U.S. Pat. No. 5,270,300 (“Hunziker”), the specification of which is incorporated by reference in its entirety.
  • In some embodiments, the sustained delivery device comprises the co-polymer poly-DL-lactide-co-glycolide (PLG). Preferably, the formulation is manufactured by combining the HSCA, the co-polymer and a solvent to form a droplet, and then evaporating the solvent to form a microsphere. The plurality of microspheres are then combined in a biocompatible diluent. Preferably, the HSCA is released from the co-polymer by its diffusion therethrough and by the biodegradation of the co-polymer. In some embodiments hereof, the ProLease™ technology of Alkermes (located in Cambridge, Mass.) is selected.
  • Hydrogels can also be used to deliver the HSCA is a time-release manner to the disc environment. A “hydrogel” is a substance formed when an organic polymer (natural or synthetic) is set or solidified to create a three-dimensional open-lattice structure that entraps molecules of water or other solution to form a gel. The solidification can occur, e.g., by aggregation, coagulation, hydrophobic interactions, or cross-linking. The hydrogels employed in this invention rapidly solidify to keep the HSCA at the application site, thereby eliminating undesired migration from the disc. The hydrogels are also biocompatible, e.g., not toxic, to cells suspended in the hydrogel.
  • A “hydrogel-HSCA composition” is a suspension of a hydrogel containing desired HSCA. The hydrogel-HSCA composition forms a uniform distribution of HSCA with a well-defined and precisely controllable density. Moreover, the hydrogel can support very large densities of HSCA. In addition, the hydrogel allows diffusion of nutrients and waste products to, and away from, the HSCA, which promotes tissue growth.
  • Hydrogels suitable for use in the present invention include water-containing gels, i.e., polymers characterized by hydrophilicity and insolubility in water. See, for instance, “Hydrogels”, pages 458-459 in Concise Encyclopedia of Polymer Science and Engineering, Eds. Mark et al., Wiley and Sons, 1990, the disclosure of which is incorporated herein by reference. Although their use is optional in the present invention, the inclusion of hydrogels is highly preferred since they tend to contribute a number of desirable qualities. By virtue of their hydrophilic, water-containing nature, hydrogels can:
  • a) house viable cells, such as mesenchymal stems cells, and
  • b) assist with load bearing capabilities of the disc.
  • In a preferred embodiment, the hydrogel is a fine, powdery synthetic hydrogel. Suitable hydrogels exhibit an optimal combination of such properties as compatibility with the matrix polymer of choice, and biocompatability. The hydrogel can include any of the following: polysaccharides, proteins, polyphosphazenes, poly(oxyethylene)-poly(oxypropylene) block polymers, poly(oxyethylene)-poly(oxypropylene) block polymers of ethylene diamine, poly(acrylic acids), poly(methacrylic acids), copolymers of acrylic acid and methacrylic acid, poly(vinyl acetate), and sulfonated polymers.
  • In general, these polymers are at least partially soluble in aqueous solutions, e.g., water, or aqueous alcohol solutions that have charged side groups, or a monovalent ionic salt thereof. There are many examples of polymers with acidic side groups that can be reacted with cations, e.g., poly(phosphazenes), poly(acrylic acids), and poly(methacrylic acids). Examples of acidic groups include carboxylic acid groups, sulfonic acid groups, and halogenated (preferably fluorinated) alcohol groups. Examples of polymers with basic side groups that can react with anions are poly(vinyl amines), poly(vinyl pyridine), and poly(vinyl imidazole).
  • In some embodiments, the sustained delivery device includes a polymer selected from the group consisting of PLA, PGA, PCL, and mixtures thereof.
  • If the half-life of the HSCA within the disc is relatively long, then it may be assumed that a relatively small dose of the HSCA can be administered into the disc. In this condition, the slow depletion of the HSCA would not cause the HSCA to fall below therapeutically effective levels in the disc until an extended period of time has elapsed.
  • In some embodiments in which HSCAs have long half-lives within the disc, the dose administered can be very small.
  • For example, if it is believed that an HSCA is effective when present in the range of 1-10 mg/kg or 1-10 ppm (as is believed to be the case for the TNF antagonist Remicade™), and since a typical nucleus pulposus of a disc has a volume of about 3 ml (or 3 cc, or 3g), then only about 3-30 ug of the HSCA need be administered to the disc in order to provide a long lasting effective amount of the drug. As a point of reference, Tobinick discloses that at least 1 mg of cytokine antagonist should be administered perispinally in order to cure back pain. Similarly, Olmarker mixed 100 ml of a formulation comprising 1.11 mg/ml of a monoclonal antibody into 40 mg of an extracted nucleus pulposus, thereby producing a monoclonal antibody concentration of about 3 parts per thousand. The smaller amounts available by this route reduce the chances of deleterious side effects of the HSCA.
  • For example, suppose a clinician administered 0.3 ml of 60 mg/ml infliximab into a 2.7 cc disc, thereby producing a infliximab concentration in the disc of about 6 mg/ml, or 6 parts per thousand. Without wishing to be tied to a theory, if infliximab has the same half-life within a nucleus pulposus as it does when administered systemically (i.e., about 1 week), then the concentration of infliximab would remain above about 10 ppm for about 9 weeks. Therefore, if another dose were needed, the clinician would only need to provide the second dose after about two months.
  • Therefore, in some embodiments, the HSCA is provided in a dose of less than 1 mg, preferably, less than 0.5 mg, more preferably, less than 0.1 mg, more preferably less than 0.01 mg. The smaller amounts available by this route reduce the chances of deleterious side effects of the HSCA. Preferably, the HSCA provided in these smaller amounts is a TNF antagonist, more preferably is infliximab.
  • In preferred embodiments, the formulation of the present invention is administered directly into the disc through the outer wall of the annulus fibrosus. More preferably, the direct administration includes depositing the HSCA in the nucleus pulposus portion of the disc. In this condition, the fibrous nature of the annulus fibrosus that surrounds the nucleus pulposus will help keep the HSCA contained within the disc.
  • Preferably, the formulation of the present invention is injected into the disc through a small bore needle. More preferably, the needle has a bore of 22 gauge or less, so that the possibilities of producing a herniation are mitigated. More preferably, the needle has a bore of 24 gauge or less, so that the possibilities of producing a herniation are even further mitigated.
  • If the volume of the direction injection of the formulation is sufficiently high so as to cause a concern of overpressurizing the nucleus pulposus, then it is preferred that at least a portion of the nucleus pulposus be removed prior to direct injection. Preferably, the volume of removed nucleus pulposus is substantially similar to the volume of the formulation to be injected. More preferably, the volume of removed nucleus pulposus is within 80-120% of the volume of the formulation to be injected. In addition, this procedure has the added benefit of at least partially removing some degenerated disc from the patient.
  • In other embodiments, the formulation is delivered into the disc space through the endplate of an opposing vertebral body. This avenue eliminates the need to puncture the annulus fibrosus, and so eliminates the possibility of herniation.
  • Although the cytokine antagonists may therapeutically treat the disc by binding the target cytokine, and thereby reducing pain and arresting degradation of the ECM, it is believed that at least some of these antagonists do not help repair the damage done by the cytokine to the ECM.
  • Therefore, there may be a need to provide a therapy that also helps repair the ECM.
  • In accordance with the present invention, there is provided a method of treating degenerative disc disease in an intervertebral disc having a nucleus pulposus, comprising the steps of:
  • a) administering a highly specific cytokine antagonist into a degenerating disc; and
  • b) administering a second therapeutic agent in an amount effective to at least partially repair the disc.
  • In accordance with one aspect of the invention, both the HSCA and second therapeutic agent are locally administered into the disc. Because the HSCA is specific, it does not interfere with the locally administered second therapeutic agent, and so each agent may independently work to provide therapy to the diseased disc.
  • In some embodiments, the HSCA and second therapeutic agent are administered simultaneously. In others, the HSCA is administered first. In still others, the second therapeutic agent is administered first.
  • Other compounds which may be added to the disc include, but are not limited to: vitamins and other nutritional supplements; hormones; glycoproteins; fibronectin; peptides and proteins; carbohydrates (both simple and/or complex); proteoglycans; oligonucleotides (sense and/or antisense DNA and/or RNA); BMPs; antibodies (for example, to infectious agents, tumors, drugs or hormones); and gene therapy reagents. Genetically altered cells and/or other cells may also be included in the matrix of this invention. If desired, substances such as pain killers and narcotics may also be admixed with a polymer for delivery and release to the disc space.
  • Preferably, healthy cells are introduced into the disc that have the capability of at least partially repairing any damage done to the disc during the degenerative process. In some embodiments, these cells are introduced into the nucleus pulposus and ultimately produce new extracellular matrix for the nucleus pulposus. In others, these cells are introduced into the annulus fibrosus and produce new extracellular matrix for the annulus fibrosus.
  • In some embodiments, these cells are obtained from another human individual (allograft), while in others, the cells are obtained from the same individual (autograft). In some embodiments, the cells are taken from an intervertebral disc (and can be either nucleus pulposus cells or annulus fibrosus cells), while in others, the cells are taken from a non-disc tissue (and may be mesenchymal stem cells). In others, autograft chondrocytes (such as from the knee, hip, shoulder, finger or ear) may be used.
  • Preferably, when viable cells are selected as the second therapeutic substance, the viable cells comprise mesenchymal stem cells (MSCs). MSCs provide a special advantage for administration into a degenerating disc because it is believed that they can more readily survive the relatively harsh environment present in the degenerating disc; that they have a desirable level of plasticity; and that they have the ability to proliferate and differentiate into the desired cells.
  • In some embodiments, the mesenchymal stems cells are obtained from bone marrow, preferably autologous bone marrow. In others, the mesenchymal stems cells are obtained from adipose tissue, preferably autologous adipose tissue.
  • In some embodiments, the mesenchymal stem cells injected into the disc are provided in an unconcentrated form. In others, they are provided in a concentrated form. When provided in concentrated form, they are preferably uncultured. Uncultured, concentrated MSCs can be readily obtained by centrifugation, filtration, or immuno-absorption. When filtration is selected, the methods disclosed in U.S. Pat. No. 6,049,026 (“Muschler”), the specification of which is incorporated by reference in its entirety, are preferably used. In some preferred embodiments, the matrix used to filter and concentrate the MSCs is also administered into the nucleus pulposus. If this matrix has suitable mechanical properties, it can be used to restore the height of the disc space that was lost during the degradation process.
  • As used herein, the term “growth factors” encompasses any cellular product that modulates the growth or differentiation of other cells, particularly connective tissue progenitor cells. The growth factors that may be used in accordance with the present invention include, but are not limited to, members of the fibroblast growth factor family, including acidic and basic fibroblast growth factor (FGF-1 and -2) and FGF-4, members of the platelet-derived growth factor (PDGF) family, including PDGF-AB, PDGF-BB and PDGF-AA; EGFs, members of the insulin-like growth factor (IGF) family, including IGF-I and -II; the TGF-β superfamily, including TGF-β1, 2 and 3 (including MP-52), osteoid-inducing factor (OIF), angiogenin(s), endothelins, hepatocyte growth factor and keratinocyte growth factor; members of the bone morphogenetic proteins (BMP's) BMP-1, (BMP-3); BMP-2; OP-1; BMP-2A, -2B, and -7, BMP-14 ; HBGF-1 and -2; growth differentiation factors (GDF's), members of the hedgehog family of proteins, including indian, sonic and desert hedgehog; ADMP-1; members of the interleukin (IL) family, including IL-1 thru -6; GDF-5 and members of the colony-stimulating factor (CSF) family, including CSF-1, G-CSF, and GM-CSF; and isoforms thereof.
  • In some embodiments, the growth factor is selected from the group consisting of TGF-B, bFGF, and IGF-1. These growth factors are believed to promote regeneration of the nucleus pulposus. Preferably, the growth factor is TGF-B. More preferably, TGF-B is administered in an amount of between 10 ng/ml and 5000 ng/ml, more preferably between 50 ng/ml and 500 ng/ml, more preferably between 100 ng/ml and 300 ng/ml.
  • In some embodiments, platelet concentrate is provided as the second therapeutic agent. Preferably, the growth factors released by the platelets are present in an amount at least two-fold (more preferably, four-fold) greater than the amount found in the blood from which the platelets were taken. More preferably, the platelet concentrate is autologous. In some embodiments, the platelet concentrate is platelet rich plasma (PRP). PRP is advantageous because it contains growth factors that can restimulate the growth of the ECM, and because its fibrin matrix provides a suitable scaffold for new tissue growth.
  • Since it is known that many pro-inflammatory proteins play a role in disc degeneration, and that the antagonists of the present invention are highly specific, it is further believed that injecting at least two of the highly specific antagonists of the present invention directly into the disc would be advantageous.
  • Therefore, in accordance with the present invention, there is provided a method of treating degenerative disc disease in an intervertebral disc having a nucleus pulposus, comprising the steps of:
  • a) administering a formulation comprising at least two highly specific antagonists of pro-inflammatory cytokines selected from the group consisting of TNF-α, an interleukin (preferably, IL-1, 11-6 and IL-8), FAS, an FAS ligand, and IFN-gamma.
  • Preferably, at least one of the substances is an antagonist of TNF-α. Preferably, the other substance is an antagonist of an interleukin.
  • In some embodiments, the formulation comprises a suitable biocompatible carrier such as saline. In some embodiments, the carrier is selected from the carriers disclosed in U.S. Pat. No. 6,277,969 (“Le”), the specification of which is incorporated by reference in its entirety.
  • Also in accordance with the present invention, there is provided a formulation for treating degenerative disc disease, comprising:
  • a) a high specificity cytokine antagonist, and
  • b) a second therapeutic agent selected from the group consisting of:
      • i) a growth factor, and
      • ii) viable cells.
  • In some embodiments of this formulation, the high specificity cytokine antagonist is selected from the group consisting of antagonists of TNF and antagonists of an interleukin.
  • Because the causes of low back pain may be myriad, and because of the significant cost of many of these specialized HSCAs, it would be useful for the clinician to first perform a diagnostic test in order to confirm that the targeted disc in fact possesses high levels of the targeted cytokine prior to providing the injection.
  • In one embodiment, the diagnostic test comprises a non-invasive diagnostic test comprising using an MRI.
  • Preferably, the clinician would first perform a discogram in order to identify which disc or discs are responsible for the patient's low back pain. Next, the clinician would perform an invasive or non-invasive test upon the targeted disc in order to confirm the presence of or quantify the level of the pro-inflammatory cytokine.
  • In one embodiment, the diagnostic test comprises an invasive test in which a portion of the disc is removed and analyzed. In some embodiments, the clinician removes a portion of the nucleus pulposus. In others, the clinician removes a portion of the annulus fibrosus. Preferably, the removed material is a portion of the nucleus pulposus. The presence of pro-inflammatory cytokines in the removed material may detected by procedures including but not limited to electrophoresis, or an enzyme-linked immunoabsorbent assay (as per Burke, Br. JBJS, 84-B(2), 2002).
  • In some embodiments, the diagnostic methods disclosed in U.S. Pat. No. 6,277,969 (“Le”), the specification of which is incorporated by reference in its entirety, are selected. In these methods, high specificity anti-TNF-α compounds are used as diagnostic tools for detecting TNF-alpha in the patient known or suspected to have a high level of TNF-alpha.
  • After determining the levels of the different pro-inflammatory cytokines in the degenerating disc, the clinician will preferably proceed to compare these diagnosed levels against pre-determined levels of the pro-inflammatory cytokines. If the diagnosed level of the pro-inflammatory cytokine exceeds the pre-determined level, then the clinician may conclude that these higher levels are causing unwanted inflammatory action and proceed to directly inject a specific HSCA into the disc capable of inhibiting the targeted protein.
  • In some embodiments, the predetermined level for an interleukin is at least 100 pg/ml. In some embodiments, the predetermined level for IL-6 is at least 250 pg/ml. In some embodiments, the predetermined level for IL-8 is at least 500 pg/ml. In some embodiments, the predetermined level for PGE2 is at least 1000 pg/ml. In some embodiments, the predetermined level for TNF-a is at least 500 pg/ml. In others, the predetermined level for TNF-α is at least 20 pg/ml, more preferably at least 30 pg/ml, more preferably at least 50 pg/ml, more preferably at least 1 ng/ml. In others, the predetermined level for TNF-α is at least 1 ng/disc.
  • It would also be useful to be able to determine whether directly administering the therapeutic substances of the present invention was in fact efficacious. Accordingly, one can measure the level of cytokine remaining in the disc after administration.
  • It is further believed that the present invention can also be used to prevent degeneration of an intervertebral disc in a human individual, namely, by following a procedure comprising the steps of:
  • a) determining a genetic profile of the individual,
  • b) comparing the profile of the individual against a pre-determined genetic profile level of at-risk humans,
  • c) determining that the individual is at at-risk patient, and
  • d) injecting an antagonist of the pro-inflammatory protein into a disc of the individual.
  • EXAMPLE I
  • This non-limiting prophetic example describes how to transdiscally administer a formulation comprising a HSCA and saline into a nucleus pulposus of a degenerating disc.
  • First, the clinician uses a diagnostic test to verify that a particular disc within a patient has high levels of a particular pro-inflammatory cytokine.
  • Next, the clinician provides a local anesthetic (such as 5 ml lidocaine) to the region dorsal of the disc of concern to reduce subcutaneous pain.
  • Next, the clinician punctures the skin of the patient dorsal the disc of concern with a relatively large (e.g., 18-19 gauge) needle having a stylet therein, and advances the needle through subcutaneous fat and dorsal sacrolumbar ligament and muscles to the outer edge of the intervertebral disc.
  • Next, the stylet is removed from the needle.
  • Next, the clinician receives a syringe having a smaller gauge needle adapted to fit within the larger gauge needle. This needle is typically a 22 or 24 gauge needle. The barrel of the syringe contains the formulation of the present invention.
  • The formulation contains infliximab, and has an infliximab concentration of between about 30 mg/ml and about 60 mg/ml.
  • Next, the physician advances the smaller needle co-axially through the larger needle and past the distal end of the larger needle, thereby puncturing the annulus fibrosus. The smaller needle is then further advanced into the center of the nucleus pulposus. Finally, the clinician depresses the plunger of the syringe, thereby injecting between about 0.1 and 1 ml of the formulation into the nucleus pulposus.
  • EXAMPLE II
  • This non-limiting prophetic example is substantially similar to that of Example I, except that the formulation comprises a sustained release device comprising the co-polymer poly-DL-lactide-co-glycolide (PLG). The formulation contains infliximab as the antagonist, and has an infliximab concentration of between about 30 mg/ml and about 60 mg/ml.
  • While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (10)

1-20. (canceled)
21. A method of assessing a degenerating disc, comprising:
a) removing a portion of the nucleus pulposus from the degenerating disc of the patient;
b) diagnosing a level of pro-inflammatory cytokine in a material derived from the portion;
c) comparing the diagnosed level against a pre-determined level of the pro-inflammatory cytokine;
d) determining whether the diagnosed level exceeds the pre-determined level of the pro-inflammatory cytokine, wherein a diagnosed level that exceeds the pre-determined level of the pro-inflammatory cytokine indicates that the pro-inflammatory cytokine is causing unwanted inflammatory action.
22. The method of claim 21, wherein the pro-inflammatory cytokine is an interleukin.
23. The method of claim 22, wherein the predetermined level for an interleukin is at least 100 pg/ml.
24. The method of claim 21 further comprising the steps of:
e) concluding that the diagnosed level is causing unwanted inflammatory action; and
f) injecting into the degenerating disc a therapeutic.
25. A method of therapeutically treating a patient having a degenerating disc, comprising:
a) removing a portion of the nucleus pulposus from the degenerating disc of the patient,
b) diagnosing a level of pro-inflammatory cytokine in a material derived from the removed nucleus pulposus,
c) comparing the diagnosed levels against pre-determined level of the pro-inflammatory cytokine,
d) determining if the diagnosed level exceeds the pre-determined level of the pro-inflammatory cytokine,
e) based upon the determination, injecting into the degenerating disc a therapeutic injectate.
26. The method of claim 25, wherein the therapeutic injectate comprises autologous cultured chondrocytes derived from the disc of the patient.
27. The method of claim 25, wherein the pro-inflammatory cytokine is an interleukin.
28. The method of claim 27, wherein the predetermined level for an interleukin is at least 100 pg/ml.
29. The method of claim 25, wherein the therapeutic injectate is selected based upon the determination that the diagnosed level exceeds the pre-determined level of the pro-inflammatory cytokine.
US15/201,032 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines Abandoned US20160310573A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/201,032 US20160310573A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US47009803P 2003-05-13 2003-05-13
US10/456,948 US7344716B2 (en) 2003-05-13 2003-06-06 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US11/881,926 US20080213261A1 (en) 2003-05-13 2007-07-30 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US12/291,378 US8728523B2 (en) 2003-05-13 2008-11-07 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US14/220,273 US20140199322A1 (en) 2003-05-13 2014-03-20 Transdiscal Administration of Specific Inhibitors of Pro-Inflammatory Cytokines
US15/201,032 US20160310573A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/220,273 Continuation US20140199322A1 (en) 2003-05-13 2014-03-20 Transdiscal Administration of Specific Inhibitors of Pro-Inflammatory Cytokines

Publications (1)

Publication Number Publication Date
US20160310573A1 true US20160310573A1 (en) 2016-10-27

Family

ID=36923759

Family Applications (14)

Application Number Title Priority Date Filing Date
US10/456,948 Active 2024-05-22 US7344716B2 (en) 2003-05-13 2003-06-06 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US11/881,926 Abandoned US20080213261A1 (en) 2003-05-13 2007-07-30 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US12/290,998 Expired - Fee Related US8877193B2 (en) 2003-05-13 2008-11-05 Transdiscal administration of anti-TNFα antibodies and growth differentiation factors
US12/291,378 Expired - Fee Related US8728523B2 (en) 2003-05-13 2008-11-07 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US14/220,273 Pending US20140199322A1 (en) 2003-05-13 2014-03-20 Transdiscal Administration of Specific Inhibitors of Pro-Inflammatory Cytokines
US15/099,909 Abandoned US20160228558A1 (en) 2003-05-13 2016-04-15 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US15/200,947 Abandoned US20160310535A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/201,024 Abandoned US20160310539A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/200,951 Abandoned US20160310536A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/200,984 Abandoned US20160310537A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/200,993 Abandoned US20160310538A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/201,032 Abandoned US20160310573A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/338,302 Abandoned US20170065641A1 (en) 2003-05-13 2016-10-28 Transdiscal Administration of Specific Inhibitors of Pro-Inflammatory Cytokines
US15/470,672 Abandoned US20170252375A1 (en) 2003-05-13 2017-03-27 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines

Family Applications Before (11)

Application Number Title Priority Date Filing Date
US10/456,948 Active 2024-05-22 US7344716B2 (en) 2003-05-13 2003-06-06 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US11/881,926 Abandoned US20080213261A1 (en) 2003-05-13 2007-07-30 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US12/290,998 Expired - Fee Related US8877193B2 (en) 2003-05-13 2008-11-05 Transdiscal administration of anti-TNFα antibodies and growth differentiation factors
US12/291,378 Expired - Fee Related US8728523B2 (en) 2003-05-13 2008-11-07 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US14/220,273 Pending US20140199322A1 (en) 2003-05-13 2014-03-20 Transdiscal Administration of Specific Inhibitors of Pro-Inflammatory Cytokines
US15/099,909 Abandoned US20160228558A1 (en) 2003-05-13 2016-04-15 Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US15/200,947 Abandoned US20160310535A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/201,024 Abandoned US20160310539A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/200,951 Abandoned US20160310536A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/200,984 Abandoned US20160310537A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines
US15/200,993 Abandoned US20160310538A1 (en) 2003-05-13 2016-07-01 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/338,302 Abandoned US20170065641A1 (en) 2003-05-13 2016-10-28 Transdiscal Administration of Specific Inhibitors of Pro-Inflammatory Cytokines
US15/470,672 Abandoned US20170252375A1 (en) 2003-05-13 2017-03-27 Transdiscal Administration Of Specific Inhibitors Of Pro-Inflammatory Cytokines

Country Status (2)

Country Link
US (14) US7344716B2 (en)
CN (1) CN1822824A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE49219E1 (en) 2003-11-26 2022-09-27 DePuy Synthes Products, Inc. Local intraosseous administration of bone forming agents and anti-resorptive agents, and devices therefor

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7115557B2 (en) 1998-09-25 2006-10-03 Sciaticon Ab Use of certain drugs for treating nerve root injury
US7429378B2 (en) * 2003-05-13 2008-09-30 Depuy Spine, Inc. Transdiscal administration of high affinity anti-MMP inhibitors
US8273347B2 (en) * 2003-05-13 2012-09-25 Depuy Spine, Inc. Autologous treatment of degenerated disc with cells
US7344716B2 (en) * 2003-05-13 2008-03-18 Depuy Spine, Inc. Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US20040229878A1 (en) * 2003-05-13 2004-11-18 Depuy Spine, Inc. Transdiscal administration of specific inhibitors of P38 kinase
US7553827B2 (en) 2003-08-13 2009-06-30 Depuy Spine, Inc. Transdiscal administration of cycline compounds
US8361467B2 (en) 2003-07-30 2013-01-29 Depuy Spine, Inc. Trans-capsular administration of high specificity cytokine inhibitors into orthopedic joints
US20060046960A1 (en) * 2004-09-02 2006-03-02 Mckay William F Controlled and directed local delivery of anti-inflammatory compositions
US20070036788A1 (en) * 2004-09-22 2007-02-15 Ahmed Sheriff Use of a compound for reducing the biological effectiveness of il-6
US7485468B2 (en) * 2004-10-15 2009-02-03 Galapagos Bv Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases
US8673268B2 (en) 2004-10-15 2014-03-18 Galapagos N.V. Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases
US20070253960A1 (en) * 2006-04-28 2007-11-01 Josee Roy Pharmaceutical removal of vascular extensions from a degenerating disc
US20080019975A1 (en) * 2006-07-07 2008-01-24 Bioassets Development Corporation Novel Regimens for Treating Diseases and Disorders
US8986696B2 (en) * 2007-12-21 2015-03-24 Depuy Mitek, Inc. Trans-capsular administration of p38 map kinase inhibitors into orthopedic joints
US20090162351A1 (en) * 2007-12-21 2009-06-25 Depuy Spine, Inc. Transdiscal administration of inhibitors of p38 MAP kinase
KR101841527B1 (en) * 2010-11-11 2018-03-23 애브비 바이오테크놀로지 리미티드 IMPROVED HIGH CONCENTRATION ANTI-TNFα ANTIBODY LIQUID FORMULATIONS
US11819522B2 (en) 2012-09-19 2023-11-21 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US9872937B2 (en) 2012-09-19 2018-01-23 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
EP3476410B1 (en) * 2012-09-19 2021-08-18 MicroVascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US10596202B2 (en) 2012-09-19 2020-03-24 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US9764122B2 (en) 2014-07-25 2017-09-19 Warsaw Orthopedic, Inc. Drug delivery device and methods having an occluding member
US9775978B2 (en) 2014-07-25 2017-10-03 Warsaw Orthopedic, Inc. Drug delivery device and methods having a retaining member
US10076650B2 (en) 2015-11-23 2018-09-18 Warsaw Orthopedic, Inc. Enhanced stylet for drug depot injector
USD802756S1 (en) 2016-06-23 2017-11-14 Warsaw Orthopedic, Inc. Drug pellet cartridge
US10434261B2 (en) 2016-11-08 2019-10-08 Warsaw Orthopedic, Inc. Drug pellet delivery system and method
CN108251351B (en) * 2018-01-10 2019-01-25 暨赛再生医学科技有限公司 A kind of inducer of intervertebral disc cells autophagocytosis

Family Cites Families (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6948A (en) * 1849-12-11 David vaughan
US32155A (en) * 1861-04-23 Jakob sttjber and friedrich frank
US198599A (en) * 1877-12-25 Improvement in tents
US26801A (en) * 1860-01-10 peters
US3678158A (en) 1971-05-11 1972-07-18 Worthington Bio Chem Corp Treatment of herniated intervertebral discs of mammals
GB1523965A (en) 1976-03-19 1978-09-06 Ici Ltd Pharmaceutical compositions containing steroids
DK144800C (en) 1980-04-21 1982-10-25 Forenede Bryggerier As PROCEDURE FOR THE EXTRACTION OF ENZYMES, PRIOR CU, ZN SUPEROXIDE DISMUTASE (SOD), CATALASE AND CARBONIC ACID ANHYDRASE, FROM BLOOD
US4435506A (en) 1982-08-13 1984-03-06 Miles Laboratories, Inc. Isolation of superoxide dismutase
US4870163A (en) 1985-08-29 1989-09-26 New York Blood Center, Inc. Preparation of pure human tumor necrosis factor and hybridomas producing monoclonal antibodies to human tumor necrosis factor
US4696816A (en) 1985-11-07 1987-09-29 Brown Mark D Method for treating intervertebral disc displacement with enzymes
DE3631229A1 (en) 1986-09-13 1988-03-24 Basf Ag MONOCLONAL ANTIBODIES AGAINST HUMAN TUMORNESCROSE FACTOR (TNF) AND THEIR USE
DE3888224T2 (en) 1987-04-24 1994-07-21 Teijin Ltd Determination of tumor necrosis factor; monoclonal antibody and composition.
KR0148009B1 (en) 1988-05-27 1998-08-01 그래고리 비. 아보트 Interleukin-1 inhibitors
JPH03215430A (en) 1990-01-19 1991-09-20 Kita Kiyoshi Anti-coagulant for articular cavity
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5116964A (en) 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
SE464761B (en) * 1989-05-31 1991-06-10 Post Mix Equipment Ab PROCEDURE AND DEVICE FOR CARBON ACID RESOLUTION AND COOLING OF A CHEER
US5194596A (en) 1989-07-27 1993-03-16 California Biotechnology Inc. Production of vascular endothelial cell growth factor
JP3443119B2 (en) 1989-08-07 2003-09-02 ペプテック リミテッド Tumor necrosis factor binding ligand
US5095037B1 (en) 1989-12-21 1995-12-19 Nissho Kk Combined anti-inflammatory agent
GB9022648D0 (en) 1990-10-18 1990-11-28 Charing Cross Sunley Research Polypeptide and its use
US5223248A (en) 1991-02-11 1993-06-29 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing antiplaque properties
US5656272A (en) 1991-03-18 1997-08-12 New York University Medical Center Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies
DE69231828T3 (en) 1991-03-18 2008-06-19 New York University Specific anti-human tumor necrosis factor monoclonal and chimeric antibodies
US6284471B1 (en) 1991-03-18 2001-09-04 New York University Medical Center Anti-TNFa antibodies and assays employing anti-TNFa antibodies
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
US5270300A (en) 1991-09-06 1993-12-14 Robert Francis Shaw Methods and compositions for the treatment and repair of defects or lesions in cartilage or bone
US5656644A (en) 1994-07-20 1997-08-12 Smithkline Beecham Corporation Pyridyl imidazoles
WO1993016099A2 (en) 1992-02-12 1993-08-19 Biopharm Gesellschaft Zur Biotechnologischen Entwicklung Von Pharmaka Mbh Dna sequences encoding novel growth/differentiation factors
US5447851B1 (en) 1992-04-02 1999-07-06 Univ Texas System Board Of Dna encoding a chimeric polypeptide comprising the extracellular domain of tnf receptor fused to igg vectors and host cells
US5258371A (en) 1992-05-29 1993-11-02 Kuraray Co., Ltd. Method to reduce connective tissue destruction
US5368841A (en) 1993-02-11 1994-11-29 The General Hospital Corporation Photodynamic therapy for the destruction of the synovium in the treatment of rheumatoid arthritis and the inflammatory arthritides
TW303299B (en) 1993-07-22 1997-04-21 Lilly Co Eli
US5602156A (en) * 1993-09-17 1997-02-11 The United States Of America As Represented By The Department Of Health And Human Services Method for inhibiting metalloproteinase expression
EP0727998B1 (en) 1993-11-08 2004-01-21 Smithkline Beecham Corporation Oxazoles for treating cytokine mediated diseases
DE69511245T2 (en) 1994-02-18 1999-12-16 Immuno Ag Wien COMPOSITION AND METHOD FOR PREVENTING AND TREATING IGNITION WITH IMMUNOGLOBIN A
US20030207827A1 (en) 1995-12-22 2003-11-06 William J. Boyle Osteoprotegerin
DK1238657T3 (en) 1996-02-02 2005-01-17 Alza Corp Sustained release of an active agent using an implantable system
AU724960C (en) 1996-02-09 2002-08-15 Swedish Orphan Biovitrum Ab (Publ) Composition comprising interleukin-1 inhibitor and controlled release polymer
US5842477A (en) * 1996-02-21 1998-12-01 Advanced Tissue Sciences, Inc. Method for repairing cartilage
US5942499A (en) 1996-03-05 1999-08-24 Orquest, Inc. Method of promoting bone growth with hyaluronic acid and growth factors
US5824084A (en) 1996-07-03 1998-10-20 The Cleveland Clinic Foundation Method of preparing a composite bone graft
US7069087B2 (en) * 2000-02-25 2006-06-27 Oratec Interventions, Inc. Apparatus and method for accessing and performing a function within an intervertebral disc
JP4771563B2 (en) 1996-12-06 2011-09-14 アムジエン・インコーポレーテツド Combination therapy using IL-1 inhibitors to treat IL-1 mediated diseases
US6294170B1 (en) 1997-08-08 2001-09-25 Amgen Inc. Composition and method for treating inflammatory diseases
US6316408B1 (en) * 1997-04-16 2001-11-13 Amgen Inc. Methods of use for osetoprotegerin binding protein receptors
NZ500447A (en) 1997-04-24 2001-09-28 Ortho Mcneil Pharm Inc Substituted imidazoles that inhibit cytokine production and are useful in the treatment of inflammatory disease
US5827886A (en) * 1997-05-07 1998-10-27 Thione International, Inc. Composition for relief of arthritis-induced symptoms
GB9713726D0 (en) * 1997-06-30 1997-09-03 Ciba Geigy Ag Organic compounds
US6623472B1 (en) 1997-08-16 2003-09-23 Orthogen Gentechnologic. Gmbh Method for inducing therapeutically-effective proteins
US6770623B1 (en) 1997-12-09 2004-08-03 Eli Lilly And Company Stabilized teriparatide solutions
HUP0100075A2 (en) 1997-12-18 2001-05-28 Eli Lilly And Co. Crystalline teriparatide
EP0950417A3 (en) 1998-02-23 2000-02-23 Pfizer Products Inc. Treatment of skeletal disorders
CA2323448A1 (en) 1998-03-13 1999-09-16 Merck & Co., Inc. Methods of inhibiting bone resorption
JP2002510646A (en) 1998-04-03 2002-04-09 カイロン コーポレイション Use of IGFI for treating articular cartilage disorders
US7115557B2 (en) 1998-09-25 2006-10-03 Sciaticon Ab Use of certain drugs for treating nerve root injury
SE9803710L (en) 1998-09-25 2000-03-26 A & Science Invest Ab Use of certain substances for the treatment of nerve root damage
US7067144B2 (en) * 1998-10-20 2006-06-27 Omeros Corporation Compositions and methods for systemic inhibition of cartilage degradation
US20010006948A1 (en) 1998-11-25 2001-07-05 James D. Kang Gene transfer to intervertebral disc cells
ES2188280T3 (en) 1998-12-16 2003-06-16 Aventis Pharma Ltd ACETALES HETEROARIL-CICLICOS.
DE19903876B4 (en) 1999-02-01 2006-09-28 Orthogen Gentechnologie Gmbh Process for the in vitro formation and accumulation of interleukin-1 receptor antagonists
US20030007972A1 (en) 1999-02-24 2003-01-09 Edward Tobinick Cytokine antagonists and other biologics for the treatment of bone metastases
US6015557A (en) 1999-02-24 2000-01-18 Tobinick; Edward L. Tumor necrosis factor antagonists for the treatment of neurological disorders
US6537549B2 (en) * 1999-02-24 2003-03-25 Edward L. Tobinick Cytokine antagonists for the treatment of localized disorders
US6419944B2 (en) 1999-02-24 2002-07-16 Edward L. Tobinick Cytokine antagonists for the treatment of localized disorders
US6982089B2 (en) 1999-02-24 2006-01-03 Tact Ip, Llc Cytokine antagonists for neurological and neuropsychiatric disorders
US6428576B1 (en) 1999-04-16 2002-08-06 Endospine, Ltd. System for repairing inter-vertebral discs
US6340369B1 (en) * 1999-08-13 2002-01-22 Bret A. Ferree Treating degenerative disc disease with harvested disc cells and analogues of the extracellular matrix
WO2001012630A1 (en) * 1999-08-13 2001-02-22 Sepracor Inc. Spirocyclic ligands for sigma receptors, and libraries and methods of use thereof
US6352557B1 (en) 1999-08-13 2002-03-05 Bret A. Ferree Treating degenerative disc disease through transplantion of extracellular nucleus pulposus matrix and autograft nucleus pulposus cells
US6541477B2 (en) 1999-08-27 2003-04-01 Scios, Inc. Inhibitors of p38-a kinase
US20020010471A1 (en) 2000-02-04 2002-01-24 Wironen John F. Methods for injecting materials into bone
US6723335B1 (en) * 2000-04-07 2004-04-20 Jeffrey William Moehlenbruck Methods and compositions for treating intervertebral disc degeneration
US6554830B1 (en) 2000-04-10 2003-04-29 Sdgi Holdings, Inc. Fenestrated surgical anchor and method
US6645485B2 (en) 2000-05-10 2003-11-11 Allan R. Dunn Method of treating inflammation in the joints of a body
US6800298B1 (en) 2000-05-11 2004-10-05 Clemson University Biological lubricant composition and method of applying lubricant composition
US20020032155A1 (en) 2000-06-30 2002-03-14 Ferree Bret A. Method of treating disc herniation and disc degeneration with concentrated growth and differentiation factors
US20020026244A1 (en) 2000-08-30 2002-02-28 Trieu Hai H. Intervertebral disc nucleus implants and methods
US7998507B2 (en) 2000-09-21 2011-08-16 Elan Pharma International Ltd. Nanoparticulate compositions of mitogen-activated protein (MAP) kinase inhibitors
US6756215B1 (en) 2000-10-20 2004-06-29 The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Functionalized TGF-β fusion proteins
CA2427795A1 (en) 2000-11-03 2002-05-10 Control Delivery Systems Sustained release device for treating conditions of the joint
US6593310B1 (en) * 2000-11-21 2003-07-15 Arthropharm Pty. Ltd. Treatment of osteoporosis
DE60120494T2 (en) 2000-12-22 2006-12-21 Ortho-Mcneil Pharmaceutical, Inc. SUBSTITUTES TRIAZOLDIAMINE DERIVATIVES AND THEIR USE AS KINASE INHIBITORS
US20020082697A1 (en) * 2000-12-22 2002-06-27 Damien Christopher J. Implantable osteogenic material
US20030069639A1 (en) 2001-04-14 2003-04-10 Tom Sander Methods and compositions for repair or replacement of joints and soft tissues
CA2817619A1 (en) * 2001-06-08 2002-12-08 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
WO2002100433A1 (en) * 2001-06-11 2002-12-19 Takeda Chemical Industries, Ltd. Medicinal compositions
SE0102067D0 (en) 2001-06-11 2001-06-11 A & Science Invest Ab Prevention of neovascularization of intervertebral discs and / or of tissues with local inflammation
WO2003000190A2 (en) 2001-06-25 2003-01-03 Depuy Liposomal encapsulation of glycosaminoglycans for the treatment of arthritic joints
US20030008817A1 (en) * 2001-07-03 2003-01-09 Tom Sander Cross-link reversing agent
CA2482309A1 (en) * 2001-12-10 2003-06-19 Colbar Lifescience Ltd. Methods, devices, and preparations for intervertebral disc treatment
US20030220692A1 (en) * 2002-02-09 2003-11-27 Shapiro Irving M. Preparations of nucleus pulposus cells and methods for their generation, identification, and use
FR2836681B1 (en) * 2002-03-04 2004-06-18 Centre Nat Rech Scient MODIFIED PHOSPHOCALCIC COMPOUND, INJECTION COMPOSITION CONTAINING THE SAME
CA2803741A1 (en) * 2002-07-19 2004-01-29 Abbott Biotechnology Ltd. Treatment of psoriasis with human anti-tnf alpha antibody
US7674250B2 (en) * 2002-08-05 2010-03-09 Boston Scientific Scimed, Inc. Methods of delivering therapeutic agents
AR047712A1 (en) 2002-09-07 2006-02-15 Royal Veterinary College TREATMENT METHOD OF A NATURAL SOFT SKELETTIC TISSUE INJURY MANAGING A COMPOSITION OF MESENQUIMATOSE MOTHER CELLS
WO2004039248A2 (en) 2002-10-31 2004-05-13 The General Hospital Corporation Repairing or replacing tissues or organs
US7135575B2 (en) 2003-03-03 2006-11-14 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
US20040193274A1 (en) 2003-03-28 2004-09-30 Trieu Hai H. Materials and methods for augmenting and/or repairing intervertebral discs
US20040193270A1 (en) 2003-03-31 2004-09-30 Depuyacromed, Inc. Implantable bone graft
US7344716B2 (en) * 2003-05-13 2008-03-18 Depuy Spine, Inc. Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US8273347B2 (en) * 2003-05-13 2012-09-25 Depuy Spine, Inc. Autologous treatment of degenerated disc with cells
US7553827B2 (en) 2003-08-13 2009-06-30 Depuy Spine, Inc. Transdiscal administration of cycline compounds
US7429378B2 (en) * 2003-05-13 2008-09-30 Depuy Spine, Inc. Transdiscal administration of high affinity anti-MMP inhibitors
US20040229878A1 (en) 2003-05-13 2004-11-18 Depuy Spine, Inc. Transdiscal administration of specific inhibitors of P38 kinase
KR20060014048A (en) 2003-05-13 2006-02-14 디퍼이 스파인 인코포레이티드 A method of treating degenerative disc disease
US8361467B2 (en) 2003-07-30 2013-01-29 Depuy Spine, Inc. Trans-capsular administration of high specificity cytokine inhibitors into orthopedic joints
US20050100538A1 (en) 2003-07-31 2005-05-12 Attawia Mohamed Intradiscal injection of anti-oxidants
US7897384B2 (en) * 2003-09-08 2011-03-01 Ethicon, Inc. Chondrocyte therapeutic delivery system
US20050090801A1 (en) * 2003-10-27 2005-04-28 Racz N. S. Safety spinal catheter
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
US8895540B2 (en) 2003-11-26 2014-11-25 DePuy Synthes Products, LLC Local intraosseous administration of bone forming agents and anti-resorptive agents, and devices therefor
JP5248107B2 (en) 2004-05-11 2013-07-31 ザ、ジェネラル、ホスピタル、コーポレイション Method for producing oxidation-resistant polymeric material
US7837733B2 (en) * 2004-06-29 2010-11-23 Spine Wave, Inc. Percutaneous methods for injecting a curable biomaterial into an intervertebral space
US7741273B2 (en) * 2006-04-13 2010-06-22 Warsaw Orthopedic, Inc. Drug depot implant designs
US20070282300A1 (en) * 2006-06-05 2007-12-06 Mohamed Attawia Intervertebral Disc Puncture and Treatment System
US8333697B2 (en) * 2006-06-08 2012-12-18 Warsaw Orthopedic, Inc. Diagnostic kits and methods for diagnosis of axial pain with or without radiculopathy
US20090162351A1 (en) * 2007-12-21 2009-06-25 Depuy Spine, Inc. Transdiscal administration of inhibitors of p38 MAP kinase
US8986696B2 (en) * 2007-12-21 2015-03-24 Depuy Mitek, Inc. Trans-capsular administration of p38 map kinase inhibitors into orthopedic joints
US20140276591A1 (en) * 2013-03-14 2014-09-18 Mi4Spine, Llc Percutaneous intervertebral annular regeneration

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE49219E1 (en) 2003-11-26 2022-09-27 DePuy Synthes Products, Inc. Local intraosseous administration of bone forming agents and anti-resorptive agents, and devices therefor

Also Published As

Publication number Publication date
US20080213261A1 (en) 2008-09-04
US20070237777A1 (en) 2007-10-11
US20170252375A1 (en) 2017-09-07
US20160228558A1 (en) 2016-08-11
US20090155364A1 (en) 2009-06-18
US20160310536A1 (en) 2016-10-27
US20160310538A1 (en) 2016-10-27
US8877193B2 (en) 2014-11-04
US8728523B2 (en) 2014-05-20
US20140199322A1 (en) 2014-07-17
US7344716B2 (en) 2008-03-18
CN1822824A (en) 2006-08-23
US20160310539A1 (en) 2016-10-27
US20090175943A1 (en) 2009-07-09
US20170065641A1 (en) 2017-03-09
US20160310537A1 (en) 2016-10-27
US20160310535A1 (en) 2016-10-27

Similar Documents

Publication Publication Date Title
US8728523B2 (en) Transdiscal administration of specific inhibitors of pro-inflammatory cytokines
US8361467B2 (en) Trans-capsular administration of high specificity cytokine inhibitors into orthopedic joints
US20040229878A1 (en) Transdiscal administration of specific inhibitors of P38 kinase
EP1631266B1 (en) A method of treating degenerative disc disease
US8067397B2 (en) Transdiscal administration of cycline compounds
US7429378B2 (en) Transdiscal administration of high affinity anti-MMP inhibitors
US8986696B2 (en) Trans-capsular administration of p38 map kinase inhibitors into orthopedic joints
AU2004294967B2 (en) Local intraosseous administration of bone forming agents and anti-resorptive agents, and devices therefor
US20090162351A1 (en) Transdiscal administration of inhibitors of p38 MAP kinase
AU2007203631A1 (en) Method and formulation of treating degenerative disc disease
MXPA06006007A (en) Local intraosseous administration of bone forming agents and anti-resorptive agents, and devices therefor

Legal Events

Date Code Title Description
AS Assignment

Owner name: DEPUY SPINE, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ATTAWIA, MOHAMED;DIMAURO, THOMAS M.;SERHAN, HASSAN;AND OTHERS;SIGNING DATES FROM 20090209 TO 20090223;REEL/FRAME:043268/0320

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION