Methods and non-immunogenic compositions for treating inflammatory disorders

Information

  • Patent Grant
  • 9950035
  • Patent Number
    9,950,035
  • Date Filed
    Friday, March 15, 2013
    11 years ago
  • Date Issued
    Tuesday, April 24, 2018
    6 years ago
Abstract
Methods for making non-immunogenic anti-inflammatory cytokine compositions, comprising (a) obtaining a liquid comprising cytokine producing cells from a mammalian donor; (b) contacting the liquid with a solid extraction material to generate a composition rich in interleukin-1 receptor antagonist; and performing one or both of (i) removing cells from the composition and (ii) freezing the composition. The compositions comprise two or more of IL1-ra, sTNF-R1, sTNF-RII, IGF-I, EGF, HGF, PDGF-AB, PDGF-BB, VEGF, TGF-β1, and sIL-1RII, Compositions may also contain white blood cells and platelets.
Description
INTRODUCTION

The present technology relates to methods of treating inflammatory disorders, including osteoarthritis. In particular, methods comprise use of solutions comprising cytokines, including non-immunogenic solutions derived from blood and other tissues.


Inflammation is a complex cellular and biochemical process that occurs in the affected blood vessels and adjacent tissues in response to an injury or abnormal stimulation caused by a physical, chemical, or biologic agent, such as a pathogen, allergen or irritant. The inflammatory process includes local reactions and resulting morphologic changes in tissue; the destruction or removal of the causative agent; and the responses that lead to repair and healing. In most instances, inflammation is a beneficial and transient process, which subsides as the body attacks and overcomes an infectious or other harmful agent. However, in some instances, inflammation can be chronic self-perpetuating process, for example, as part of an ongoing degenerative process (such as arthritis) or autoimmune disease, leading to destruction of tissue. Chronic inflammation is associated with a variety of disorders, including rheumatoid arthritis, atherosclerosis, ischemic heart disease, periodontitis, colitis, and some cancers.


An inflammatory response consists of a cascade of biochemical events, involving the local vascular system and immune system, and various cells within the injured tissue. The process involves the release of numerous cell-derived mediators, including histamine, interferon-gamma, interleukin-8, leukotriene, nitric oxide, prostaglandins, tumor necrosis factor-alpha, and interleukin-1. In particular, interleukin-1 (IL-1) includes a family of cytokines that can stimulate lymphocytes and macrophages, activate phagocytes, increase prostaglandin production, contribute to degeneration of bone joints, increase bone marrow cell proliferation, and are involved in many chronic inflammatory conditions. IL-1 can be generated by macrophages, monocytes, and dendritic cells, and can be part of the inflammatory response against infection.


The mode of action of IL-1 can be mediated by interleukin-1 receptor antagonist protein (IL-1ra; also known as “IRAP”). IL-1ra binds to the same receptor on the cell surface as IL-1, and thus prevents IL-1 from sending a signal to that cell. IL-1ra is secreted from white blood cells, including monocytes, macrophages, neutrophils, polymorphonuclear cells (PMNs), and other cells, and can modulate a variety of IL-1 related immune and inflammatory responses, as described by Arend W P, Malyak M, Guthridge C J, Gabay C (1998) “Interleukin-1 receptor antagonist: role in biology” Annu. Rev. Immunol. 16: 27-55. Production of IL-1ra is stimulated by several substances including adherent immunoglobulin G (IgG), other cytokines, and bacterial or viral components. IL-1ra, as well as other cytokines such as soluble tumor necrosis factor receptor 1 (sTNF-R1), soluble tumor necrosis factor receptor 2 (sTNF-R2) and (soluble interleukin receptor 1 (sIL-1RII), is an important natural anti-inflammatory protein in arthritis, colitis, and granulomatous pulmonary disease.


IL-1ra can be used in the treatment of rheumatoid arthritis, an autoimmune disease in which IL-1 plays a key role, reducing inflammation and cartilage degradation associated with the disease. For example, Kineret™ (anakinra) is a recombinant, non-glycosylated form of IL-1ra (Amgen Manufacturing, Ltd., Thousand Oaks, Calif.). Various recombinant interleukin-1 inhibitors and methods of treatment are described in U.S. Pat. No. 6,599,873, Sommer et al., issued Jul. 29, 2003; U.S. Pat. No. 5,075,222, Hannum et al., issued Dec. 24, 1991; and U.S. Application Publication No. 2005/0197293, Mellis et al., published Sep. 8, 2005 In addition, methods for producing IL-1ra from body fluids, including the use of autologous fluids, are described in U.S. Pat. No. 6,623,472, Reinecke et al., issued Sep. 23, 2003; U.S. Pat. No. 6,713,246, Reinecke et al., issued Mar. 30, 2004; and U.S. Pat. No. 6,759,188, Reinecke et al., issued Jul. 6, 2004.


Many such treatments for inflammation are known in the art. Therapies known in the art may be directed to removal of the underlying irritant or agent causing the inflammatory reaction, or by mediating one or more aspects of the inflammatory response. Examples include glucocorticoid steroids (such as hydrocortisone, cortisone, prednisone, and beclomethasone), non-steroidal anti-inflammatory drugs (such as aspirin, ibuprofen and naproxen), and immune selective anti-inflammatories. However, many such treatments present side effects, particularly during chronic administration, or have pharmacologic characteristics that limit their use. For example, while compositions and methods using IL-1ra are known in the art, they may be associated with issues regarding stability and half-life of IL-1ra as well as the amount and rate of IL-1ra provided. Moreover, many treatments do nothing to address the underlying causes of the inflammatory process. Accordingly, improved methods of treating inflammation are needed, offering one or more of improved efficacy, reduced side effects, and improved dosing characteristics.


SUMMARY

The present technology provides methods for generating solutions rich in anti-inflammatory cytokines for use in treatment of inflammation and other disorders mediated by interleukin-1 and tumor necrosis factor-alpha. Methods for generating such solutions include contacting a liquid volume of cytokine-producing cells (e.g., white blood cells) with a solid extraction material, removing the solid extraction material from the liquid. Such methods further comprise one or both of removing the cells from the liquid during or after contacting the liquid with the solid extraction material, and freeze drying (lyophilizing) the solution after the solid extraction material is removed from the liquid. In some embodiments, the liquid volume of cells is obtained from a plurality of mammalian subjects. Compositions of this technology are non-immunogenic, allowing their use in treating mammalian subjects regardless of the source of the cells used in making the solution.


In particular, the present technology provides method for making a non-immunogenic anti-inflammatory cytokine composition, including processes comprising:

  • (a) obtaining a liquid comprising cytokine producing cells, such as white blood cells, from a mammalian donor;
  • (b) contacting the liquid with a solid extraction material to generate a composition rich in interleukin-1 receptor antagonist; and
  • (c) removing cells from the composition.


    Methods also comprise
  • (a) obtaining a liquid comprising cytokine producing cells from a mammalian donor;
  • (b) contacting the liquid with a solid extraction material to generate a composition rich in interleukin-1 receptor antagonist; and
  • (c) freezing the composition to produce the non-immunogenic anti-inflammatory cytokine composition.


In other embodiments, methods comprise:

  • (a) obtaining a liquid comprising cytokine-producing cells from a mammalian donor;
  • (b) contacting the liquid with a solid extraction material to generate a composition rich in interleukin-1 receptor antagonist; and
  • (c) isolating the liquid from the solid extraction material;
  • (d) mixing the liquid with a storage solution to make a storage stabilized solution; and
  • (e) refrigerating the stabilized solution.


In various embodiments, the liquid comprising cytokine-producing cells may comprise whole blood, bone marrow aspirate, adipose tissue, urine, fractions thereof (such as platelet-rich plasma, and bone marrow concentrate), and mixtures thereof, containing (for example) white blood cells, adipose stromal cells, adipocytes, and bone marrow stromal cells. The solid extraction material may be selected from the group consisting of corundum, quartz, titanium, dextran, agarose, polyacrylamide, polystyrene, polyethylene, polyvinyl chloride, polypropylene, and combinations thereof.


In some embodiments, the liquid comprising cytokine-producing cells is prepared by a process comprising:

  • (1) loading tissue comprising cytokine-producing cells into a tube comprising a buoy disposed in the tube, wherein the buoy has a density such that the buoy is operable to reach an equilibrium position upon centrifugation of the tissue in the tube, the equilibrium position being between a first fraction and a second fraction comprising cytokine-producing cells, the second fraction having a concentration of cells greater than the concentration of cells in the first fraction;
  • (2) centrifuging the tube so that the buoy defines an interface between the first fraction and the second fraction; and
  • (3) removing the cell fraction to form the liquid comprising cytokine-producing cells.


In various embodiments, the obtaining and contacting steps comprise:

  • (1) loading tissue comprising whole blood, bone marrow aspirate, or both, and an anticoagulant into a tube comprising a buoy disposed in the tube, wherein the buoy has a density such that the buoy reaches an equilibrium position upon centrifugation of the tissue in the tube, the equilibrium position being between a first fraction and a second fraction comprising cytokine-producing cells, the second fraction having a concentration of cells greater than the concentration of cells in the first fraction;
  • (2) centrifuging the tube so that the buoy defines an interface between the first fraction and the second fraction; and
  • (3) collecting the second fraction, as the liquid comprising cytokine-producing cells;
  • (4) loading the second fraction into a concentrator assembly comprising the solid extraction material and incubating the second fraction in contact with the solid extraction material; and
  • (5) rotating the concentrator assembly at centrifugal speeds to separate from the solid extraction material a solution rich in interleukin-1 receptor antagonist having a concentration of interleukin-1 receptor antagonist greater than that of the whole blood.


In various embodiments, the anti-inflammatory cytokine composition comprises

  • (i) interleukin-1 receptor antagonist (IL-1ra) at a concentration of at least about 10,000 pg/ml;
  • (ii) soluble Tumor Necrosis Factor Receptor 1 (sTNF-R1) at a concentration of at least about 1,200 pg/ml; and
  • (iii) a protein selected from the group consisting of sTNF-RII (soluble tumor necrosis factor-receptor 2), IGF-I (insulin-like growth factor 1), EGF (epidermal growth factor), HGF (hepatocyte growth factor), PDGF-AB (platelet-derived growth factor AB), PDGF-BB (platelet-derived growth factor BB), VEGF (vascular endothelial growth factor), TGF-β1 (transforming growth factor-β1, and sIL-1RII (soluble interleukin receptor II), and mixtures thereof, wherein the concentration of the protein in the composition is greater than the concentration of the protein in normal blood.


    In some embodiments, the compositions additionally comprise white blood cells, platelets, or combinations thereof.


Methods of treating a condition mediated by interleukin-1 receptor, such as inflammation, in a human or other mammalian subject, comprising topical administration of a composition of the present technology. In various embodiments, the composition is made using a cytokine cell suspension obtained from a mammalian subject, or plurality of subjects, other than the subject to be treated.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a block diagram illustrating a method for producing an anti-inflammatory cytokine composition;



FIG. 2 is a diagram of a fractionation device;



FIG. 3 shows a device for activating a sample to generate anti-inflammatory cytokines, before (FIG. 3A) and after (FIG. 3B) centrifugation;



FIG. 4 is a diagram of a device for generating a blood clot;



FIG. 5 is a diagram of a single device capable of generating an anti-inflammatory cytokine composition;





Corresponding reference numerals indicate corresponding parts throughout the several views of the drawings. It should be noted that the figures set forth herein are intended to exemplify the general characteristics of materials, compositions, devices, and methods among those of the present technology, for the purpose of the description of certain embodiments. These figures may not precisely reflect the characteristics of any given embodiment, and are not necessarily intended to fully define or limit specific embodiments within the scope of this technology.


DESCRIPTION

The following description of technology is merely exemplary in nature of the composition, manufacture and use of one or more inventions, and is not intended to limit the scope, application, or uses of any specific invention claimed in this application or in such other applications as may be filed claiming priority to this application, or patents issuing therefrom. A non-limiting discussion of terms and phrases intended to aid understanding of the present technology is provided at the end of this Detailed Description.


The present technology relates to compositions, methods of making compositions, and methods of using compositions for the treatment of inflammatory disorders, and other disorders mediated by interleukin-1. In general, such compositions are made by a process comprising:

  • (a) obtaining a liquid comprising cytokine-producing cells (a “cytokine cell suspension,” as discussed further below) from one or more mammalian subjects; and
  • (b) fractionating the liquid to produce a protein solution comprising one or more proteins, such as interleukin-1 receptor antagonist.


Protein Compositions

The present technology provides methods for treating inflammatory disorders other disorders mediated by interleukin-1 in humans or other mammalian subjects using compositions (herein referred to as “Protein Solutions”) comprising proteins dissolved, suspended or otherwise carried for delivery to a mammalian subject in a physiologically-acceptable medium. In various embodiments, such compositions comprise proteins (e.g., cytokines) that are native to whole blood in normal mammal subjects. Such compositions may also contain viable cells, including platelets, white blood cells, and combinations thereof.


In various embodiments, the Protein Solution comprises at least two proteins selected from the group consisting of IL-1ra, sTNF-RI, sTNF-RII (soluble tumor necrosis factor-receptor 2), IGF-I (insulin-like growth factor 1), EGF (epidermal growth factor), HGF (hepatocyte growth factor), PDGF-AB (platelet-derived growth factor AB), PDGF-BB (platelet-derived growth factor BB), VEGF (vascular endothelial growth factor), TGF-β1 (transforming growth factor-β1, and sIL-1RII (soluble interleukin receptor II), wherein the concentration of each of the two proteins in the composition is greater than the concentration of each protein in normal blood. For the sake of clarity, the Protein Solution may contain three or more of the proteins from the recited group. While the concentration of every such protein in the composition may be greater than its respective concentrations in in normal blood, it is not necessary that the concentration of more than two of the proteins be greater than their respective concentrations in normal blood.


In various embodiments, a Protein Solution comprises the following components.









TABLE 1







Protein Solution Exemplary Protein Components











Normal




Whole Blood



Composition Concentration
Concentration





Component




plasma proteins
about 80 mg/ml or greater
about 67 mg/ml


(total)
about 100 mg/ml or greater



about 200 mg/ml or greater



about 250 mg/ml or greater


albumin
about 60 mg/ml or greater
about 56 mg/ml



about 100 mg/ml of greater


fibrinogen
about 3.2 mg/ml or greater
about 2.9 mg/ml



about 4 mg/ml or greater


IL-1ra
about 10,000 pg/ml or greater
about 4200 pg/ml



about 25,000 pg/ml or greater



about 30,000 pg/ml or greater



from about 25,000 to



about 110,000 pg/ml



from about 25,000 to



about 40,000 pg/ml


sTNF-RI
about 1,200 pg/ml or greater
about 630 pg/ml



about 1,800 pg/ml or greater



about 3,000 pg/ml or greater


sTNF-RII
about 3,000 pg/ml or greater
about 1200 pg/ml



about 5,000 pg/ml or greater



about 7,000 pg/ml or greater



about 9,000 pg/ml or greater


sIL-1RII
about 15,000 pg/ml or greater
about 11,800 pg/ml



about 20,000 pg/ml or greater



about 25,000 pg/ml or greater


Growth factors


EGF
about 800 pg/ml or greater
about 250 pg/ml



about 1,000 pg/ml or greater



about 1,200 pg/ml or greater


HGF
about 1,000 pg/ml or greater
about 500 pg/ml



about 2,500 pg/ml or greater



about 2,800 pg/ml or greater



about 3,000 pg/ml or greater


PDGF-AB
about 35,000 pg/ml or greater
about 6,000 pg/ml



about 50,000 pg/ml or greater



about 70,000 pg/ml or greater


PDGF-BB
about 10,000 pg/ml or greater
about 1,500 pg/ml



about 15,000 pg/ml or greater



about 20,000 pg/ml or greater


TGF-β1
about 100,000 pg/ml or greater
about 10,000 pg/ml



about 150,000 pg/ml or greater



about 190,000 pg/ml or greater


IGF-1
about 130,000 pg/ml or greater
about 70,000 pg/ml



about 150,000 pg/ml or greater



about 160,000 pg/ml or greater


VEGF
about 500 pg/ml or greater
about 150 pg/ml



about 600 pg/ml or greater



about 800 pg/ml or greater










Protein concentrations can be measured using the methods set forth in Example 4.


The composition further preferably comprises viable white blood cells, lysed white blood cells, or both. In a preferred composition, the Protein Solution comprises monocytes, granulocytes, and platelets. In various embodiments, a Protein Solution comprises the following components.









TABLE 2







Protein Solution Exemplary Cellular Components











Normal Whole Blood


Component
Composition Concentration
Concentration





white blood cells
at least about 15 k/μl
 6.5 k/μl



at least about 30 k/μl



from about 30 to about 60 k/μl



from about 40 to about 50 k/μl


red blood cells
less than about 3 M/μl
 4.5 M/μl



less than about 2 M/μl



less than about 2.5 M/μl


platelets
at least about 400 k/μl
240 k/μl



at least about 800 k/μl



at least about 1,000 k/μl


neutrophils
at least about 5 k/μl
 3.7 k/μl



at least about 10 k/μl



at least about 12 k/μl


monocytes
at least about 1 k/μl
 0.5 k/μl



at least about 2 k/μl



at least about 3 k/μl


lymphocytes
at least about 5 k/μl
 2 k/μl



at least about 10 k/μl



at least about 20 k/μl


eosinophiles
at least about 0.15 k/μl
 0.1 k/μl



at least about 0.18 k/μl


basophils
at least about 0.2 k/μl
 0.1 k/μl



at least about 0.4 k/μl



at least about 0.6 k/μl









It will be understood that this concentration is species specific. Further, it is understood that concentrations may vary among individual subjects. Thus, in methods comprising production of a Protein Solution from the blood or other tissue containing cytokine-producing cells, the concentration of proteins and cells in the Protein Solution may vary from those recited above; the values recited above are mean values for concentrations as may be seen in a population of subjects.


In various embodiments, the concentration of one or more of the proteins or other components in the Protein Solution is greater than the concentration of the component in normal blood. (Compositions with such higher concentrations of components are said to be “rich” in such components.) As referred to herein, the concentration of a component in “normal” blood or other tissue is the concentration found in the general population of mammalian subjects from which the tissue is obtained, e.g., in normal whole blood. In methods wherein the anti-inflammatory cytokine composition is derived from tissue from a specific subject, the “normal” concentration of a protein or cell may be the concentration in the blood of that individual before processing is performed to derive the protein or cell.


Thus, in various embodiments, the concentration of one or more components of the Protein Solution is greater than about 1.5 times, about 2 times, or about 3 times, greater than the concentration of the component in normal blood. For example, components may have greater concentrations in the compositions, relative to normal (whole) blood, as follows:

    • IL-1ra, at a concentration that is at least 2.5, or at least 3 or at least 5, times greater;
    • sTNF-RI, at a concentration that is at least 2, or at least 2.5 or at least 3, times greater;
    • sTNF-RII, at a concentration that is at least 2, or at least 2.5 or at least 3, times greater;
    • sIL-1RII, at a concentration that is at least 1.5, or at least 1.8 or at least 2, times greater;
    • EGF, at a concentration that is at least 2, or at least 3 or at least 5, times greater;
    • HGF, at a concentration that is at least 2, or at least 3 or at least 4, times greater;
    • PDGF-AB, at a concentration that is at least 2, or at least 3 or at least 5, times greater;
    • PDGF-BB, at a concentration that is at least 2, or at least 3 or at least 5, times greater;
    • TGF-β1, at a concentration that is at least 3, or at least 4 or at least 6, times greater;
    • IGF-1, at a concentration that is at least 1.2, or at least 1.4 or at least 1.5, times greater;
    • VEGF, at a concentration that is at least 2, or at least 2.5 or at least 3, times greater;
    • white blood cells, at a concentration that is at least about 2, or at least about 3 or at least about 4, times greater;
    • platelets, at a concentration that is at least about 2, or at least about 3 or at least 4, times greater;
    • neutrophils, at a concentration that is at least 1.5, or at least 2 or at least 3, times greater;
    • monocytes, at a concentration that is at least 3, or at least 4 or at least 6, times greater;
    • lymphocytes, at a concentration that is at least 5, or at least 8 or at least 10, times greater; and
    • basophils, at a concentration that is at least 2, or at least 4 or at least 6, times greater


      Also, the concentration of erythrocytes in the Protein Solution is preferably at least half, or at least a third, of the centration of erythrocytes in normal blood.


For example, a Protein Solution may comprise:

    • (a) at least about 10,000 pg/ml IL1-ra;
    • (b) at least about 1,200 pg/ml sTNF-RI; and
    • (c) a protein selected from the group consisting of sTNF-RII, IGF-I, EGF, HGF, PDGF-AB, PDGF-BB, VEGF, TGF-β1, and sIL-1RII, and mixtures thereof, wherein the protein has a concentration higher than the protein's baseline concentration in normal blood. In another example, a Protein Solution comprises:
    • (a) interleukin-1 receptor antagonist (IL-1ra), at a concentration at least 3 times greater than the concentration of IL-1ra in normal blood;
    • (b) soluble tissue necrosis factor-r1 (sTNF-r1), at a concentration at least 2 times greater than the concentration of IL-1ra in normal blood;
    • (c) white blood cells at a concentration at least 2 times greater than the concentration of white blood cells in normal blood; and
    • (d) platelets, at a concentration at least 2 times greater than the concentration of platelets in normal blood.


In some embodiments, the concentration of IL-1ra in the Protein Solution is preferably at least 5,000, or at least 10,000, times greater than the concentration of interleukin-1α in the Protein Solution. The ratio of IL-1ra:interleukin-1β (IL-1β) concentrations is preferably at least 100. In some embodiments, the concentration of IL-1ra in the Protein Solution is preferably at least 1500, or at least 8000, times greater than the concentration of IL-1β in the Protein Solution. The ratio of sIL-1RII:interleukin-1β (IL-1β) concentrations is preferably greater than 1. In some embodiments, the sIL-1RII in the Protein Solution is preferably at least 2000, or at least 45000, times greater the concentration of interleukin-1β in the Protein Solution.


The present technology provides Protein Solutions wherein one or more components of the Protein Solution are obtained from non-autologous sources, such as through recombinant or synthetic methods, or by isolation from allogeneic sources (i.e., from subjects of the same species as the subject to whom the solution is to be administered) or xenogeneic sources (i.e., from animal sources other than the species to whom the solution is to be administered). In some embodiments, the Protein Solutions consists, or consists essentially, of such allogeneic components. However, in various embodiments, the Protein Solution additionally comprises one or more components (e.g., white blood cells and platelets) derived from the subject to whom the solution is to be administered in a treatment methods according to this technology. Such components are, accordingly, “autologous.” In some embodiments, the Protein Solution comprises mixtures of autologous and allogeneic components.


Methods of Making Protein Solutions

Protein Solutions may be made by any of a variety of methods, including admixture of individual components, and processes wherein one or more components are derived from a source material. In various embodiments, the Protein Solution is made by fractionating a cytokine cell suspension, to produce a protein solution comprising IL1-ra.


Obtaining Protein Solutions by Contacting Cytokine-Producing Cells with an Extraction Material


In various embodiments, Protein Solutions are made by derivation of one or more components from tissue comprising cytokine-producing cells. As referred to herein, a “cytokine producing tissue” is a tissue obtained from a mammalian subject, comprising cells that are capable of producing cytokines. Such cells include white blood cells, adipose stromal cells, bone marrow stromal cells, and combinations thereof. It is understood that white blood cells include monocytes, lymphocytes, and granulocytes such as neutrophils, eosinophils, and basophils. White blood cell useful in the methods of this technology preferably include monocytes and neutrophils. Cytokine producing tissues among those useful herein include blood, adipose tissue, bone marrow, and fractions thereof, as further discussed below.


Blood useful herein includes whole blood, plasma, platelet-rich plasma, platelet-poor plasma, and blot clots. In a preferred embodiment, methods of the present technology use platelet-rich plasma (PRP), containing white blood cells and platelets, comprising the buffy coat layer created by sedimentation of whole blood. Adipose tissue useful herein includes any fat tissue, including white and brown adipose tissue, which may be derived from subcutaneous, omental/visceral, mammary, gonadal, or other adipose tissue sites. Bone marrow useful herein includes red marrow and yellow marrow. In a preferred embodiment, bone marrow is bone marrow concentrate, obtained from the red marrow of long bones, comprising hematopoietic and mesenchymal stems cells. As discussed above, compositions of the present technology may be made from blood, adipose, and bone marrow tissue from allogeneic sources, relative to the subject to be treated according to methods of this technology. Compositions may also be made from combinations of allogeneic and autologous tissues.


In some embodiments, methods comprise fractionating a liquid (a “cytokine cell suspension.”) comprising cells capable of producing cytokines, such as IL1-ra and sTNF-R1. As discussed above, such cells include white blood cells, adipose stromal cells, bone marrow stromal cells, and combinations thereof. In some embodiments, the cytokine cell suspension is a liquid comprising white blood cells. It should be understood that the cytokine cell suspension comprises cells and an extra-cellular liquid, regardless of the relative proportions of the cells and liquid. In some embodiments, the suspension may comprise primarily cells, with liquid being present as only a minor component, essentially wetting the cells. In some embodiments, the liquid may comprise two phases, consisting of a phase primarily consisting of liquid and a phase primarily consisting of cells, forming a suspension of cells in the liquid only upon agitation or other mixing.


As exemplified in FIG. 1, methods of the present technology include those comprising:

    • (a) obtaining a cytokine cell suspension, such as a liquid comprising white blood cells (steps 105, 115 or 135, or combinations thereof);
    • (b) contacting the suspension with a solid extraction material (step 140); and
    • (c) isolating a protein-containing liquid from the solid extraction material (step 150).


      Obtaining the suspension 105, 115, 135 can comprise any of a variety of methods for creating a liquid containing cells among those known in the art. Such methods include isolation from tissue and culturing. Obtaining may be performed directly in the method, whereby a health care practitioner or other individual performs isolation, processing, culturing or other processes for creating the suspension, in a procedure that includes the contacting and isolating steps. In some embodiments, the processes for creating the suspension are performed contemporaneously with the contacting and isolating steps, as part of a point-of-care procedure, as discussed further herein. Alternatively, obtaining the suspension may be indirect, involving only the acquisition of the suspension for use in the contacting and isolating steps, wherein the processing to create the suspension has previously been performed by another party.


In various embodiments, obtaining comprises isolating a cytokine cell suspension, comprising white blood cells or other cytokine-producing cells, from blood, adipose tissue, bone marrow aspirate or other tissue comprising cytokine-producing cells, as exemplified in Steps 110, 120 and 125 of FIG. 1. Methods may comprise obtaining a cytokine cell suspension from two, three or more tissue sources.


Obtaining a Cytokine Cell Suspension from Blood


In embodiments comprising the use of blood, the blood may be used directly in contacting the solid extraction material, as exemplified in step 140 of FIG. 1, or may be processed to provide a blood fraction, such as PRP, in a preferred embodiment. Many devices and methods for creating blood fractions are known in the art, using such means as centrifugation and filtering.


In various embodiments, methods of the present technology comprise creating PRP as the cytokine cell suspension, using centrifugation. Such methods generally comprise placing blood in a container a separator operable to separate the blood into two or more fractions, and centrifuging the separator to create a platelet-rich plasma fraction. Such devices may include a tube and a buoy disposed in the tube, wherein the buoy has a density such that the buoy reaches an equilibrium position upon centrifugation of the tissue in the tube, the equilibrium position being between a first fraction and a second fraction comprising white blood cells, the second fraction having a concentration of white blood cells greater than the concentration of white blood cells in the first fraction. Such methods further comprise centrifuging the tube so that the buoy defines an interface between the first fraction and the second fraction comprising white blood cells. The second fraction is then collected for further use in the methods of this technology.


One such device useful herein is described in U.S. Pat. No. 7,992,725, Leach et al., issued Aug. 9, 2011. Such a device is commercially available as GPS III Platelet Concentrate and Separation System, from Biomet Biologics, LLC (Warsaw, Ind., USA). The device can be used in a clinical or laboratory environment to isolate fractions from a suspension or multi-component tissue material obtained from a subject, such as blood, bone marrow aspirate, cerebrospinal fluid, adipose tissue, Isolated fractions can include platelets, platelet poor plasma, platelet rich plasma and stromal cells. The isolated fractions can each have equilibrium point or positions within the separation container that are achieved when separation has occurred. For example, a buffy coat (PRP) of whole blood may have an equilibrium position above that of the red blood cells when a sample of whole blood is separated.


The fractionation device 200 is exemplified in FIG. 2. The fractionation device 200 comprises a buoy 210 and a container wall 215. When the separation container 205 is centrifuged, the buoy perimeter 210a and the container wall 215 have clearance allowing the buoy 210 to move within the separation container 205 and a material to pass between the buoy perimeter 210a and the container wall 215. Alternatively, the buoy 210 could have an opening, such as a centrally or internally located opening or a peripheral channel running the height of the buoy, which would allow a material to move through the buoy.


The buoy 210 is carried in the separation container 205 and has a tuned density that is configured to reach a selected equilibrium position in a suspension. The buoy can have its density tuned in the range from about 1.0 g/cc to about 1.10 g/cc, such as about 1.06 g/cc. The buoy 210, according to various embodiments, can be formed to include the tuned density and can be formed of one or more materials to achieve the tuned density.


Referring to FIG. 2, a collection compartment 220 is positioned within the device 200 after a separation procedure has occurred. The collection compartment 220, defined relative to the buoy 210, is positioned at an equilibrium position of a separated or isolated middle fraction 225 in the container. The equilibrium position of a selected fraction can be defined as its position within the container relative to other fractions in the container of a separated sample or material. The equilibrium position can also be defined relative to the axis X of the buoy 210 or the container 12. The equilibrium position, however, may depend upon the amount of the sample of the amount of a selected fraction within a sample. According to the illustration in FIG. 2, the equilibrium position of the fraction 230 is above or nearer a top 235 of the device 200 than the equilibrium position of the middle fraction 225. Thus, the buoy 210 can be tuned, such as including a selected density or specific gravity, to position the collection compartment 220 relative to an equilibrium position of any selected fraction.


In some embodiments, the buoy 210 can comprise a collection port 240. The collection port 240 communicates with access port 245 and communicates with a collection space 220 above buoy upper surface 250 and can be located near the buoy perimeter 210a. In some embodiments, the collection port 240 is not carried on the buoy, but rather the collection port is a withdraw device such as a syringe that is inserted through an access port or top of the device 200.


According to various embodiments, an isolator 255, is coupled to the buoy 210. The combination of the isolator and buoy, according to various embodiments, can also be referred to as a separation assembly member. The isolator 255, for example, provides a means for creating the collection compartment 220 and comprises one or more spacers 260, 265 to position the isolator 255 apart from the buoy 210 to create the collection compartment 220. A withdraw port 270 can be carried on the isolator 255 communicating with the withdraw port 245 and the collection port 240. The spacer 260, 265 can also serve as a conduit 275 between the collection port 50 and a withdraw or withdraw port 245. The withdraw port 245 serves as a structure for withdrawing the isolated or middle fraction 225 from the collection compartment 220.


After centrifuging the device 200 containing whole blood, the first fraction or top fraction 230, can be platelet-poor-plasma, the middle fraction 225 can be platelet-rich plasma or platelet concentrate, and a bottom fraction 278 can be red blood cells. Therefore, the fractionation method further comprises withdrawing a desired fraction from the device 200. Various ports 205, 245 and 280 can be provided to allow access to any appropriate compartment of the device 200. The access ports 205, 245, 280 can be any means that allow communication from outside the separation device 200 to the device's interior, such as a Luer lock port, a septum, a valve, or other opening. Additionally, collection vent tube 285 allows removal of a fractionated suspension in the collection compartment 220 through opening 290 without the need to remove the fraction, such as plasma, above the isolator 255. Although, without a collection vent tube 285, the fraction above the isolator could be removed and the collection area could be vented to the area above the isolator.


A method for using the fractionation device 200 can begin by inputting whole blood via an access port 205. The fractionation device 200 is placed into a centrifuge and spun for a period that is appropriate for fractionating whole blood. An exemplary period can be for about five minutes to about twenty minutes at a rate of about 320 rpm to about 5000 rpm. This speed may produce a selected gravity that may be approximately 7.17×g to about 1750×g (times greater than the normal force of gravity).


Other devices that may be used to isolate platelet-rich plasma described, for example, in U.S. Pat. No. 5,585,007, Antanavich, issued Dec. 17, 1996; U.S. Pat. No. 6,398,972, Blasetti et al., issued Jun. 4, 2002; U.S. Pat. No. 6,649,072, Brandt et al., issued Nov. 18, 2003; U.S. Pat. No. 6,790,371, Dolocek, issued Sep. 14, 2004; U.S. Pat. No. 7,011,852, Sukavaneshvar et al., issued Mar. 14, 2006; U.S. Pat. No. 7,179,391, Leach et al., issued Feb. 20, 2007; U.S. Pat. No. 7,374,678, Leach et al., issued May 20, 2008; U.S. Pat. No. 7,223,346, Dorian et al., issued May 29, 2007; and U.S. Pat. No. 7,708,152, Dorian et al., issued May 4, 2010.


In addition to the GPS® Platelet Concentrate and Separation Systems, a variety of other commercially available devices may be used to isolate platelet-rich plasma, including the Magellan™ Autologous Platelet Separator System, commercially available from Medtronic, Inc. (Minneapolis, Minn., USA); SmartPReP™ commercially available from Harvest Technologies Corporation (Plymouth, Mass., USA); the AutoloGel™ Process, commercially available from Cytomedix, Inc. (Rockville, Md., USA); the GenesisCS System, commercially available from EmCyte Corporation (Fort Myers, Fla., USA); the PCCS System, commercially available from Biomet 3i, Inc. (Palm Beach Gardens, Fla., USA); and the Arthrex ACP™ Double Syringe System, commercially available from Arthrex, Inc. (Naples, Fla., USA).


Referring again to FIG. 1, blood drawn from the patient may be mixed with an anticoagulant in one or more of Steps 115, 120, 125, and 130, so as to facilitate processing. Suitable anticoagulants include heparin, citrate phosphate dextrose (CPD), ethylenediaminetetraacetic acid (EDTA), anticoagulant citrate dextrose solution (ACD), and mixtures thereof. For example, the anticoagulant may be placed in the syringe used for drawing blood from the subject, or may be mixed with the blood after it is drawn.


A liquid containing white blood cells may be prepared by admixing cells with a suitable liquid, as shown in step 125, using methods known in the art. For example, white blood cells may be isolated from whole blood by lysing red blood cells or by centrifugation of whole blood utilizing a density gradient where the white blood cells sediment to the bottom of a centrifuge tube. An example of density centrifugation includes the Ficoll-Paque™ Plus (GE Healthcare Bio-Sciences, Piscataway, N.J., USA). In some cases, a density gradient may be used to further separate mononuclear and polymorphonuclear cells. White blood cells may also be prepared from whole blood using filtration; an example includes the Acelere™ MNC Harvest System (Pall Life Sciences, Ann Arbor, Mich., USA). White blood cells can also be obtained from bone marrow. The white blood cells may be then suspended in a suitable medium, such as plasma, so as to maintain their viability.


Other methods may be used to create platelet-rich plasma or other liquid containing white blood cells. For example, whole blood can be centrifuged without using a buoy system, whole blood may be centrifuged in multiple stages, continuous-flow centrifugation can be used, and filtration can also be used. In addition, a blood component including platelet-rich plasma can be produced by separating plasma from red blood cells using a slow speed centrifugation step to prevent pelleting of the platelets. In other embodiments, the buffy coat fraction formed from centrifuged blood can be separated from remaining plasma and re-suspended to form platelet-rich plasma.


Obtaining a Cytokine Cell Suspension from Adipose Tissue


In embodiments comprising the use of adipose tissue, the adipose tissue may be used directly in contacting the solid extraction material, as exemplified in step 140 of FIG. 1, or the adipose tissue may be processed to provide isolated adipocytes in step 110. Cell fractions comprising adipose-derived stem cells are also useful in this method. In some embodiments, adipose tissue is derived from human subcutaneous fat isolated by suction assisted lipectomy or liposuction. Stromal cells may be isolated from the adipose tissue and/or tissue portions using any suitable method, including methods known in the art such as mechanical and breakdown centrifugation. Stromal cells can also be isolated using enzymatic digestion. For example, stromal cells can be isolated from lipoaspirate, treated by sonication and/or enzymatic digestion, and enriched by centrifugation. Stromal cells isolated from adipose tissue may be washed and pelleted.


For example, adipose tissue can be collected by suction-assisted tumescent liposuction inside a specialized collection container attached to suction hoses and to a liposuction cannula. The collection container can have a gauze-type grid filter that allows the tumescent fluid to pass through and retains the solid adipose tissue. After collecting the adipose tissue, the collection container is removed from the suction device and reattached to a centrifugation device. The filter unit may further contain a filter having approximately a 100 micrometer pore size. Once the collection container containing the adipose tissue is attached to the centrifugation device, the tissue is sonicated. After sonication, the entire apparatus is inserted into a centrifuge bucket and centrifuged at, for example, 300×g for 5 minutes. After centrifugation, the collection container together with the filter unit is detached and can be discarded. The pellet containing the stromal cells can then be re-suspended in biocompatible solutions, such as plasma, plasma concentrate and platelet-rich plasma.


Various methods and devices for isolating and/or fractionating adipose tissue and adipocytes include those as described by U.S. Pat. No. 7,374,678, Leach, issued May 20, 2008; U.S. Pat. No. 7,179,391 to Leach et al., issued Feb. 20, 2007; U.S. Pat. No. 7,992,725, Leach et al., issued Aug. 9, 2011; U.S. Pat. No. 7,806,276, Leach et al., issued Oct. 5, 2010; and U.S. Pat. No. 8,048,297, Leach et al., issued Nov. 1, 2011. A device, such as the GPS™ Platelet Concentrate System, commercially available from Biomet Biologics, LLC (Warsaw, Ind., USA), may be used to isolate adipocytes.


Obtaining a Liquid Containing White Blood Cells from Bone Marrow


In embodiments comprising the use of bone marrow, the marrow may be used directly in contacting the solid extraction material, as exemplified in step 140 of FIG. 1, or may be processed to provide a bone marrow concentrate, as in step 135. Many devices and methods for obtaining and concentrating bone marrow are known in the art.


An exemplary process for isolating and creating a bone marrow concentrate (cBMA) is diagrammed in FIG. 6. Generally, the method 600 may start in step 605 with obtaining a bone marrow aspirate volume. The bone marrow aspirate (BMA) may be obtained in any selected or generally known manner. For example, a selected region of bone, such as a portion near an operative procedure, may be used to obtain the bone marrow aspirate. Generally, an accessing device, such as a syringe and needle, may be used to access an intramedullary area of a selected bone. A small volume of the selected portion may be drawn from a plurality of locations to obtain an appropriate volume of BMA or selected fraction of the BMA.


Once a selected volume of the BMA is obtained in step 605, it may be separated and concentrated using a gravimetric separator. Separators among those useful herein are operable to separate a multi-component fluid that generally includes various components or constituents of varying densities that are commingled or mixed together, including those described above for separation of fractions from blood and adipose tissue. The separator may include a buoy that is of a selected density relative to BMA. Such separators include those described above for use in concentrating and isolating fractions from blood and adipose tissue, including those described in U.S. Pat. No. 7,374,678, Leach, issued May 20, 2008; U.S. Pat. No. 7,179,391 to Leach et al., issued Feb. 20, 2007; U.S. Pat. No. 7,992,725, Leach et al., issued Aug. 9, 2011; U.S. Pat. No. 7,806,276, Leach et al., issued Oct. 5, 2010; and U.S. Pat. No. 8,048,297, Leach et al., issued Nov. 1, 2011. A device, such as the GPS™ Platelet Concentrate System, commercially available from Biomet Biologics, LLC (Warsaw, Ind., USA), may be used to isolate adipocytes. Separators and methods that may be used to fractionate BMA at steps 610 and 615 are also described, for example, in U.S. Application Publication No. 2006/0278588, Woodell-May, published Dec. 14, 2006. The BMA may be positioned in a separator according to various embodiments in step 610. Once the BMA is positioned in the separator, a selected fraction of the BMA may be separated from the BMA in step 615.


Once the BMA is placed in the separator, separator is spun in a centrifuge in a range between about 1,000 and about 8,000 RPM. This produces a force between about 65 and about 4500 times greater than the force of normal gravity, as generally calculated in the art, on the separator and the BMA. At this force, the more dense material in a BMA sample is forced toward the bottom end of the tube. The separator can thus be used to remove nucleated cells from the bone marrow sample. In various embodiments, concentrated BMA has a concentration of nucleated cells that is at least 2, at least 3, at least 4, or at least 5 times the concentration of nucleated cells in BMA.


Obtaining a Cytokine Cell Suspension from Blood Clots


In other embodiments comprising the use of blood, a liquid comprising cytokine-producing cells may be trapped in a blood clot. Cell releasate can be generated from the blood clot by either compression (“squeezing”), clot disruption, or centrifugation. The blood clot can be made with or without anticoagulant and with or without exogenous thrombin by combining blood or a blood fraction with a clotting agent. Suitable clotting agents include thrombin (e.g., bovine, recombinant human, pooled human, or autologous), autologous clotting protein, and polyethylene glycol. Calcium may be in the form of a calcium salt, such as calcium chloride.


In some embodiments, the clotting agent comprises a clotting protein. A suitable clotting fraction can be obtained by a process of: loading whole blood or plasma with a calcium solution (e.g., calcium chloride in ethanol) into a blood isolation device; optionally heating the whole blood or plasma for at least about 20 minutes, at a temperature of at least about 20° C.; and isolating the clotting fraction. The isolating may be performed by centrifuging the heated whole blood or plasma. A suitable isolation device is commercially available as the Clotalyst™ Autologous Thrombin Collection System (hereinafter “Clotalyst System”), sold by Biomet Biologics LLC, Warsaw, Ind., USA.


An exemplary procedure for producing a clotting agent using a device 400 of FIG. 4 begins with injecting a reagent comprising calcium chloride and ethanol into the main chamber 405 through the first port 410. Glass beads are also placed in the main chamber 405. After the reagent has been injected, the first port 410 is closed using the first replacement cap 415. Blood with anticoagulant is injected into the main chamber 405 through the second port 420. After the blood has been injected, the second port 420 is closed using the second replacement cap 425. Optionally, the syringes and blood separation device 400 are pre-heated to a temperature of about 25° C.


The contents of the blood component separation device 400 are mixed by repeatedly inverting the device 400, e.g. about twelve times, so as to contact the blood with the glass beads. After mixing, the device is incubated The incubation process can be at a temperature and for a duration that will permit the contents of the device 400 to be heated at about 25° C. for about 15 minutes. Upon completion of the incubation period, a clotted mass of red blood cells, blood plasma, and glass beads forms at a second end 406 of the main chamber 405. After incubation is complete, the device 400 is shaken enough to dislodge and break-up any gel that may be present.


Obtaining a Cytokine Suspension Using Non-Centrifugal Methods


As noted above, the liquid containing white blood cells can be obtained by non-centrifugal means, such as by culturing. As referred to herein, a “non-centrifugal method” comprises a process for obtaining tissue fractions comprising cytokine-producing cells from tissue without use of a centrifuge. In some embodiments, methods are “non-gravimetric,” wherein, based on physical, chemical or physicochemical properties of the cells other than density, wherein the concentration of white blood cells in the fraction are higher than the concentration of white blood cells in the tissue. Such non-gravimetric methods are, in particular, distinguished from methods wherein a white blood cell fraction is created by centrifugation of whole blood or other tissue. In some embodiments, the non-centrifugal method comprises a process solely based on such properties of white blood cells other than density. Non-centrifugal methods include filtration, antibody binding, and electrophoretic methods.


For example, as discussed above, white blood cells may be prepared from whole blood, bone marrow aspirate or other tissue, using filtration. White blood cells and other cytokine-producing cells obtained from blood, bone marrow, adipose tissue or other sources may also be cultured, using methods among those known in the art. The cells may be then suspended in a suitable medium, such as plasma, so as to maintain their viability and facilitate mixing or other contact with a solid extraction material. A liquid containing the cells may also be produced by compression or disruption of blood clots, as described above.


Contacting a Cytokine Cell Suspension with an Extraction Material and Isolating a Protein Solution


In further reference to the exemplified process of FIG. 1, the cytokine cell suspension is incubated or otherwise contacted with a solid extraction material (step 140) to produce a protein-containing liquid. This liquid is then isolated (step 150) from the solid extraction material, as a Protein Solution of the present technology. Without limiting the scope, mechanism or function of the present technology, solid extraction materials useful herein concentrate cytokines or other proteins in the liquid volume of white blood cells and may, in some embodiments, activate, stimulate or otherwise increase production of cytokines, including IL-1ra. Thus, in some embodiments, methods comprising activating a cytokine cell suspension with a solid extraction material.


The solid extraction material can include various materials that provide a particular surface area to contact the cells. The solid extraction material may be a continuous material or may be discontinuous and comprise a plurality of separate particles. For example, the solid extraction material may be in the form of a plurality of beads, fibers, powder, a porous material, or a surface of a container comprising the liquid containing the cells. The solid extraction material may comprise geometric forms having various cross-sectional shapes, such as spherical, oval, or polygonal, among others. The solid extraction material can also comprise a continuous porous network, similar to a sponge, or can include a plurality of individual porous particles. The solid extraction material may also provide a larger surface area by being porous in comparison to a non-porous material.


In some embodiments, the solid extraction material includes particles having a large aspect ratio, for example, where the particles are needle-like in shape. The solid extraction material may also be formed as long fibers and may be or take a form similar to glass wool.


In some cases, the solid extraction material can comprise the internal walls of a container holding the cytokine cell suspension. For example, the solid extraction material may comprise the lumen of a syringe that contains the cytokine cell suspension. Other containers include tubes, such as centrifuge tubes, or a blood fractionation device or concentrator assembly as described elsewhere herein.


Where the solid extraction material is a continuous material, such as a porous sponge-like material, the solid extraction material can be used in an amount sufficient to absorb or adsorb or include substantially the entire liquid volume of white blood cells within the pores or interstices of the solid extraction material. Where the solid extraction material is a discontinuous material, such as a plurality of particles, the solid extraction material can be combined with the liquid containing the cells to form a slurry-like composition. The slurry can vary in consistency from paste-like, having a high-solids fraction, to a readily flowable slurry having a low-solids fraction.


The solid extraction material can provide a large surface area with which to contact the cells. However, in some cases, the solid extraction material can be further treated to increase its surface area, for example, by physically or chemically etching or eroding the surface of the solid extraction material. With respect to chemical etching, a corrosive agent can be used to modify the surface of the solid extraction material depending on the nature of the material. The modified surface may be produced by employing an alkali or an acid, for example chromosulphonic acid, in particular about 20% to about 80% in strength, preferably about 50% chromosulphonic acid. The solid extraction material can be incubated with the corrosive agent for about 5 min to about 30 min in order to chemically etch the surface and increase the surface area. The solid extraction material can then be washed to remove the corrosive agent. For example, the solid extraction material can include the internal walls of a container for holding the cytokine cell suspension where the internal walls are etched to subsequently increase the surface area in contact with the liquid.


Various polymers, metals, ceramics, and glasses can be used as the solid extraction material. In some embodiments, the solid extraction material comprises a hygroscopic material. Examples of suitable solid extraction material materials include glasses, minerals, polymers, metals, and polysaccharides. Minerals include corundum and quartz. Polymers include polystyrene, polyethylene, polyvinyl chloride, polypropylene, and polyacrylamide. Metals include titanium. Polysaccharides include dextran and agarose. A preferred solid extraction material comprises, or consists essentially of, polyacrylamide, as further described below.


The solid extraction material may comprise, for example, continuous solid extraction material of glass or a plurality of glass particles, glass wool, a continuous solid extraction material of metal such as titanium, a plurality of metal beads, metal powder, and combinations thereof. A continuous solid extraction material of metal can include a block or other three-dimensional shape formed of porous metal or metal alloys with an open cell structure. The solid extraction material may include various beads or particles of various sizes including substantially spherical beads. Beads include polystyrene beads, polyacrylamide beads, glass beads, metal (e.g., titanium) beads, or any other appropriate beads. Beads may be any size appropriate for the container and the amount of cytokine cell suspension being used. In some instances, bead sizes can range from about 0.001 millimeters to about 3 millimeters in diameter. Where the bead size is sufficiently small, the beads can appear more like a powder.


Polyacrylamide beads used as the solid extraction material can be formed by polymerizing acrylamide monomer using controlled and standardized protocols as known in the art to produce relatively uniform beads formed of polyacrylamide gel. In general, polyacrylamide is formed by polymerizing acrylamide with a suitable bifunctional crosslinking agent, most commonly N,N′-methylenebisacrylamide (bisacrylamide). Gel polymerization is usually initiated with ammonium persulfate and the reaction rate is accelerated by the addition of a catalyst, such as N,N,N′,N′-tetramethylethylenediamine (TEMED). In various embodiments, polyacrylamide beads comprise 0.5 micromole of carboxyl groups per milliliter of beads, imparting a slight anionic character (negative charge). The beads are also typically resistant to changes in pH, and are stable in many aqueous and organic solutions. By adjusting the total acrylamide concentration, the polyacrylamide gel can be formed in a wide range of pore sizes. Moreover, the polyacrylamide beads can be formed in many sizes and can have relatively uniform size distributions. Bead size may range from several micrometers in diameter to several millimeters in diameter. For example, various types of Bio-Gel™ P polyacrylamide gel beads (Bio-Rad Laboratories, Hercules, Calif., USA) have particle sizes ranging from less than about 45 μm up to about 180 μm. Polyacrylamide beads are also available from SNF Floerger (Riceboro, Ga., USA), Pierce Biotechnology, Inc. (Rockford, Ill., USA), and Polymers, Inc. (Fayetteville, Ark., USA).


Once polymerized, polyacrylamide beads can be dried and stored in a powder-like form. The dry beads are insoluble in water but can swell considerably upon being rehydrated. Rehydration returns the polyacrylamide beads to a gel consistency that can be from about two to about three times the dry state size. Thus, dry polyacrylamide beads (i.e., desiccating polyacrylamide beads) may be used to absorb a portion of a liquid volume, including solutes smaller than the bead pore size, and can serve to concentrate IL-1ra and other proteins produced by the white blood cells. For example, combining dry polyacrylamide beads with the blood and/or platelet-rich plasma activates production of IL-1ra by the white blood cells and also reduces the total liquid volume as the dry beads rehydrate and swell.


Without limiting the scope, mechanism or function of the present technology, it has been discovered that surface contact with the solid extraction material can activate the cells and the solid extraction material can, in some cases, assist in the separation and concentration of the resulting Protein Solution rich in cytokines, including IL-1ra. For example, in the case of a porous solid extraction material, a portion of the liquid comprising the cells can enter the pores and remain therein. Cells in the liquid may contact this additional surface area. In some embodiments, the pores are too small for the cells to enter, but a portion of the liquid can enter the pores. Liquid can be removed from the solid extraction material and pores by centrifuging, for example.


The solid extraction material is preferably sterilized, using techniques among known in the art, in order to prevent contamination of the cytokine cell suspension. For example, heat and pressure sterilization methods, such as autoclaving, may be used depending on the particular composition of the solid extraction material. Alternative methods, such as chemical sterilization or irradiation, can be used where the solid extraction material may be adversely affected by the autoclaving process.


In some embodiments, the cytokine cell suspension is incubated with solid extraction material for a time effective to remove a portion of the liquid. The incubation may be carried out over a period from about 30 seconds to about 72 hours and may be carried out at a temperature from about 20° C. to about 41° C. For example, the incubation may be 24 hours or less, 10 hours or less, 5 hours or less, 2 hours or less, 1 hour or less, 30 minutes or less, 15 minutes or less 10 minutes or less, 5 minutes or less, 4 minutes or less, 3, minutes or less, or 2 minutes or less. Incubation may be at least about 15 seconds, at least about 30 seconds, at least about 1 minutes, at least about 90 seconds, at least about 2 minutes, at least about 10 minutes, or at least about 30 minutes. In some embodiments, incubations from about 1 minute to about 3 minutes. In some embodiments, the incubation is conducted at about 37° C. In some embodiments the liquid is not incubated, but is contacted with the solid extraction material for only so long as necessary to perform subsequent processing. The contacting may occur at ambient conditions, e.g., at a temperature of about 20-25° C.


In some embodiments, the cytokine cell suspension and the solid extraction material are agitated to more thoroughly mix these components during contact. The agitation may be accomplished by inverting, shaking, rocking, stirring, or vortexing the liquid and solid extraction material. Agitation may increase contact of the cells within the liquid with the solid extraction material. Agitation may be performed once, repeated multiple times, repeated periodically, or may be continuous. The liquid comprising the cells and the solid extraction material may also be agitated while the liquid is stimulated with the electromagnetic field. Additional aspects and features relating to producing protein-rich solutions using polyacrylamide beads and other solid extraction materials are described in: U.S. Patent Application Publication No. 2009/0220482, Higgins et al., published Sep. 3, 2009; U.S. Patent Application Publication No. 2010/0055087, Higgins et al., published Mar. 4, 2010; U.S. Patent Application Publication 2011/0052561, Hoeppner, published Mar. 3, 2011; International Application Publication 2012/030593, Higgins et al., published Mar. 8, 2012; and U.S. Patent Application Publication 2012/0172836, Higgins et al., published Jul. 5, 2012. Compositions and methods useful in aspects of the present technology are also described in the following applications filed concurrently with this disclosure: U.S. patent application Ser. No. 13/841,083, Landrigan, et al., Treatment of Inflammatory Respiratory Disease Using Protein Solutions; U.S. patent application Ser. No. 13/837,005, Woodell-May et al., Methods and Acellular Compositions for Treating Inflammatory Disorders; U.S. patent application Ser. No. 13/837,480, O' Shaughnessey, et al., Treatment of Pain Using Protein Solutions; U.S. patent application Ser. No. 13/839,280, Leach et al., Methods for Making Cytokine Compositions from Tissue Using Non-Centrifugal Methods; U.S. patent application Ser. No. 13/840,129, Matusuka, et al., Treatment of Collagen Defects Using Protein Solutions; and U.S. patent application Ser. No. 13/841,103, Landrigan, et al., Treatment of Peripheral Vascular Disease Using Protein Solutions, all of which are incorporated by reference herein.


Contacting of the liquid containing white blood cells with the solid extraction material may be performed using a suitable container or other apparatus to affect the contact. Contacting may be performed in a continuous process wherein a flow of the liquid is passed over or through the solid extraction material, or the liquid and solid extraction material may be contained in a vessel. As discussed above, the vessel may comprise the solid extraction material, or may merely serve as a container holding the beads or other forms of the material. Containers useful in the present technology include those known in the art, such as the Plasmax™ Plus Plasma Concentrator, commercially available from Biomet Biologics, LLC (Warsaw, Ind., USA) and may include those devices and methods of use as described in U.S. Pat. No. 7,553,413, Dorian et al., issued Jun. 30, 2009; and U.S. Pat. No. 7,694,828, Swift et al., issued Apr. 13, 2010.


Such a device is shown in FIGS. 3A and 3B, for exemplary use with a polyacrylamide gel bead solid extraction material. The device 300 has an upper chamber 305 and a lower chamber 310. The upper chamber 305 has an end wall 315 through which the agitator stem 320 of a gel bead agitator 325 extends. The device 300 also has an inlet port 330 that extends through the end wall 315 and into the upper chamber 305. The device 300 also includes an outlet port 335 that communicates with a plasma concentrate conduit 340. The floor of upper chamber 305 includes a filter 345, the upper surface of which supports desiccated concentrating polyacrylamide beads 350.


During use, a fluid 355 containing white blood cells and, optionally, platelets is injected to the upper chamber 305 via the inlet port 330 and mixed with the polyacrylamide beads 350. The fluid 355 and polyacrylamide beads 350 may be mixed by rotating the agitator stem 320 and the gel bead agitator 325, to help mix the fluid 355 and beads 350. The mixed fluid 355 and polyacrylamide beads 350 are then incubated for the desired time at the desired temperature. The device 300 is then centrifuged so that liquid passes to the lower chamber 310 while the polyacrylamide beads 350 are retained by a filter 345, thereby separating the polyacrylamide beads 350 from the resulting solution 360 of IL-1ra and other proteins that collects in the lower chamber 310. The solution 360 may be removed from the device via outlet port 335.


In some embodiments, a Protein Solution can be made in a process wherein a liquid containing white blood cells is isolated from a tissue and then contacted with a solid extraction material in a continuous process. Referring again to FIG. 1, in some embodiments the isolating 110, 120, 135 and contacting 140 are performed using a single apparatus, referred to herein as a single separation and concentration device (“S/C device”). One such device is described in U.S. patent application Ser. No. 13/434,245, O'Connell, filed Mar. 29, 2012.


The S/C device comprises a separation region, a first concentration region, a second concentration region, a buoy system, an inlet port, a check valve, a first withdrawal port and a second withdrawal port. FIG. 5 shows an S/C device 500 capable of generating an anti-inflammatory cytokine composition from whole blood. For example, the method may start with obtaining a volume of whole blood, which is filled into a separation region 505 of the S/C device 500 by injecting through the inlet port 510. A buoy system 515 is located within the separation region 505. The buoy system comprises a first buoy member 520, a second buoy member 525, and a third buoy member 530 that couples the first buoy member 520 to the second buoy member 525. A space between the first and second buoy members 520, 525 defines a buoy separation region 535. A density of each buoy member can be selected depending on what blood fraction is desired as a result of a separation. The buoy system 515 can include a selected buoy system, such as the buoy system generally used in the GPS® II or GPS® III gravity platelet separation system sold by Biomet Biologics, LLC. (Warsaw, Ind., USA). Buoy systems are disclosed in U.S. Pat. No. 7,845,499 and U.S. Pat. No. 7,806,276, and U.S. Pat. No. 7,992,725.


A method for obtaining a Protein Solution comprises spinning the S/C device 500 by centrifugation. Centrifugal forces allow the buoy system 515 to move through the whole blood, resulting in a fraction of the whole blood to be located in the buoy separation region 535. For example, this fraction may comprise platelet-rich plasma. With a use of a withdrawal syringe, the selected fraction can be removed from the collection volume 535 through the third buoy member 530 that defines a removal passage 540 that is connected with collection passages 545. A connection elbow 550 can interconnect with the removal passage 540 to allow a vacuum to be formed through the connection elbow 550, the collection passage 540, and the buoy collection passages 545. A collection tube 555 can interconnect the connection elbow 550 with a withdrawal elbow 560 that extends from a wall 565 that can be a bottom wall of concentration region 570. A second withdrawal tube 575 can be first connected with a check valve assembly 580 and a first withdrawal port 585. The first withdrawal port 585 can be connected with the withdrawal syringe with a Luer lock type connection or other appropriate connection.


The check valve assembly 580 ensures the fraction being removed flows in one direction and prevents the fraction being removed from reentering the second withdrawal tube 575. Furthermore, when material is pushed back into the check valve assembly 580 from the first withdrawal port 585, such that material will enter the concentration region 570, a disc within the check valve assembly 580 can flex down towards the second withdrawal tube 575 and close an opening and thereby open a second opening within the check valve assembly 580. The second opening allows the fraction to be pushed into the concentration region 570.


Therefore, the blood fraction is then re-injected through the first withdrawal port 285, through the check valve assembly 580, and into an upper volume 588 of the concentration region 570. Polyacrylamide beads 590 are added to the blood fraction in the upper volume 588 and the blood fraction and the polyacrylamide beads 590 can be mixed by shaking. Optionally, the blood fraction and the beads 590 can be incubated for a selected period of time before proceeding with the method.


The method comprises a second step of spinning by centrifugation. During the second centrifugation, the anti-inflammatory cytokine composition is separated from the beads 590 by being forced through a filter 592 and into a lower concentration region 595 of the concentration region 570. The Protein Solution can be withdrawn through a third withdrawal tube 596 and out a second withdrawal port 598 by use of a second withdrawal syringe. Again, the syringe can be connected to the second withdrawal port by a Luer® lock type connection.


Referring again to FIG. 1, following contacting the liquid with the solid extraction materials, a Protein Solution is isolated, as indicated at step 150. Isolation may be accomplished by drawing off at least a portion of the liquid volume and leaving the beads. In some cases, the extraction material may be sedimented by centrifugation prior to drawing off the Protein Solution. Isolation may also be performed by filtration, where the material is retained by a filter and the Protein Solution passes through the filter using centrifugal force or by using vacuum, for example. If the incubation with extraction material utilizes dry polyacrylamide beads, the liquid volume may be reduced as the beads swell upon rehydration, thereby concentrating the resulting Protein Solution. To maintain the increased concentration, care should be taken in the isolation step so as to avoid compressing the beads or drawing liquid out from the swollen beads. For example, high centrifugal force or high vacuum may collapse the beads and/or draw liquid out of the internal volume of the beads.


Optional Electromagnetic Stimulation


The cytokine cell suspension can be stimulated with an electromagnetic field, before or during the contacting of the liquid with a solid extraction material. Thus, in some embodiments, stimulation of the liquid comprising the cells can be performed prior to contacting the liquid and the solid extraction material. However, it is preferred that at least a portion of the contacting step and at least a portion of the stimulating step overlap in time such that the liquid comprising the cells is concurrently in contact with the solid extraction material and stimulated with the electromagnetic field.


Stimulating the cytokine cell suspension with an electromagnetic field may involve various forms of electromagnetic stimulation, such as a pulsed electromagnetic field or a capacitively coupled electromagnetic field. In some embodiments, the liquid is stimulated using a power source coupled to a stimulation coil. The current passing through the coil produces a pulsing magnetic field which induces in the liquid a pulsing electric field. The coil may partially surround the liquid as it is held within a container, such as a tube or syringe. The coil may be integrated into to the container holding the cytokine cell suspension or may be removable. For example, a plastic tube can be formed with an integrated coil or the coil can be temporarily coupled to the container or placed within the container; for example, the tube can be configured so that the coil can be snapped onto the container. The power source can be coupled to the coil as needed to perform the stimulating step.


Stimulation of the liquid with an electromagnetic field may also include placing at least two electrodes across the liquid. Electrical energy may then be applied to the electrodes so as to capacitively couple the electrodes and generate the electromagnetic field there between. The electromagnetic field is therefore able to pass through the liquid so as to increase the rate and/or amount of cytokine production. In other embodiments, electrodes can be used to produce a direct current or one or more coils can be used to produce a pulsed electromagnetic field.


The strength of the electromagnetic field during stimulation can be at least about 0.5 microvolts per centimeter, whether produced by direct current, capacitively coupled current, or pulsed electromagnetic field. In the case of a direct current electrode, the amplitude of the current can be from about 1 to about 200 microamperes, and in some embodiments, the amplitude may be from about 20 to about 100 microamperes. In still further embodiments, the current may be about 20, about 60, or about 100 microamperes. It should be understood, however, that the amplitude of the current may be of other suitable magnitudes.


The electromagnetic field applied during the stimulating step may be constant or vary over time. For example, a sinusoidal time varying electromagnetic field can be applied using the electrodes placed across the liquid. Such a sinusoidal time varying electromagnetic field can have a peak voltage across the electrodes from about 1 volt to about 10 volts, and in some embodiments, the peak voltage can be about 5 volts. The corresponding electric field produced can have an amplitude of from about 0.1 millivolt per centimeter (mV/cm) to about 100 mV/cm, and in some embodiments can be about 20 mV/cm. The sinusoidal time varying electric field may have a frequency of from about 1,000 Hz to about 200,000 Hz, and in some embodiments the frequency may be about 60,000 Hz.


The electromagnetic field applied to the liquid may also be a pulsed electromagnetic field. The pulsed electromagnetic field can be induced using an external coil and a pulse generator. In this regard, a pulsed electromagnetic field may have a pulse duration of from about 10 microseconds per pulse to about 2000 microseconds per pulse. The pulse duration in one embodiment can be about 225 microseconds. The pulses may include electromagnetic bursts, in which a burst can comprise from 1 pulse to about 200 pulses. Alternatively, the electromagnetic field may have bursts that comprise from about 10 pulses to about 30 pulses. In this regard, in one embodiment each burst may comprise about 20 pulses.


The frequency at which bursts in the pulsed electromagnetic are applied may vary. In this regard, bursts can be repeated at a frequency of from about 1 Hz to about 100 Hz in some embodiments, and can be repeated at a frequency of about 10 Hz to about 20 Hz in other embodiments. Furthermore, bursts can repeat at a frequency of about 1.5 Hz, about 15 Hz or about 76 Hz. A burst can have a duration from about 10 microseconds up to about 40,000 microseconds. In this regard, a burst can have a duration of about 4.5 milliseconds.


Suitable devices for generating a capacitively coupled electromagnetic field include SpinalPak® spinal stimulator (EBI, L.P., Parsippany, N.J.) or a DC stimulation device such as an SpF® XL IIb spinal fusion stimulator (EBI, L.P., Parsippany, N.J.). Pulsed electromagnetic fields can be produced using various known methods and apparatuses, such as using a single coil or a pair of Helmholtz coils. For example, a suitable apparatus includes the EBI Bone Healing System® Model 2001 (EBI, L.P., Parsippany, N.J.) and the BTBS stimulation coil. With respect to direct current, an electric field may be generated using any known device for generating a direct current electric field, such as for example, the Osteogen™ implantable bone growth stimulator (EBI, L.P., Parsippany, N.J.). Other suitable devices for generating electromagnetic fields may be used.


Electromagnetic stimulation of the cytokine cell suspension can be continued and/or repeated as desired with respect to contacting the liquid and the solid extraction material. It should be understood, however, that the step of stimulating the liquid with an electromagnetic field includes fields other than, or in addition to, electric or electromagnetic fields associated with ambient conditions (such the electromagnetic fields generated by casual exposure to radios, telephones, desktop computers or similar devices).


In some embodiments, both the contacting and stimulating steps as shown in FIG. 1 are performed in less than about 1 hour. The contacting and stimulating steps can also be performed at temperatures ranging from about 20° C. to about 37° C. In a preferred embodiment, the temperature of the cytokine cell suspension is kept at about 37° C. during the contacting and stimulating steps. One or both of the contacting and stimulating steps are typically performed ex vivo.


Other Methods for Forming Protein Solutions


The present technology provides other methods for forming components of Protein Solutions, such as the admixture of proteins and other components and the isolation and concentration of proteins and components without using solid extraction materials. For example, Protein Solutions of the present technology can be made by mixing of proteins made by such methods with components or solutions made by tissue isolation from allogeneic sources and processing with solid extraction materials, as described above.


For example, various methods provide acellular or substantially acellular Protein Solutions, comprising one or more proteins as described above. Without limiting the scope, mechanism or function of the present technology, such acellular anti-inflammatory cytokine compositions may offer advantages in certain applications, insofar as they may not create an immunogenic response in subjects to whom they are administered.


In particular, by way of example, a Protein Solution may comprise interleukin-1 receptor antagonist (IL-1ra) that is synthetic or recombinant, or isolated from autologous, allogeneic or xenogeneic blood or other biologic sources, aside from the methods described above. For example, Kineret™ (anakinra) is a recombinant, non-glycosylated form of IL-1ra, sold by Amgen Manufacturing, Ltd. (Thousand Oaks, Calif.). Various recombinant interleukin-1 inhibitors and methods of treatment are described in U.S. Pat. No. 6,599,873, Sommer et al., issued Jul. 29, 2003; U.S. Pat. No. 5,075,222, Hannum et al., issued Dec. 24, 1991; and U.S. Application Publication No. 2005/0197293, Mellis et al., published Sep. 8, 2005. In addition, methods for producing IL-1ra from body fluids, including the use of autologous fluids, are described in U.S. Pat. No. 6,623,472, Reinecke et al., issued Sep. 23, 2003; U.S. Pat. No. 6,713,246, Reinecke et al., issued Mar. 30, 2004; and U.S. Pat. No. 6,759,188, Reinecke et al., issued Jul. 6, 2004. When an allogeneic anti-inflammatory cytokine composition is to be generated, multiple sources of IL-1ra from multiple subjects may be pooled together.


More generally, methods for making acellular Protein Solutions can comprise culturing cells in a cell culture that either naturally produce anti-inflammatory cytokines, such as IL-1ra, or cells that are engineered to produce such cytokines. Non-limiting examples of cells that naturally produce anti-inflammatory cytokines include adipose tissue cells, adipocytes, adipose-derived stem cells, stromal cells, bone marrow cells, mesenchymal stem cells, and blood cells.


In various embodiments, cell lines can be engineered to overproduce an anti-inflammatory cytokine. Non-limiting examples of anti-inflammatory cytokines include VEGF, TNF-α, IL-1ra, sTNF-RI, sTNF-RII, PGDF-AB, PDGF-BB, IGF-I, EGF, TGF-β1, sIL-1RII, and HGF. Stable eukaryotic cell lines can be generated that overexpress an anti-inflammatory cytokine by transfecting eukaryotic cells, such as mammalian cells, with recombinant DNA comprising a gene encoding an anti-inflammatory cytokine and a selectable marker. Alternatively, prokaryotes and yeast can be engineered to overexpress an anti-inflammatory cytokine by transformation with recombinant DNA comprising a gene encoding an anti-inflammatory cytokine and a selectable marker. Transformations and transfections can be performed with recombinant DNA molecules comprising a DNA sequencing encoding an anti-inflammatory cytokine, such as IL-1ra, and a selectable marker. Eukaryotic and prokaryotic cells can be engineered to overexpress the anti-inflammatory cytokine constitutively or by induction. Methods for expressing anti-inflammatory cytokines, such as IL-1ra, sTNF-RI, and sTNF-RII, and sIL1-RII in eukaryotic and prokaryotic cells are described in U.S. Pat. No. 6,337,072, Ford et al., issued Jan. 8, 2002; and U.S. Application Publication No. 2001/0053764, Sims et al., published Dec. 20, 2001.


When a IL-1ra gene is transcribed in humans, the mRNA can be spliced into four variants, resulting in four isoforms of translated IL-1ra. SEQ ID NOs: 1, 3, 5, and 7 are the cDNAs for IL-1ra isoforms 1-4 respectively, and SEQ ID NOs: 2, 4, 6, and 8 are the amino acid sequences of IL-1ra isoforms 1-4 respectively. Collectively, the IL-1ra isoforms are referred to as “IL-1ra.” SEQ ID NO: 9 is the cDNA sequence for sTNF-RI and SEQ ID NO:10 is the amino acid sequence for sTNF-RI. SEQ ID NO:11 is the cDNA sequence for sTNF-RII and SEQ ID NO:12 is the amino acid sequence for sTNF-RII. SEQ ID NO:13 is the cDNA sequence for sIL-1RI and SEQ ID NO:14 is the amino acid sequence for sIL-1RI. SEQ ID NOs 15 and 17 are the cDNAs for sIL-1RIIv1 and sIL-1RIIv3 respectively, and SEQ ID NOs:16 and 18 are the amino acid sequences for sIL-1RIIv1 and sIL-1RIIv3 respectively. The cDNA sequence for IL-1RIIv2 is a non-coding sequence; therefore, it is not included.


To express either IL-1ra, sTNF-RI, or sTNF-RII (generically referred to as a “protein of interest”) in a prokaryotic culture, for example in a particular bacteria, a cDNA sequence (SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, or 17) is cloned into an expression vector suitable for the bacteria. The expression vector should comprise a strong promoter, and a selectable marker, such as antibiotic resistance. Non-limiting examples of antibiotics capable of killing bacteria cells include ampicillin, tetracycline, kanamycin, and chloramphenicol. The expression vector should further comprise elements that result in constitutive or inducible expression of the protein of interest. Optionally, a DNA sequence corresponding to a tag functionally coupled to the protein of interest that allows for identification and purification of the protein can be present in the vector adjacent to the gene for the protein of interest. For example, an N or C-terminal His tag can be used to detect proteins with anti-His antibodies, and they allow for purification on nickel columns. When the expression vector comprising a gene expressing a protein of interest is prepared, a bacteria cell, for example E. coli, can be transformed with the expression vector. The selectable marker ensures that only cells transformed with the vector will survive in LB broth supplemented with an antibiotic corresponding to the selectable marker. The bacteria can then be grown in LB broth supplemented with the antibiotic for expression and purification. Expression vectors, methods for cloning a protein of interest into an expression vector, methods for transforming prokaryotic cells, methods for expressing protein from transformed prokaryotic cells, and protein purification methods are commonly known by those with ordinary skill in the art.


To express a protein of interest in a eukaryotic culture, for example in mammalian cells, a cDNA sequence (SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, or 17) is cloned into an expression vector suitable for a particular mammalian cell. The expression vector should comprise a strong promoter, and a selectable marker, such as antibiotic resistance. Non-limiting examples of antibiotics capable of killing mammalian cells include geneticin and gentamicin. The expression vector should further comprise elements that result in constitutive or inducible expression of the protein of interest. Optionally, a DNA sequence corresponding to a tag functionally coupled to the protein of interest that allows for identification and purification of the protein can be present in the vector adjacent to the gene for the protein of interest. When the expression vector comprising a gene expressing a protein of interest is prepared, a mammalian cell, such as a human cell, can be transfected with the expression vector. Transfected cells can be grown in a cell culture medium supplemented with an antibiotic corresponding to the selectable marker. The presence of the antibiotic allows for the isolation of stable cell lines. Stable cell lines can then be grown in cell culture medium supplemented with antibiotic for expression and purification. Expression vectors, methods for cloning a protein of interest into an expression vector, methods for transfecting eukaryotic cells and developing stable cell lines, methods for expressing protein from transfected eukaryotic cells, and protein purification methods are commonly known by those with ordinary skill in the art.


Alternatively, eukaryotic cells that have not been genetically altered by DNA transfection can be cultured. The eukaryotic cells can be primary cultures, i.e. cells grown directly from a eukaryotic donor, such as a human, or the eukaryotic cells can be established cell lines. Many established cell lines are available commercially from American Type Culture Collection, Inc. (Manassas, Va., USA). The cells can be grown with or an exogenous signal, such as a recombinant protein. Eukaryotic cells are often cultured in culture flasks with cell culture medium. The cell culture medium can be recovered from the flasks, and centrifuged to remove any non-adherent cells.


A cell culture can be a monolayer culture, a non-adherent culture, or a bioreactor. A monolayer culture comprises anchorage-dependent cells that are cultured on a suitable substrate that allows cell adhesion and spreading, such as cell culture flasks and cell culture dishes. A non-adherent culture comprises cells that are maintained in a suspension. Suitable cells are either not anchorage-dependent, or they are anchorage-dependent cells that have been adapted for culture in a suspension. Many cell lines, for example many insect cells, can be grown in either a monolayer or a suspension. A bioreactor is a device that can support a biologically active environment in which chemical processes are carried out and/or biochemically active substances are derived. Bioreactors can include suspended or immobilized cells. Monolayer cultures, non-adherent cultures, and bioreactors can be maintained by methods commonly used in the art.


In some embodiments, the cell culture is subjected to an electromagnetic field, so as to stimulate the production of one or more proteins. Stimulating the culture with an electromagnetic field may involve various forms of electromagnetic stimulation, such as a pulsed electromagnetic field or a capacitively coupled electromagnetic field. Methods and conditions for stimulation include those discussed above.


Cell cultures can either release anti-inflammatory cytokines into culture medium naturally, or the cultures can be induced to release the anti-inflammatory cytokines into the culture medium. The culture medium can be isolated by aspiration, centrifugation or filtration for use in forming the acellular anti-inflammatory cytokine composition.


In some embodiments, an anti-inflammatory cytokine is isolated from urine, for use in producing a Protein Solution of the present technology. Proteins can be isolated from urine by methods among those known in the art. One such method is employed in the ProteoSpin™ Urine Protein Concentration Maxi Kit sold by Norgen Biotek Corp. (Thorold, Ontario, Canada). This kit utilizes an ion exchange resin integrated into a spin column. Briefly, a urine sample is obtained and its pH adjusted to 3.5. The urine is then transferred to a spin column containing the ion exchange resin, which is placed in a collection tube. The column is then centrifuged, wherein the proteins attach to the resin, and the remaining fluids and salts flow into the collection tube and are discarded. The proteins are then washed by applying supplied column activation and wash buffer followed by centrifugation. The flow through is discarded and the wash procedure is repeated. An elution buffer (10 mM sodium phosphate, pH 12.5) is added to the column and neutralizer is added to an elution tube. The spin column containing the elution buffer is placed in the elution tube and centrifuged, whereby the proteins are eluted and captured in the elution tube containing neutralizer.


Therapeutic Compositions

The present technology also provides compositions comprising a Protein Solution and a second component comprising active materials, physiological carriers, and combinations thereof. In some embodiments, compositions comprise a safe and effective amount of the Protein Solution and a safe and effective amount of a second active. A “safe and effective” amount of a component is an amount that is sufficient to have the desired therapeutic effect in the human or other mammalian subject, without undue adverse side effects (such as toxicity, irritation, or allergic response), commensurate with a reasonable benefit/risk ratio when used in the manner of this technology. The specific safe and effective amount of the component will, obviously, vary with such factors as the particular condition being treated, the physical condition of the patient, the nature of concurrent therapy (if any), the specific components used, the specific route of administration and dosage form, the carrier (if any) employed, and the desired dosage regimen.


Active materials among those useful herein include biologics and pharmaceutical actives. Biologics include blood fractions, such as PRP, blood products, and concentrated bone marrow aspirate (cBMA).


Accordingly, in some embodiments, the present technology provides compositions comprising a safe and effective amount of a Protein Solution and a safe and effective amount of cBMA. cBMA can include hematopoietic, stem cells, stromal stem cells, mesenchymal stem cells, endothelial progenitor cells, red blood cells, white blood cells, fibroblasts, reticulocytes, adipose cells, or endothelial cells. As described above, the Protein Solution may be made using bone marrow aspirate as a cytokine containing tissue. However, a therapeutic composition may additionally comprise cBMA with Protein Solution. In one embodiment, a therapeutic composition comprises a Protein Solution and cBMA in an Protein Solution:cBMA ratio of about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9 or about 1:10. Alternatively, the Protein Solution:cBMA ratio can be about 2:1, about 3:1, about 4:1, about 5:1, about 6:1, about 7:1, about 8:1, about 9:1 or about 10:1. The cBMA and Protein Solution may also be produced simultaneously. Thus, in reference to FIG. 1 and the processes described above, bone marrow aspirate may be added to the whole blood obtained in step 115, prior to or during the contacting with a solid extraction material in step 140; such a process involves operation of both steps 115 and 130. For example, bone marrow aspirate may be added to whole blood prior or during isolation of platelet-rich plasma in step 120. Such methods include those described in U.S. Application Publication No. 2006/0278588, Woodell-May, published Dec. 14, 2006.


Pharmaceutical actives among those useful herein include herein include organic molecules, proteins, peptides, peptidomimetics, nucleic acids, nucleoproteins, antisense molecules, polysaccharides, glycoproteins, lipoproteins, carbohydrates and polysaccharides, botanical extracts, and synthetic and biologically engineered analogs thereof, living cells (other than white blood cells stromal cells) such as chondrocytes, bone marrow cells, viruses and virus particles, natural extracts, and combinations thereof. Specific non-limiting examples of bioactive materials include hormones, antibiotics and other anti-infective agents, hematopoietics, thrombopoietics, antiviral agents, antitumor agents (chemotherapeutic agents), antipyretics, analgesics, anti-inflammatory agents, antiallergy agents, vasodilators, cytokines, growth factors, gene regulators, vitamins, minerals and other nutritionals, nutraceuticals and combinations thereof. In particular, actives include bronchodilators (such as albuterol, levabuterol, irbuterol, ipatropium, salmeterol, and formoterol), glucocortico steroids (such as mometasone, fluticasone, budesonide, and beclomethosone), antibiotics, antivirals, and combinations thereof. In some embodiments, compositions may comprise growth factors in addition to those present in the Protein Solution, such Platelet-Derived Growth Factor (PDGF), Transforming Growth Factor Beta (TGF-β), Insulin-Like Growth Factor (IGF), Fibroblast Growth Factor (FGF), Epidermal Growth Factor (EGF), Vascular Endothelial Growth Factor (VEGF), and Bone Morphogenetic Proteins (BMPs).


The compositions may comprise a carrier material, in addition to any liquid comprising the Protein Solution. It should be understood that in various embodiments of the present technology, methods of treatment employ the Protein Solution as comprised and made above, without further carrier, by direct injection or other application to the site of treatment. However, in other embodiments, an additional carrier material may be used for such reasons as for ease of administration, to facilitate administration using a particular delivery device, enhancing activity, an increasing the length of time the Protein Solution remains at the site of administration. Carriers among those useful herein include saline, hyaluronic acid, collagen, buffers (such as Hank's Buffer), cell culture media, blood products (such as PRP and platelet poor plasma), and mixtures thereof.


Protein Solutions, and compositions comprising Protein Solutions may be sterilized prior to administration, by any suitable method. For example, a Protein Solution may be sterilized by including a sterile filter to process the product made by the processes described above. In some embodiments, an antibiotic may be included in the solid extraction material during the contacting step described above, or may be added at one or more of the various steps in the methods and treatments described herein. Alternatively, or in addition, the Protein Solution may be produced asceptically.


Protein Solutions and compositions comprising Protein Solutions may also be lyophilized (freeze drying, or cryodesiccation) after production, using methods among those known in the art. Thus, as depicted in FIG. 1, the Protein Solution can be lyophilized 160 after it is isolated from the solid extraction material. When freeze dried, the anti-inflammatory cytokine composition can be hydrated with a suitable media 170, at a time before administration or at a time of administration. Hydration may be accomplished by mixing the composition with a solution including saline, buffers, blood, blood fractions, bone marrow aspirate, concentrated bone marrow aspirate, and combinations thereof.


The present technology provides compositions comprising components derived from blood or other tissue that are suitable for allogeneic administration. In particular, such compositions may comprise proteins and other components isolated from a mammalian subject, or a plurality of mammalian subjects, other than the subject to whom the composition is to be administered in a method of this technology. In some embodiments, Protein Solutions comprise components isolated from the subject to be treated and components isolated from one or more subjects other than the subject to be treated.


Methods of the present technology include those wherein a Protein Solution or components thereof are isolated from blood obtained from mammalian blood donors other than the subject to be treated, such as may be obtained from a blood bank. In some embodiments, the blood may typed according to the ABO blood typing system so as to assess compatibility with the blood of the donor. In other embodiments, the blood is not typed. The blood may be obtained from multiple donors, and mixed (“pooled”) as part of the obtaining steps discussed above.


In further reference to FIG. 1, compositions made by contacting a cytokine cell suspension with a solid extraction material may be made suitable for allogeneic administration by freeze drying, as depicted in step 160, after isolation of the Protein Solution from the solid extraction material.


In some embodiments, the composition can be processed to remove white blood cells (or other cytokine-producing cells) present in the Protein Solution composition after contacting step 140. Methods for removing white blood cells include those known in the art, including filtering, clotting, and gravimetric methods. Isolating the blood fraction comprising plasma and removing white blood cells may performed essentially simultaneously. Thus, the present technology provides methods for making a non-immunogenic anti-inflammatory cytokine composition, comprising:

    • (a) obtaining a cytokine cell suspension from a mammalian donor;
    • (b) contacting the suspension with solid extraction material to generate a composition rich in cytokines, such as interleukin-1 receptor antagonist; and
    • (c) performing one or both of:
      • (i) removing cells from the composition; and
      • (ii) freezing (such as by freeze drying) the composition to produce the non-immunogenic anti-inflammatory cytokine composition.


        Thus, the present technology provides anti-inflammatory compositions comprising a lyophilized activated plasma composition, wherein:
    • (a) the plasma is obtained from a plurality of mammalian subjects;
    • (b) the plasma is activated by contacting the plasma with a solid extraction material; and
    • (c) the plasma is substantially free of white blood cells.


In other embodiments, the Protein Solution is not freeze dried after contacting, and cells are not removed. In such embodiments, a crypreservative storage solution may be added to the composition, to provide stability for subsequent storage at reduced temperatures. Suitable storage solutions include those in the art, such as glycerol and dimethylsulfoxide (DMSO). The composition may be stored at reduced temperatures, such as from about 1° C. to about 6° C. In some embodiments, the composition is stored under liquid nitrogen, at about −80° C. Preferably, the cryopreservative storage solution is removed from the Protein Solution prior to administration to a mammalian subject. Removal of the storage solution may be performed by methods including those known in the art for processing stored blood comprising cryopreservatives. Washing may be performed using a wash solution, such as saline. In such embodiments, the blood type of the subject to be treated may be matched to the blood type of the donor from whom the cytokine cell suspension was obtained.


Methods of Treatment

The present technology provides methods for the treatment of an inflammatory disorder or other disorder mediated by IL1-ra in a human or other mammalian subject, comprising administration of a Protein Solution of the present technology the subject. Methods include treating an inflammatory disorder using a Protein Solution that is made by contacting a cytokine cell suspension with a solid extraction material, wherein the liquid is whole blood, bone marrow aspirate, adipose tissue, or fractions thereof obtained from a donor that is not the subject.


Without limiting the mechanism, utility or function of the present technology, the methods and treatments of this technology mediate the effects of interleukin-1 and its role in the inflammation cascade. As generally discussed above, interleukin-1 (IL-1) includes a family of cytokines that can stimulate lymphocytes, neutrophils, and macrophages, activate phagocytes, increase airway fibrosis, promote lymphocyte nodules in the airways, increase production of both MMP-9 and MMP-12, and are involved in many chronic inflammatory conditions. IL-1 can be generated by macrophages, monocytes, and dendritic cells, and can be part of the inflammatory response against infection. See, Lappalainen et al., “Interleukin-1β Causes Pulmonary Inflammation, Emphysema, and Airway Remodeling in the Adult Murine Lung” American Journal of Respiratory Cell and Molecular Biology, vol. 32, no. 4, pages 311-318 (April 2005).


The mode of action of IL-1 can be mediated by IL-1ra. IL-1ra binds to the same receptor on the cell surface as IL-1, and thus prevents IL-1 from sending a signal to that cell. IL-1ra is secreted from white blood cells, including monocytes, macrophages, neutrophils, polymorphonuclear cells (PMNs), and other cells, and can modulate a variety of IL-1 related immune and inflammatory responses, as described by Arend W P, Malyak M, Guthridge C J, Gabay C (1998) “Interleukin-1 receptor antagonist: role in biology” Annu. Rev. Immunol. 16: 27-55. Production of IL-1ra is stimulated by several substances including adherent immunoglobulin G (IgG), other cytokines, and bacterial or viral components. Likewise, the mode of action of TNF-α can be mediated by sTNF-RI and sTNF-RII, which prevent TNF-α from binding to membrane bound TNF-RI and/or TNF-RII.


Examples of inflammatory disorders treated by the methods of this technology include rheumatoid arthritis, osteoarthritis, osteolytis, tendonitis, synovitis, peripheral vascular disease, and inflammatory respiratory diseases (such as chronic obstructive pulmonary disease, fibrosis, emphysema, acute respiratory distress syndrome, and pneumonia). Treatment methods also include the prevention, reduction or elimination of pain associated with various disorders, such as pain associated with traumatic injury, muscle strain, arthritis (rheumatoid arthritis and osteoarthritis), synovitis, sacroiliac joint disorders, back disorders, post-surgical injections, tendon injections, sports medicine procedure (for example, ACL repair, MCL repair, BTB repair, patella repair, or cartilage repair), contusions, muscle strains, post traumatic osteoarthritis. Methods also include stimulation of chondrocyte production at the site of a collagen defect, such as defects at joints associated with arthritis, injuries or surgical procedures.


In some embodiments, methods of the present technology comprise administration of a Protein Solution to the site of a tissue defect to prevent or treat a disorder associated with IL-1ra. As referred to herein, such “tissue defects” include any condition involving tissue which is inadequate for physiological or cosmetic purposes. Examples of such defects include those that are congenital, those that result from or are symptomatic of disease, disorder, or trauma, and those that are consequent to surgical or other medical procedures. Embodiments include treatment for vascular, bone, skin, nerve, and organ tissue defects. Examples of such defects include those resulting from osteoporosis, spinal fixation procedures, hip and other joint replacement procedures, chronic wounds, fractures, sclerosis of tissues and muscles, and spinal cord or other nerve injury. In various embodiments, the compositions and methods of this invention may be used in methods associated with the repair bone or cartilage defects.


In various embodiments, methods are for the treatment of inflammatory disorders in a human. In other embodiments, treatment is for non-human mammals, such as companion, working, and sports animals. For example, such methods of this technology may be used for the treatment of inflammatory disorders in horses.


In various embodiments, methods of the present technology comprise a point-of-care method for making a Protein Solution. As referred to herein, a “point-of-care method” wherein the processes of the present technology are performed at a time proximate to the administration of the Protein Solution to the subject being treated. Such methods may be performed at a location proximate, such as in the same room (for example, bed side) or otherwise immediately adjacent, to the mammalian subject to be treated with the Protein Solution. In various embodiments, a “proximate time” may be, for example, within 12 hours, within 8 hours, within 2 hours, within 1 hour or within 30 minutes of administration of the Protein Solution to the subject.


In some embodiments, the Protein Solution is administered with a concomitant therapy. Such therapies include, for example, the administration of pharmaceutical actives or biologics, as described above. In some embodiments, concomitant therapies are administered concurrently with a Protein Solution. For example, methods may comprise administration of a Protein Solution with a safe and effective amount of an active selected from the group consisting of glucocorticosteroids, non-steroidal anti-inflammatories, antibiotics, antivirals, and combinations thereof.


In some embodiments, methods comprise administration of a Protein Solution with concentrated bone marrow aspirate, as described above. For example, cBMA and a Protein Solution may be administered concomitantly.


Methods of the present technology generally comprise administration of a Protein Solution to the site of inflammation in a mammalian subject. Administration of the Protein Solution can be performed with any suitable device, including such devices known in the art for topical delivery of compositions to the muscle, joint, vascular, lung or other tissue. For example, topical delivery for treatment of inflammation or pain associated with joint disorders may comprise injection of a Protein Solution at or near the joint. Treatment for inflammatory respiratory diseases may comprise delivery of a Protein Solution by endotracheal tubes, inhalers and nebulizers.


Embodiments of the present technology are further illustrated through the following non-limiting examples.


Example 1
Preparing and Characterizing a Protein Solution

A Protein Solution rich in interleukin-I receptor antagonist is prepared from seven consented human providers. Blood (55 mL) is drawn into a 60 cc syringe with 5 mL of anticoagulant citrate dextrose solution A (ACD-A, Citra Labs, Braintree, Mass.). Platelet-rich plasma (PRP) is created using the GPS® III platelet concentration system (800-1 003A, Biomet Biologics, Warsaw, Ind.) according to the instructions for use. The solution is generated by adding 6 mL of PRP to a modified Plasmax device containing 1 gram of polyacrylamide beads (Biomet Biologics, Warsaw, Ind.). The IL-Ira solution is removed from the Plasmax devices and frozen at minus 50° C. for the assay. Cytokine content is assayed on a 16-plex ELISA (Searchlight® Protein Array ELISA system, Aushon Biosystems, Billerica, Mass.). The analytes included IL-4, IL-10, IL-11, IL-I3, IL-Ira, IFN-γ, sTNF-RI, sTNF-RII, IL-1α, IL-1β, TNF-α, IL-17, IL-18, bFGF, TBF-β1, and TBF-β2.


The solution contains both anabolic (bFGF, TGF-β1, TGF-β2 (see Table 3)) and anti-inflammatory (IL-1ra, sTNF-RI, sTNF-RII, IL-4, IL-10, IL-11, IL-13, IFN-γ, (see Table 4)) cytokines without expressing large doses of catabolic cytokines (IL-1α, IL-1β, TNF-α, IL-17, IL-18 (see Table 5)). The anti-inflammatory cytokines IL-Ira and sTNF-R are all detected in ng/mL quantities, while all of the catabolic analytes were in pg/mL quantities. However, donor-to-donor variability is detected. Correlations between the catabolic cytokines IL-1 and TNF-α and anti-inflammatory analytes IL-1ra and sTNF-R are compared, but no large correlations detected (Table 6). On average, there is about 13,260 times more IL-1ra than IL-1a and about 7,561 times more than IL-1β.









TABLE 3







Anabolic cytokines in the solution.












Donor
bFGF
TGF-β1
TGF-β2
















1
18.5
1,458,008
153,833



2
10.7
1,137,404
119,545



3
11.9
585,298
70,544



4
4.9
1,342,442
162,707



5
20.0
1,579,361
204,670



6
7.7
1,393,746
170,345



7
13.9
1,474,155
174,502



Average
12.5
1,281,488
150,878



±SD
±5.5
±336,345
±43,617

















TABLE 4







Anti-inflammatory cytokines in the solution.






















TNF-



Donor
IFN-γ
IL-4
IL-10
IL-13
IL-1ra
TNF-RI
RII
IL-11


















1
<0.4
2.1
0.5
3.5
9,660
2,728
2,249
<2.0


2
<0.4
1.3
0.3
2.8
17,477
5,120
2,900
<2.0


3
<0.4
<0.8
0.3
0.1
23,126
6,247
2,446
<2.0


4
40.4
59.9
8.9
19.9
10,458
4,374
2,612
<2.0


5
30.2
33.9
23.3
15.8
13,462
2,763
1,394
<2.0


6
2.6
23.3
1.4
25.6
8,813
2,992
2,716
<2.0


7
0.7
1.2
0.6
1.8
11,277
3,330
1,915
<2.0


Average
10.7
17.5
5.0
9.9
13,468
3,936
2,319
<2.0


±SD
±17.0
±22.9
±8.7
±10.3
±5,154
±1,356
±520
±0
















TABLE 5







Catabolic cytokines in the solution.














Donor
IL-17
TNF-α
IL-1α
IL-1β
IL-18


















1
3.1
16.0
<0.8
1.5
239



2
1.2
<2.3
2.5
3.3
559



3
0.7
<2.3
1.8
2.3
511



4
28.9
195
0.8
1.3
329



5
33.8
661
0.8
2.0
450



6
22.0
105
0.3
1.7
333



7
6.7
<2.3
1.9
1.0
787



Average
13.8
141
1.3
1.9
458



±SD
±14.1
±241
±0.8
±0.8
±183

















TABLE 6







Correlation analysis.











Analytes compared
R2
Ratio







IL-1ra and IL-1α
0.46
13,260X  



IL-1ra and IL-1β
0.45
7,561X  



TNF-RI and TNF-α
0.17
945X



TNF-RII and TNF-α
0.47
477X










Example 2
Generation of IL-1Ra from Platelet-Rich Plasma

An IL-1ra-rich solution is created as follows. Whole blood (70 mL) anticoagulated (10%) with ACD-A (Braintree, Mass., USA) is drawn from 5 healthy volunteers. A portion (10 mL) is reserved for a whole blood measurement. Platelet-rich plasma (PRP) (6 mL) is produced using the GPS® II System (Biomet Biologics, LLC, Warsaw, Ind., USA). Complete blood counts are collected for the whole blood and PRP samples following a validated procedure, as described in Woodell-May J E, Ridderman D N, Swift M J, Higgins J. “Producing Accurate Platelet Counts for Platelet Rich Plasma: Validation of a Hematology Analyzer and Preparation Techniques for Counting” J. Craniofac. Surg. (2005) September 16(5):749-56.


Following the PRP production, 5 mL of the PRP is added to a modified plasma concentration device (Plasmax™, Biomet Biologics LLC, Warsaw, Ind., USA) and incubated with polyacrylamide desiccating beads in the device for 24 hours at room temperature. Following the contact with polyacrylamide beads the electromagnetic field, the plasma concentration device is centrifuged to separate the serum fraction.


To analyze baseline IL-1ra levels at time zero, the whole blood and PRP samples are activated with 50 μL of thrombin and 10% CaCl2 (1,000 units/mL). A blood clot is formed and incubated for 30 minutes at room temperature. Following incubation, the clot is centrifuged for 5 minutes at 3,000 rpm. Serum is collected from the clots and retained for ELISA analysis. The serum fraction from the plasma concentrator does not require activation by thrombin, and is tested directly. All samples are analyzed for IL-1ra using an ELISA kit (IL-1ra Quantikine™ Kit, R&D Systems, Minneapolis, Minn., USA).


The PRP samples result in about an eight-fold increase in platelets, about five-fold increase in total white blood cells (WBCs), about nine-fold increase in the monocyte fraction of the WBCs, and about a three-fold increase in the PMN fraction of the WBCs. The IL-1ra production in the whole blood and PRP samples is correlated most closely to the WBC concentration. The five-fold increase in the PRP is likely due to the increase in WBCs, and both the whole blood and PRP IL-1ra values can be considered baseline IL-1ra content. This is in contrast to the 195-fold increase in IL-1ra following incubation in the plasma concentrator. This plasma concentration device typically results in a 3-fold increase in plasma protein concentration due to a volume reduction caused by the desiccation process. This 3-fold decrease in volume does not account for the levels of increase seen in the amount of IL-1ra. Therefore, this level of increase indicates stimulation of WBCs to produce IL-1ra during the contact with the solid extraction material (e.g., polyacrylamide beads) and electromagnetic field stimulation.


Correlation analysis demonstrates that IL-1ra production is more closely correlated with the increase in WBCs than the platelet content. The IL-1ra levels do not correlate as closely with the WBC population in the PRP. This is not surprising since the WBC are not activated, and the serum is collected by thrombin activation of the plasma. However, it is possible that the WBC, once activated in the plasma concentration device, participate in the significant production of IL-1ra seen in this example.


Example 3
Production of Protein Solution from PRP

Anticoagulated blood (120 cc) is collected from 5 human donors. Platelet-rich plasma (PRP) is prepared using GPS® III disposables (Biomet Biologics LLC, Warsaw, Ind., USA). PRP is loaded into modified plasma concentration devices (Plasmax™, Biomet Biologics LLC, Warsaw, Ind., USA) and processed. The output is divided into 4 groups: IL-1ra in concentrated plasma with and without thrombin activation (1000 U/mL in 1M CaC12), or cell-free IL-1ra with and without thrombin activation. IL-1ra is measured using ELISA (R&D Systems) over time.


The PRP contacts polyacrylamide beads in the Plasmax™ device while electromagnetic field stimulation is provided using a capacitively coupled electromagnetic field.


Unclotted PRP produces an average of about 50 ng over 24 hrs. The cell-free samples produce about 34 ng without changing over 24 hrs. Once clotted, the elution of IL-1ra is slowed, with only about 30% being eluted after 10 hours. Release in the cell-free samples is also delayed, but eluted 100% of available IL-1ra after 10 hours.


Example 4
Generation of Protein Solution and Characterization of Cytokine Levels in Healthy Subjects and Osteoarthritis Subjects

Protein Solutions from healthy patients are prepared as follows for the measurement of growth factors. 72 ml of anticoagulated whole blood are drawn by venipuncture from each of six donors. 3 ml of each donor's anticoagulated whole blood are aliquoted into microcentrifuge tubes and frozen at −50° C. 60 ml of the anticoagulated whole blood is loaded into GPS® III disposable devices (Biomet Biologics LLC, Warsaw, Ind., USA), which is processed according to the manufacturer's instructions to produce PRP. The PRP is removed from the GPS® III devices and added to Plasmax™ devices (Biomet Biologics LLC, Warsaw, Ind., USA), which is processed according to the manufacturer's instructions to produce APS. APS is extracted from each device, aliquoted into microcentrifuge tubes, and frozen at −50° C. Each sample, whole blood and PRP, is subjected to three freeze-thaw cycles. Quantikine Human Immunoassays (R&D Systems, Inc., Minneapolis, Minn.) for VEGF, PDGF-BB, PDGF-AB, EGF, TGF-β1, TGF-β2, and IGF-1 are run in duplicate according to the manufacturer's instructions for each APS and whole blood sample.


APS from healthy patients is prepared as above for the measurement of anti-inflammatory cytokines. Quantikine Human Immunoassays (R&D Systems, Inc., Minneapolis, Minn.) for IL-1ra, IL-1β, IL-8, sTNF-RI, TNF-α, IL-6, sTNF-RII, IL-10, IL-13, and IL-4 are run in duplicate according to the manufacturer's instructions for each APS and whole blood sample. Immunoassays are also performed to detect hepatocyte growth factor (HGF) and soluble IL-1RII.


APS from 105 osteoarthritis patients is prepared as above for the measurement of growth factors anti-inflammatory cytokines. The APS is stored at −50° C. or in dry ice.


Cytokine concentrations are compared between healthy donors and OA patients in baseline blood and APS. IL-1β is concentrated at a higher level in OA patients, but the fold increase is still much lower than that of IL-1ra. Other cytokines and growth factors that are concentrated at least to the level of that observed in healthy donors include sTNF-RI, IGF-I, IL-8, VEGF, and IL-6. The soluble cytokines sTNF-RII and sIL-1RII are concentrated to a level not quite as high but very similar to the healthy concentration level. The results are displayed in Table 7.









TABLE 7







Concentration of growth factors and anti-inflammatory


cytokines from APS derived from healthy patients


and patients with osteoarthritis (in pg/ml).













Fold



Baseline
APS
Increase












Cytokine
Average
StDev
Average
StDev
Average
















VEGF
Healthy
276
109
742
494
2.7



OA
484
201
1710
1025
3.8


IL-1β
Healthy
3.4
2
3.8
0.8
1.1



OA
3.3
1.1
8.9
7.3
2.8


IL-8
Healthy
74
16
315
198
4.3



OA
73.5
29.6
287.9
192.7
4.2


IL-6
Healthy
3.1
0.4
3.4
0.7
1.1



OA
1.8
1.3
3
3.5
1.6


TNF-α
Healthy
ND
ND
3.4
0.7
ND



OA
2.4
2
4.3
3
5.3


IL-1ra
Healthy
8092
2536
30853
16737
3.8



OA
7576
2469
41896
19669
5.9


sTNF-
Healthy
2485
338
9491
1387
3.8


RII
OA
1491
492
5060
1946
3.5


PDGF-
Healthy
13400
3400
91700
24100
6.8


AB
OA
16799
5731
37889
24922
2.5


PDGF-
Healthy
4702
1027
23810
6126
5.1


BB
OA
5306
2422
11936
8655
2.5


IGF-I
Healthy
114000
30000
155000
34000
1.4



OA
79072
22137
118060
42827
1.5


EGF
Healthy
240
71
1227
300
5.1



OA
374
199
707
489
2.2


sTNF-
Healthy
629
76
2408
338
3.8


RI
OA
808
275
3011
964
3.9


TGF-β1
Healthy
25717
11131
181245
56420
7.1



OA
56594
56940
153567
145973
4.2


sIL-
Healthy
11,786
ND
26,000
ND
2.2


1RII
OA
ND
ND
ND
ND
ND


HGF
Healthy
782
ND
3244
ND
4.1



OA
ND
ND
ND
ND
ND









Example 5
Generation of a Protein Solution from Adipose Tissue

Adipose stromal cells are prepared as follows. Adipose tissue is minced into small pieces (about 1 cm3) and digested in 2 mg/mL type I collagenase (Worthington Biochemical Corp., Lakewood, N.J.) under intermittent mechanical agitation in a water bath at 37° C. for 180 minutes. Digestion can be neutralized by the addition of medium or a blood-derived solution. The cell suspension is centrifuged (300×g for 7 minutes at 25° C.) followed by removal of the supernatant from the cell pellet. The pellet is then re-suspended in a compatible solution to provide a liquid volume comprising Adipose stromal cells.


Alternatively, the pellet is suspended with whole blood obtained from the subject, and added to a GPS™ Platelet Concentrate System, from Biomet Biologics, Inc. (Warsaw, Ind.). Following centrifugation, the platelet-rich plasma layer, which also contains the adipose stromal cells, is extracted from the system.


The adipose stromal cells, optionally including platelet-rich plasma, are then combined with polyacrylamide beads to stimulate production of IL-1ra. The adipose stromal cells and polyacrylamide beads are separated from the liquid solution to obtain a solution rich in IL-1ra.


Example 6
Generation of Protein Solution from Lipoaspirate

A therapeutic composition of IL-1ra is generated from stromal cells isolated from adipose tissue. Isolation of human stromal cells is performed by obtaining human subcutaneous adipose tissue from lipoaspiration/liposuction procedures and digesting the tissue in collagenase type I solution (Worthington Biochemical Corp., Lakewood, N.J.) under gentle agitation for 1 hour at 37° C. The dissociated cells are filtered with 500 μm and 250 μm Nitex filters. The fraction is centrifuged at 300×g for 5 minutes. The supernatant is discarded and the cell pellet is re-suspended in a compatible liquid solution, such as a blood-derived solution.


Non-Limiting Discussion of Terminology

The headings (such as “Introduction” and “Summary”) and sub-headings used herein are intended only for general organization of topics within the present disclosure, and are not intended to limit the disclosure of the technology or any aspect thereof. In particular, subject matter disclosed in the “Introduction” may include novel technology and may not constitute a recitation of prior art. Subject matter disclosed in the “Summary” is not an exhaustive or complete disclosure of the entire scope of the technology or any embodiments thereof. Classification or discussion of a material within a section of this specification as having a particular utility is made for convenience, and no inference should be drawn that the material must necessarily or solely function in accordance with its classification herein when it is used in any given composition.


The disclosure of all patents and patent applications cited in this disclosure are incorporated by reference herein.


The description and specific examples, while indicating embodiments of the technology, are intended for purposes of illustration only and are not intended to limit the scope of the technology. Equivalent changes, modifications and variations of specific embodiments, materials, compositions and methods may be made within the scope of the present technology, with substantially similar results. Moreover, recitation of multiple embodiments having stated features is not intended to exclude other embodiments having additional features, or other embodiments incorporating different combinations of the stated features. Specific examples are provided for illustrative purposes of how to make and use the compositions and methods of this technology and, unless explicitly stated otherwise, are not intended to be a representation that given embodiments of this technology have, or have not, been made or tested.


As used herein, the words “prefer” or “preferable” refer to embodiments of the technology that afford certain benefits, under certain circumstances. However, other embodiments may also be preferred, under the same or other circumstances. Furthermore, the recitation of one or more preferred embodiments does not imply that other embodiments are not useful, and is not intended to exclude other embodiments from the scope of the technology.


As used herein, the word “include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the materials, compositions, devices, and methods of this technology. Similarly, the terms “can” and “may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.


Although the open-ended term “comprising,” as a synonym of non-restrictive terms such as including, containing, or having, is used herein to describe and claim embodiments of the present technology, embodiments may alternatively be described using more limiting terms such as “consisting of” or “consisting essentially of.” Thus, for any given embodiment reciting materials, components or process steps, the present technology also specifically includes embodiments consisting of, or consisting essentially of, such materials, components or processes excluding additional materials, components or processes (for consisting of) and excluding additional materials, components or processes affecting the significant properties of the embodiment (for consisting essentially of), even though such additional materials, components or processes are not explicitly recited in this application. For example, recitation of a composition or process reciting elements A, B and C specifically envisions embodiments consisting of, and consisting essentially of, A, B and C, excluding an element D that may be recited in the art, even though element D is not explicitly described as being excluded herein. Further, as used herein the term “consisting essentially of” recited materials or components envisions embodiments “consisting of” the recited materials or components.


A” and “an” as used herein indicate “at least one” of the item is present; a plurality of such items may be present, when possible. “About” when applied to values indicates that the calculation or the measurement allows some slight imprecision in the value (with some approach to exactness in the value; approximately or reasonably close to the value; nearly). If, for some reason, the imprecision provided by “about” is not otherwise understood in the art with this ordinary meaning, then “about” as used herein indicates at least variations that may arise from ordinary methods of measuring or using such parameters.


As referred to herein, ranges are, unless specified otherwise, inclusive of endpoints and include disclosure of all distinct values and further divided ranges within the entire range. Thus, for example, a range of “from A to B” or “from about A to about B” is inclusive of A and of B. Disclosure of values and ranges of values for specific parameters (such as temperatures, molecular weights, weight percentages, etc.) are not exclusive of other values and ranges of values useful herein. It is envisioned that two or more specific exemplified values for a given parameter may define endpoints for a range of values that may be claimed for the parameter. For example, if Parameter X is exemplified herein to have value A and also exemplified to have value Z, it is envisioned that Parameter X may have a range of values from about A to about Z. Similarly, it is envisioned that disclosure of two or more ranges of values for a parameter (whether such ranges are nested, overlapping or distinct) subsume all possible combination of ranges for the value that might be claimed using endpoints of the disclosed ranges. For example, if Parameter X is exemplified herein to have values in the range of 1-10, or 2-9, or 3-8, it is also envisioned that Parameter X may have other ranges of values including 1-9, 1-8, 1-3, 1-2, 2-10, 2-8, 2-3, 3-10, and 3-9.

Claims
  • 1. A method for making a non-immunogenic anti-inflammatory cytokine composition, the method comprising: contacting a suspension comprising white blood cells, obtained from a non-autologous source, with a solid extraction material to generate a fluid rich-in interleukin-1 receptor antagonist;separating the white blood cells from the fluid rich in interleukin-1 receptor antagonist; andfreeze drying the fluid rich in interleukin-1 receptor antagonist, after the step of separating, to produce a non-immunogenic anti-inflammatory cytokine composition.
  • 2. The method according to claim 1, wherein the suspension comprises any one of: a. whole blood, bone marrow aspirate, bone marrow concentrate, adipose tissue, platelet-rich plasma, or urine; b. a fraction thereof; or c. a mixture thereof of any of the foregoing.
  • 3. The method according to claim 1, wherein the composition rich in interleukin-1 receptor antagonist comprises at least one of soluble tumor necrosis factor receptor I, soluble tumor necrosis factor receptor II, soluble interleukin-1 receptor II, hepatocyte growth factor, platelet-derived growth factor AB, platelet-derived growth factor BB, epidermal growth factor, vascular endothelial growth factor, insulin-like growth factor-1, and transforming growth factor-β1.
  • 4. The method according to claim 1, wherein the solid extraction material is selected from corundum, quartz, titanium, dextran, agarose, polyacrylamide, polystyrene, polyethylene, polyvinyl chloride, polypropylene, and a combination thereof.
  • 5. The method according to claim 1, wherein the solid extraction material comprises polyacrylamide.
  • 6. The method according to claim 1, wherein the solid extraction material comprises a form selected from a bead, fiber, powder, porous material, and a combination thereof.
  • 7. The method according to claim 1, wherein after the separating step the fluid rich in interleukin-1 receptor antagonist is free of the white blood cells.
  • 8. The method of claim 1, wherein the suspension is prepared by a process comprising: loading cytokine-producing tissue into a tube comprising a buoy disposed in the tube, wherein the buoy has a density such that the buoy is operable to reach an equilibrium position upon centrifugation of the tissue in the tube, the equilibrium position being between a first fraction and a second fraction comprising the white blood cells, the second fraction having a concentration of white blood cells greater than a concentration of white blood cells in the first fraction;centrifuging the tube so that the buoy defines an interface between the first fraction and the second fraction; and
  • 9. The method according to claim 1, comprising: loading tissue comprising whole blood, bone marrow aspirate, or both, into a tube comprising a buoy disposed in the tube, wherein the buoy has a density such that the buoy reaches an equilibrium position upon centrifugation of the tissue in the tube, the equilibrium position being between a first fraction and a second fraction comprising white blood cells, the second fraction having a concentration of white blood cells greater than the concentration of white blood cells in the first fraction;centrifuging the tube so that the buoy defines an interface between the first fraction and the second fraction;collecting the second fraction as the suspension;loading the suspension into a concentrator assembly comprising the solid extraction material and incubating the suspension in contact with the solid extraction material.
  • 10. The method according to claim 1, wherein the contacting comprises subjecting the suspension to an electromagnetic field.
  • 11. The method according to claim 10, wherein the electromagnetic field comprises a pulsed electromagnetic field or a capacitively coupled electromagnetic field.
  • 12. The method according to claim 1, further comprising reconstituting the non-immunogenic anti-inflammatory cytokine composition in a liquid comprising any one of saline, a buffer, blood, a blood fraction, a blood fraction concentrate, a bone marrow aspirate, or a bone marrow concentrate; or a combination of any of the foregoing.
  • 13. The method according to claim 1, wherein before the freeze-drying the fluid rich in interleukin-1 receptor antagonist comprises: interleukin-1 receptor antagonist (IL-1ra) at a concentration of at least about 10,000 pg/ml;soluble tumor necrosis factor receptor 1 (sTNF-R1) at a concentration of at least about 1,200 pg/ml; anda protein selected from soluble tumor necrosis factor-receptor 2 (sTNF-RII), insulin-like growth factor 1 (IGF-I), epidermal growth factor (EGF), hepatocyte growth factor (HGF), platelet-derived growth factor AB (PDGF-AB), platelet-derived growth factor BB (PDGF-BB), vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1), soluble insulin receptor II (sIL-1RII), and a mixture thereof, wherein the concentration of the selected protein in the fluid is greater than the concentration of the protein in the suspension.
  • 14. A method for making a non-immunogenic anti-inflammatory cytokine composition, the method comprising: contacting a suspension comprising a liquid volume of white blood cells from a non-autologous source with a solid extraction material to generate a composition rich in interleukin-1 receptor antagonist;isolating the composition from the solid extraction material and the white blood cells to provide an isolated composition;mixing the isolated composition with a storage solution to make a stabilized composition; andfreezing the stabilized composition to generate a non-immunogenic anti-inflammatory cytokine composition.
  • 15. The method according to claim 14, wherein the storage solution comprises glycerol, dimethylsulfoxide, or a mixture thereof.
  • 16. The method according to claim 15, wherein freezing comprises storage at a temperature from 1° C. to about −80° C.
  • 17. A method for making an anti-inflammatory composition, the method comprising: receiving a suspension of white blood cells from a non-autologous source;activating the white blood cells to generate a composition rich in interleukin-1 receptor antagonist;separating the white blood cells from the composition rich in interleukin-1 receptor antagonist; andfreeze drying the composition rich in interleukin-1 receptor antagonist, after the separating step, to produce an anti-inflammatory composition that is non-immunogenic and free of viable white blood cells.
  • 18. The method according to claim 17, wherein the suspension comprises any one of platelets, stromal cells, hematopoietic stem cells, mesenchymal stem cells, or endothelial progenitor cells; or a combination of any of the foregoing.
  • 19. The method according to claim 17, wherein the suspension comprises any one of: a. whole blood, a blood clot, bone marrow aspirate, adipose tissue, a platelet-rich plasma, or urine; b. a fraction thereof; or c. a mixture thereof of any of the foregoing.
  • 20. The method according to claim 17, wherein the composition rich in interleukin-1 receptor antagonist further comprises a protein selected from soluble tumor necrosis factor-receptor I, soluble tumor necrosis factor-receptor II, insulin-like growth factor-1, epidermal growth factor, hepatocyte growth factor, platelet-derived growth factor AB, platelet-derived growth factor BB, vascular endothelial growth factor, transforming growth factor-β1 (TGF-1β), soluble insulin receptor II, and a mixture thereof, wherein the concentration of the selected protein in the composition is greater than the concentration of the selected protein in the suspension.
  • 21. The method according to claim 17, wherein activating includes subjecting the suspension to an electromagnetic field.
  • 22. The method according to claim 17, wherein activating includes contacting the suspension with a solid extraction material.
  • 23. The method according to claim 22, wherein the solid extraction material is selected from corundum, quartz, titanium, dextran, agarose, polyacrylamide, polystyrene, polyethylene, polyvinyl chloride, polypropylene, and a combination thereof.
  • 24. The method according to claim 22, wherein the solid extraction material comprises a form selected from a bead, fiber, powder, porous material, and a combination thereof.
US Referenced Citations (648)
Number Name Date Kind
593333 Park Nov 1897 A
1468313 Fritz Sep 1923 A
1593814 Robert Jul 1926 A
2722257 Lockhart Nov 1955 A
3013557 Pallotta Dec 1961 A
3141846 Laven, Jr. Jul 1964 A
3159159 Cohen Dec 1964 A
3300051 Mitchell Jan 1967 A
3409165 Creith Nov 1968 A
3420374 Umeda Jan 1969 A
3441143 Kudlaty Apr 1969 A
3453364 Flodin et al. Jul 1969 A
3469369 Helmke Sep 1969 A
3508653 Coleman Apr 1970 A
3545671 Ross Dec 1970 A
3583627 Wilson Jun 1971 A
3596652 Winkelman Aug 1971 A
3647070 Adler Mar 1972 A
3654925 Holderith Apr 1972 A
3661265 Greenspan May 1972 A
3706305 Berger et al. Dec 1972 A
3706306 Berger et al. Dec 1972 A
3723244 Breillatt Mar 1973 A
3741400 Dick Jun 1973 A
3779383 Ayres Dec 1973 A
3785549 Latham Jan 1974 A
3814248 Lawhead Jun 1974 A
3849072 Ayres Nov 1974 A
3850369 Bull et al. Nov 1974 A
3879295 Glover et al. Apr 1975 A
3887466 Ayres Jun 1975 A
3894952 Ayres Jul 1975 A
3896733 Rosenberg Jul 1975 A
3897337 Ayres Jul 1975 A
3897343 Ayres Jul 1975 A
3909419 Ayres Sep 1975 A
3929646 Adler Dec 1975 A
3931010 Ayres et al. Jan 1976 A
3931018 North, Jr. Jan 1976 A
3935113 Ayres Jan 1976 A
3937211 Merten Feb 1976 A
3941699 Ayres Mar 1976 A
3945928 Ayres Mar 1976 A
3951801 Ayres Apr 1976 A
3957654 Ayres May 1976 A
3962085 Liston et al. Jun 1976 A
3965889 Sachs Jun 1976 A
3972812 Gresl, Jr. Aug 1976 A
3982691 Schlutz Sep 1976 A
4001122 Griffin Jan 1977 A
4020831 Adler May 1977 A
4046699 Zine, Jr. Sep 1977 A
4055501 Cornell Oct 1977 A
4059108 Latham, Jr. Nov 1977 A
4066549 Oeser et al. Jan 1978 A
4077396 Wardlaw et al. Mar 1978 A
4088582 Murty May 1978 A
4146172 Cullis et al. Mar 1979 A
4152270 Cornell May 1979 A
4154690 Ballies May 1979 A
4159896 Levine et al. Jul 1979 A
4187979 Cullis et al. Feb 1980 A
4189385 Greenspan Feb 1980 A
4203840 Stoeppler et al. May 1980 A
4204537 Latham, Jr. May 1980 A
4225580 Rothman et al. Sep 1980 A
4229298 Bange Oct 1980 A
4269718 Persidsky May 1981 A
4294707 Ikeda et al. Oct 1981 A
4298598 Schwarz et al. Nov 1981 A
4300717 Latham, Jr. Nov 1981 A
4303193 Latham, Jr. Dec 1981 A
4314823 Rich, Jr. Feb 1982 A
4322298 Persidsky Mar 1982 A
4332351 Kellogg Jun 1982 A
4362567 Schwarz et al. Dec 1982 A
4364832 Ballies Dec 1982 A
4377572 Schwarz et al. Mar 1983 A
4379849 Heimreid Apr 1983 A
4411794 Schwinn et al. Oct 1983 A
4414976 Schwarz et al. Nov 1983 A
4416654 Schoendorfer Nov 1983 A
4417981 Nugent Nov 1983 A
4424132 Iriguchi Jan 1984 A
4427650 Stroetmann Jan 1984 A
4427651 Stroetmann Jan 1984 A
4442655 Stroetmann Apr 1984 A
4443345 Wells Apr 1984 A
4445550 Davis et al. May 1984 A
4446021 Aufderhaar et al. May 1984 A
4453927 Sinko Jun 1984 A
4453939 Zimmerman et al. Jun 1984 A
4464167 Schoendorfer Aug 1984 A
4511662 Baran et al. Apr 1985 A
4537767 Rothman et al. Aug 1985 A
RE32089 Blatt et al. Mar 1986 E
4577514 Bradley et al. Mar 1986 A
4610656 Mortensen Sep 1986 A
4617009 Ohlin Oct 1986 A
4627879 Rose et al. Dec 1986 A
4631055 Redl et al. Dec 1986 A
4632761 Bowers Dec 1986 A
4639316 Eldegheidy Jan 1987 A
4650678 Fuhge et al. Mar 1987 A
4655211 Sakamoto et al. Apr 1987 A
4672969 Dew Jun 1987 A
4675117 Neumann et al. Jun 1987 A
4680025 Kruger Jul 1987 A
4708799 Gerlach et al. Nov 1987 A
4714457 Alterbaum Dec 1987 A
4722790 Cawley et al. Feb 1988 A
4724317 Brown Feb 1988 A
4735616 Eibl et al. Apr 1988 A
4735726 Duggins Apr 1988 A
4738655 Brimhall et al. Apr 1988 A
4755300 Fischel et al. Jul 1988 A
4755301 Bowers Jul 1988 A
4770779 Ichikawa et al. Sep 1988 A
4776964 Schoendorfer et al. Oct 1988 A
4818291 Iwatsuki et al. Apr 1989 A
4818386 Burns Apr 1989 A
4828710 Itoh et al. May 1989 A
4832851 Bowers May 1989 A
4834890 Brown et al. May 1989 A
4839058 Cawley et al. Jun 1989 A
4844818 Smith Jul 1989 A
4846780 Galloway et al. Jul 1989 A
4846835 Grande Jul 1989 A
4850952 Figdor et al. Jul 1989 A
4853137 Ersson Aug 1989 A
4871462 Fischel et al. Oct 1989 A
4874368 Miller et al. Oct 1989 A
4877520 Burns Oct 1989 A
4879031 Panzani Nov 1989 A
4900453 Sedlmayer Feb 1990 A
4902281 Avoy Feb 1990 A
4909251 Seelich Mar 1990 A
4915847 Dillon et al. Apr 1990 A
4917801 Luderer et al. Apr 1990 A
4928603 Rose et al. May 1990 A
4929242 Desecki et al. May 1990 A
4933291 Daiss et al. Jun 1990 A
4939081 Figdor et al. Jul 1990 A
4943273 Pages Jul 1990 A
4946601 Fiehler Aug 1990 A
4950220 Wells et al. Aug 1990 A
4957637 Cornell Sep 1990 A
4957638 Smith Sep 1990 A
4973168 Chan Nov 1990 A
4983157 Pober et al. Jan 1991 A
4983158 Headley Jan 1991 A
4985153 Kuroda et al. Jan 1991 A
5000970 Shanbhag et al. Mar 1991 A
5002571 O'donnell, Jr. Mar 1991 A
5019243 McEwen et al. May 1991 A
5024613 Vasconcellos et al. Jun 1991 A
5030215 Morse et al. Jul 1991 A
5030341 McEwen et al. Jul 1991 A
5039401 Columbus et al. Aug 1991 A
5045048 Kaleskas Sep 1991 A
5047004 Wells Sep 1991 A
5053127 Schoendorfer et al. Oct 1991 A
5053134 Luderer et al. Oct 1991 A
5071570 Shiraki et al. Dec 1991 A
5075222 Hannum et al. Dec 1991 A
5080262 Herold et al. Jan 1992 A
5086784 Levine et al. Feb 1992 A
5100564 Pall et al. Mar 1992 A
5104375 Wolf et al. Apr 1992 A
5112484 Zuk, Jr. May 1992 A
5112490 Turpen May 1992 A
5131907 Williams et al. Jul 1992 A
5137832 Levine et al. Aug 1992 A
5141645 Shiraki et al. Aug 1992 A
5147290 Jonsson Sep 1992 A
5152905 Pall et al. Oct 1992 A
5156613 Sawyer Oct 1992 A
5165938 Knighton Nov 1992 A
5171456 Hwang et al. Dec 1992 A
5173295 Wehling Dec 1992 A
5178602 Wells Jan 1993 A
5185001 Galanakis Feb 1993 A
5188583 Guigan Feb 1993 A
5190057 Sarfarazi Mar 1993 A
5190759 Lindblad et al. Mar 1993 A
5197985 Caplan Mar 1993 A
5203825 Haynes Apr 1993 A
5204537 Bennet et al. Apr 1993 A
5206023 Hunziker Apr 1993 A
5207638 Choksi et al. May 1993 A
5217426 Bacehowski et al. Jun 1993 A
5217627 Pall et al. Jun 1993 A
5219328 Morse et al. Jun 1993 A
5226877 Epstein Jul 1993 A
5226914 Caplan et al. Jul 1993 A
5234608 Duff Aug 1993 A
5236604 Fiehler Aug 1993 A
5251786 Sarrine Oct 1993 A
5258126 Pall et al. Nov 1993 A
5260420 Burnouf-radosevich et al. Nov 1993 A
5269927 Fiehler Dec 1993 A
5271852 Luoma, II Dec 1993 A
5279825 Wehling Jan 1994 A
5281342 Biesel et al. Jan 1994 A
5290552 Sierra et al. Mar 1994 A
5290918 Bui-khac Mar 1994 A
5298171 Biesel Mar 1994 A
5304372 Michalski et al. Apr 1994 A
5316674 Pall et al. May 1994 A
5318524 Morse et al. Jun 1994 A
5318782 Weis-fogh Jun 1994 A
5321126 Van Dommelen et al. Jun 1994 A
5322620 Brown et al. Jun 1994 A
5330974 Pines et al. Jul 1994 A
5344752 Murphy Sep 1994 A
5354483 Furse Oct 1994 A
5359032 Dayer et al. Oct 1994 A
5370221 Magnusson et al. Dec 1994 A
5370802 Brown Dec 1994 A
5372945 Alchas et al. Dec 1994 A
5376263 Fischel Dec 1994 A
5387187 Fell et al. Feb 1995 A
5393674 Levine et al. Feb 1995 A
5395923 Bui-khac et al. Mar 1995 A
5403272 Deniega et al. Apr 1995 A
5405607 Epstein Apr 1995 A
5409833 Hu et al. Apr 1995 A
5411885 Marx May 1995 A
5417650 Gordon May 1995 A
5420250 Lontz May 1995 A
5443481 Lee Aug 1995 A
5454958 Fiehler Oct 1995 A
5456693 Conston et al. Oct 1995 A
5456885 Coleman et al. Oct 1995 A
5474687 Van Vlasselaer Dec 1995 A
5480378 Weis-fogh et al. Jan 1996 A
5484383 Fitch, Jr. et al. Jan 1996 A
5486359 Caplan et al. Jan 1996 A
5494578 Brown Feb 1996 A
5494592 Latham, Jr. et al. Feb 1996 A
5501371 Schwartz-feldman Mar 1996 A
5505685 Antwiler Apr 1996 A
5510102 Cochrum Apr 1996 A
5520885 Coelho et al. May 1996 A
5525477 Hassouna Jun 1996 A
5533518 Vogler Jul 1996 A
5560830 Coleman et al. Oct 1996 A
5571418 Lee et al. Nov 1996 A
5575778 Hardt et al. Nov 1996 A
5577513 Van Vlasselaer Nov 1996 A
5585007 Antanavich et al. Dec 1996 A
5588958 Cunningham et al. Dec 1996 A
5589462 Patat et al. Dec 1996 A
5599558 Gordinier et al. Feb 1997 A
5601711 Sklar et al. Feb 1997 A
5601727 Bormann et al. Feb 1997 A
5603845 Holm Feb 1997 A
5607579 Latham, Jr. et al. Mar 1997 A
5614106 Payrat et al. Mar 1997 A
5618663 Delmas Apr 1997 A
5632895 Tsukagoshi et al. May 1997 A
5632905 Haynes May 1997 A
5641414 Brown Jun 1997 A
5641622 Lake et al. Jun 1997 A
5643192 Hirsh Jul 1997 A
5643193 Papillon et al. Jul 1997 A
5645540 Henniges et al. Jul 1997 A
5646004 Van Vlasselaer Jul 1997 A
5648223 Van Vlasselaer Jul 1997 A
5649903 Deniega et al. Jul 1997 A
5663051 Vlasselaer Sep 1997 A
5674173 Hlavinka et al. Oct 1997 A
5707331 Wells et al. Jan 1998 A
5707647 Dunn et al. Jan 1998 A
5707876 Levine Jan 1998 A
5716616 Prockop et al. Feb 1998 A
5723331 Tubo et al. Mar 1998 A
5724988 Dennehey et al. Mar 1998 A
5733466 Benebo et al. Mar 1998 A
5733545 Hood, III Mar 1998 A
5736033 Coleman et al. Apr 1998 A
5738784 Holm et al. Apr 1998 A
5738796 Bormann et al. Apr 1998 A
5750025 Holmes et al. May 1998 A
5750658 Coelho et al. May 1998 A
5762798 Wenthold et al. Jun 1998 A
5785700 Olson Jul 1998 A
5786217 Tubo et al. Jul 1998 A
5788662 Antanavich et al. Aug 1998 A
5792344 Holm Aug 1998 A
5792450 Wilson et al. Aug 1998 A
5795489 Holm Aug 1998 A
5795571 Cederholm-Williams et al. Aug 1998 A
5795751 Apel Aug 1998 A
5811094 Caplan et al. Sep 1998 A
5811151 Hendriks et al. Sep 1998 A
5817519 Zelmanovic et al. Oct 1998 A
5823986 Peterson Oct 1998 A
5824084 Muschler Oct 1998 A
5830359 Knight et al. Nov 1998 A
5833866 Brown Nov 1998 A
5834418 Brazeau et al. Nov 1998 A
5837150 Langley et al. Nov 1998 A
5840502 Van Vlasselaer Nov 1998 A
5842477 Naughton et al. Dec 1998 A
5846427 Kessler et al. Dec 1998 A
5853600 McNeal et al. Dec 1998 A
5860937 Cohen Jan 1999 A
5863892 Stern et al. Jan 1999 A
5865785 Bischof Feb 1999 A
5885239 Headley et al. Mar 1999 A
5889584 Wardlaw Mar 1999 A
5895346 Wells et al. Apr 1999 A
5899874 Jonsson May 1999 A
5900245 Sawhney et al. May 1999 A
5906934 Grande et al. May 1999 A
5916557 Berlowitz-tarrant et al. Jun 1999 A
5916743 Lake et al. Jun 1999 A
5918622 Perez Jul 1999 A
5924972 Turvaville et al. Jul 1999 A
5934803 Hutter Aug 1999 A
5938621 Kelly et al. Aug 1999 A
5951160 Ronk Sep 1999 A
5955032 Kelly et al. Sep 1999 A
5955436 Kunkle, Jr. Sep 1999 A
5958250 Brown et al. Sep 1999 A
5958253 Holm Sep 1999 A
5961210 McCardel et al. Oct 1999 A
5980734 Itoh Nov 1999 A
5980757 Brown et al. Nov 1999 A
5985315 Patat et al. Nov 1999 A
5997544 Nies et al. Dec 1999 A
6007811 Sawyer et al. Dec 1999 A
6010627 Hood, III Jan 2000 A
6011490 Tonnesen et al. Jan 2000 A
6020196 Hu et al. Feb 2000 A
6022306 Dumont et al. Feb 2000 A
6025201 Zelmanovic et al. Feb 2000 A
6027655 Holm Feb 2000 A
6049026 Muschler Apr 2000 A
6051146 Green et al. Apr 2000 A
6051147 Bischof Apr 2000 A
6053856 Hlavinka Apr 2000 A
6054122 MacPhee et al. Apr 2000 A
6063297 Antanavich et al. May 2000 A
6063624 Kandler et al. May 2000 A
6071421 Brown Jun 2000 A
6071422 Hlavinka et al. Jun 2000 A
6071423 Brown et al. Jun 2000 A
6090793 Zimmermann et al. Jul 2000 A
6096309 Prior et al. Aug 2000 A
6096728 Collins et al. Aug 2000 A
6102843 Kelley et al. Aug 2000 A
6117425 MacPhee et al. Sep 2000 A
6123655 Fell Sep 2000 A
6150163 McPherson et al. Nov 2000 A
6153113 Goodrich et al. Nov 2000 A
6183737 Zaleske et al. Feb 2001 B1
6196987 Holmes et al. Mar 2001 B1
6197325 MacPhee et al. Mar 2001 B1
6200287 Keller et al. Mar 2001 B1
6200606 Peterson et al. Mar 2001 B1
6214338 Antanavich et al. Apr 2001 B1
6221315 Giesler et al. Apr 2001 B1
6245900 Yamasaki et al. Jun 2001 B1
6264890 Boehringer et al. Jul 2001 B1
6274090 Coelho et al. Aug 2001 B1
6277961 Hock et al. Aug 2001 B1
6280400 Niermann Aug 2001 B1
6286670 Smith Sep 2001 B1
6287558 Lanza et al. Sep 2001 B1
6296602 Headley Oct 2001 B1
6316247 Katz et al. Nov 2001 B1
6322785 Landesberg et al. Nov 2001 B1
6327491 Franklin et al. Dec 2001 B1
6328765 Hardwick et al. Dec 2001 B1
6334842 Hlavinka et al. Jan 2002 B1
6337072 Ford et al. Jan 2002 B1
6342157 Hood, III Jan 2002 B1
6351659 Vilsmeier Feb 2002 B1
6355239 Bruder et al. Mar 2002 B1
6368298 Beretta et al. Apr 2002 B1
6368498 Guilmette Apr 2002 B1
6398972 Blasetti et al. Jun 2002 B1
6406671 Dicesare et al. Jun 2002 B1
6409528 Bodnar Jun 2002 B1
6410344 Chung Jun 2002 B1
6417004 Brady et al. Jul 2002 B1
6440444 Boyce et al. Aug 2002 B2
6444228 Baugh et al. Sep 2002 B1
6464624 Pages Oct 2002 B2
6471069 Lin et al. Oct 2002 B2
6472162 Coelho et al. Oct 2002 B1
6487992 Hollis Dec 2002 B1
6508778 Verkaart et al. Jan 2003 B1
6516953 Dicesare et al. Feb 2003 B1
6523698 Dennehey et al. Feb 2003 B1
6544162 Van et al. Apr 2003 B1
6544727 Hei Apr 2003 B1
6558341 Swisher May 2003 B1
6563953 Lin et al. May 2003 B2
6596180 Baugh et al. Jul 2003 B2
6599873 Sommer et al. Jul 2003 B1
6623472 Reincke et al. Sep 2003 B1
6623959 Harris Sep 2003 B2
6629919 Egozy et al. Oct 2003 B2
6638503 Chitte Oct 2003 B2
6649072 Brandt et al. Nov 2003 B2
6676629 Andrew et al. Jan 2004 B2
6713246 Reinecke et al. Mar 2004 B1
6716187 Jorgensen Apr 2004 B1
6719901 Dolecek et al. Apr 2004 B2
6733471 Ericson et al. May 2004 B1
6758978 Bedell Jul 2004 B1
6759188 Reinecke et al. Jul 2004 B2
6764531 Hogan Jul 2004 B2
6777231 Katz et al. Aug 2004 B1
6790371 Dolecek Sep 2004 B2
6803022 Dicesare et al. Oct 2004 B2
6811777 Mishra Nov 2004 B2
6830762 Baugh et al. Dec 2004 B2
6835353 Smith et al. Dec 2004 B2
6835377 Goldberg et al. Dec 2004 B2
RE38730 Jakary et al. Apr 2005 E
6899813 Dolecek et al. May 2005 B2
6905612 Dorian et al. Jun 2005 B2
6911202 Amir et al. Jun 2005 B2
RE38757 Jakary et al. Jul 2005 E
6979307 Beretta et al. Dec 2005 B2
7011644 Andrew et al. Mar 2006 B1
7011852 Sukavaneshvar et al. Mar 2006 B2
7077273 Ellsworth et al. Jul 2006 B2
7077827 Greenfield Jul 2006 B2
7155288 Soykan et al. Dec 2006 B2
7166283 Tsuji et al. Jan 2007 B2
7179391 Leach et al. Feb 2007 B2
7195606 Ballin Mar 2007 B2
7223346 Dorian et al. May 2007 B2
7273886 Olivero Sep 2007 B2
7354515 Coull et al. Apr 2008 B2
7374678 Leach et al. May 2008 B2
7411006 Shanbrom Aug 2008 B2
7465293 Reinecke et al. Dec 2008 B2
7470371 Dorian et al. Dec 2008 B2
7531355 Rodríguez et al. May 2009 B2
7553413 Dorian et al. Jun 2009 B2
7608258 Mishra Oct 2009 B2
7678385 Reddi Mar 2010 B2
7694828 Swift et al. Apr 2010 B2
7708152 Dorian et al. May 2010 B2
7806276 Leach et al. Oct 2010 B2
7845499 Higgins et al. Dec 2010 B2
7867765 Faustman et al. Jan 2011 B2
7901344 Yoo Mar 2011 B2
7901584 Dorian et al. Mar 2011 B2
7914689 Higgins et al. Mar 2011 B2
7987995 Dorian et al. Aug 2011 B2
7992725 Leach et al. Aug 2011 B2
8048297 Leach et al. Nov 2011 B2
8048321 Leach et al. Nov 2011 B2
8062534 Higgins et al. Nov 2011 B2
8067534 Jagota Nov 2011 B2
8202539 Behnam et al. Jun 2012 B2
8567609 Landrigan et al. Oct 2013 B2
8596470 Leach et al. Dec 2013 B2
8753690 Higgins et al. Jun 2014 B2
8783470 Hecker et al. Jul 2014 B2
8801586 Dorian et al. Aug 2014 B2
8808551 Leach et al. Aug 2014 B2
8950586 Dorian et al. Feb 2015 B2
8992862 Leach et al. Mar 2015 B2
9011800 Leach et al. Apr 2015 B2
9556243 Leach et al. Jan 2017 B2
20010009757 Bischof et al. Jul 2001 A1
20010053764 Sims et al. Dec 2001 A1
20020009454 Boone et al. Jan 2002 A1
20020032112 Pages Mar 2002 A1
20020035820 Farris Mar 2002 A1
20020076400 Katz et al. Jun 2002 A1
20020077276 Fredeking et al. Jun 2002 A1
20020082220 Hoemann et al. Jun 2002 A1
20020090711 Karlsson Jul 2002 A1
20020104808 Blasetti et al. Aug 2002 A1
20020114775 Pathak Aug 2002 A1
20020119179 Rezania et al. Aug 2002 A1
20020161449 Muschler Oct 2002 A1
20020169408 Beretta et al. Nov 2002 A1
20020172666 Sacchi et al. Nov 2002 A1
20020182664 Dolecek et al. Dec 2002 A1
20020192632 Hei et al. Dec 2002 A1
20030033021 Plouhar et al. Feb 2003 A1
20030033022 Plouhar et al. Feb 2003 A1
20030050709 Noth et al. Mar 2003 A1
20030050710 Petersen et al. Mar 2003 A1
20030055511 Schryver et al. Mar 2003 A1
20030082152 Hedrick et al. May 2003 A1
20030091536 Frisbie et al. May 2003 A1
20030099650 Ho et al. May 2003 A1
20030138910 Reinecke et al. Jul 2003 A1
20030185803 Kadiyala et al. Oct 2003 A1
20030191429 Andrew et al. Oct 2003 A1
20030194397 Mishra Oct 2003 A1
20030198687 Bennett Oct 2003 A1
20030205538 Dorian et al. Nov 2003 A1
20040005246 Efthimiadis et al. Jan 2004 A1
20040013575 Stevens et al. Jan 2004 A1
20040120942 McGinnis et al. Jun 2004 A1
20040171146 Katz et al. Sep 2004 A1
20040182395 Brookman Sep 2004 A1
20040182788 Dorian et al. Sep 2004 A1
20040182795 Dorian et al. Sep 2004 A1
20040219182 Gomes et al. Nov 2004 A1
20040251217 Leach et al. Dec 2004 A1
20040258671 Watkins Dec 2004 A1
20050059589 Mullarkey Mar 2005 A1
20050076396 Katz et al. Apr 2005 A1
20050084961 Hedrick et al. Apr 2005 A1
20050084962 Simon Apr 2005 A1
20050100536 Mishra May 2005 A1
20050109716 Leach et al. May 2005 A1
20050130301 McKay et al. Jun 2005 A1
20050145187 Gray Jul 2005 A1
20050152905 Omoigui Jul 2005 A1
20050153441 Hedrick et al. Jul 2005 A1
20050153442 Katz et al. Jul 2005 A1
20050186120 Dorian et al. Aug 2005 A1
20050196393 Shanbrom Sep 2005 A1
20050196874 Dorian et al. Sep 2005 A1
20050197293 Mellis et al. Sep 2005 A1
20050247715 Ellsworth et al. Nov 2005 A1
20050260174 Fraser et al. Nov 2005 A1
20050260175 Hedrick et al. Nov 2005 A1
20050282275 Katz et al. Dec 2005 A1
20060046960 McKay et al. Mar 2006 A1
20060051865 Higgins et al. Mar 2006 A1
20060057223 DiMauro et al. Mar 2006 A1
20060057693 Simon Mar 2006 A1
20060083720 Fraser et al. Apr 2006 A1
20060121002 Rolland et al. Jun 2006 A1
20060140923 Evangelista Jun 2006 A1
20060151384 Ellsworth et al. Jul 2006 A1
20060171948 Weinstein et al. Aug 2006 A1
20060175242 Dorian et al. Aug 2006 A1
20060175244 Dorian et al. Aug 2006 A1
20060175268 Dorian et al. Aug 2006 A1
20060178610 Nowakowski Aug 2006 A1
20060196885 Leach et al. Sep 2006 A1
20060243676 Swift et al. Nov 2006 A1
20060263407 Mishra Nov 2006 A1
20060273049 Leach et al. Dec 2006 A1
20060273050 Higgins et al. Dec 2006 A1
20060278588 Woodell-May Dec 2006 A1
20070027082 Hasty et al. Feb 2007 A1
20070034579 Dorian et al. Feb 2007 A1
20070036768 Fraser et al. Feb 2007 A1
20070075016 Leach Apr 2007 A1
20070092494 Higgins et al. Apr 2007 A1
20070207161 Ralph Sep 2007 A1
20070208321 Leach et al. Sep 2007 A1
20080011684 Dorian et al. Jan 2008 A1
20080019964 Olmarker et al. Jan 2008 A1
20080064626 Zanella Mar 2008 A1
20080145834 Ho Jun 2008 A1
20080164204 Hatamian et al. Jul 2008 A1
20080173593 Coull et al. Jul 2008 A1
20080193424 McKale et al. Aug 2008 A1
20080217263 Higgins et al. Sep 2008 A1
20080217264 Leach et al. Sep 2008 A1
20080217265 Leach et al. Sep 2008 A1
20080268064 Woodell-May Oct 2008 A1
20080269762 Simon et al. Oct 2008 A1
20080283474 Leach et al. Nov 2008 A1
20080306431 Yoo Dec 2008 A1
20080318317 Roche et al. Dec 2008 A1
20090014391 Leach et al. Jan 2009 A1
20090018313 Shanbrom Jan 2009 A1
20090047242 Reinecke et al. Feb 2009 A1
20090101599 Dorian et al. Apr 2009 A1
20090112146 Wratten et al. Apr 2009 A1
20090181019 Solinger Jul 2009 A1
20090191217 de Wildt et al. Jul 2009 A1
20090192528 Higgins et al. Jul 2009 A1
20090220482 Higgins et al. Sep 2009 A1
20090221075 Dorian et al. Sep 2009 A1
20090236297 Dorian et al. Sep 2009 A1
20090250413 Hoeppner Oct 2009 A1
20090253566 Chavarria Oct 2009 A1
20090263319 Wohabrebbi et al. Oct 2009 A1
20090289014 Hoeppner Nov 2009 A1
20090317439 Turzi et al. Dec 2009 A1
20100008992 Ichim Jan 2010 A1
20100015129 Abramson et al. Jan 2010 A1
20100055087 Higgins et al. Mar 2010 A1
20100125236 Bare et al. May 2010 A1
20100140182 Chapman et al. Jun 2010 A1
20100186676 Van Der Jul 2010 A1
20100198130 Swift et al. Aug 2010 A1
20100206798 Dorian et al. Aug 2010 A1
20100226909 Hecker et al. Sep 2010 A1
20100256595 Leach et al. Oct 2010 A1
20100323870 Leach et al. Dec 2010 A1
20100324450 Leach et al. Dec 2010 A1
20110014705 Leach et al. Jan 2011 A1
20110020196 Grippi et al. Jan 2011 A1
20110021334 Leach et al. Jan 2011 A1
20110036786 Ellsworth Feb 2011 A1
20110052561 Hoeppner Mar 2011 A1
20110056893 Leach et al. Mar 2011 A1
20110059082 Germer et al. Mar 2011 A1
20110059083 Aigner et al. Mar 2011 A1
20110059084 Osterroth et al. Mar 2011 A1
20110065183 Dorian et al. Mar 2011 A1
20110077596 Higgins et al. Mar 2011 A1
20110129441 Lentz Jun 2011 A1
20110168193 Leach et al. Jul 2011 A1
20110189172 Solinger et al. Aug 2011 A1
20110192804 Landrigan et al. Aug 2011 A1
20110251041 Chavarria et al. Oct 2011 A1
20110268708 Lin et al. Nov 2011 A1
20120015796 Leach et al. Jan 2012 A1
20120027746 Dorian et al. Feb 2012 A1
20120093936 Lindenberg et al. Apr 2012 A1
20120145652 Leach et al. Jun 2012 A1
20120150086 Cohen Jun 2012 A1
20120172836 Higgins et al. Jul 2012 A1
20120228203 Hecker et al. Sep 2012 A1
20130068676 Leach et al. Mar 2013 A1
20130102452 Leach et al. Apr 2013 A1
20130119549 Cheng et al. May 2013 A1
20130178425 Higgins et al. Jul 2013 A1
20130196425 Dorian et al. Aug 2013 A1
20130259951 O'Connell, Jr. Oct 2013 A1
20130294983 Dorian et al. Nov 2013 A1
20140051061 Landrigan et al. Feb 2014 A1
20140054246 Landrigan et al. Feb 2014 A1
20140091048 Leach et al. Apr 2014 A1
20140242045 Higgins et al. Aug 2014 A1
20140271587 Landrigan et al. Sep 2014 A1
20140271588 Landrigan et al. Sep 2014 A1
20140271589 Matuska et al. Sep 2014 A1
20140271870 O'Shaughnessey et al. Sep 2014 A1
20140274893 Woodell-May et al. Sep 2014 A1
20140274894 Leach et al. Sep 2014 A1
20140275497 Leach et al. Sep 2014 A1
20140349388 Dorian et al. Nov 2014 A1
20140356446 Leach et al. Dec 2014 A1
20150141332 Toler May 2015 A1
20150147300 Woodell-May et al. May 2015 A1
Foreign Referenced Citations (114)
Number Date Country
696278 Sep 1998 AU
748575 Jun 2002 AU
9103724 Mar 1993 BR
1321138 Aug 1993 CA
2182862 Jun 1996 CA
2448415 Dec 2002 CA
1074709 Jul 1993 CN
1321103 Nov 2001 CN
1322146 Nov 2001 CN
103702729 Apr 2014 CN
105209478 Dec 2015 CN
105338990 Feb 2016 CN
105339007 Feb 2016 CN
105358161 Feb 2016 CN
105358162 Feb 2016 CN
56103 Oct 1960 DE
1443359 Nov 1968 DE
4202667 May 1993 DE
090997 Oct 1983 EP
0102773 Mar 1984 EP
0109374 May 1984 EP
0142339 May 1985 EP
0244834 Nov 1987 EP
0253198 Jan 1988 EP
0295771 Dec 1988 EP
0417818 Mar 1991 EP
0534178 Mar 1993 EP
0592242 Apr 1994 EP
1005910 Jun 2000 EP
1006360 Jun 2000 EP
1289618 Mar 2003 EP
1427279 Jun 2004 EP
1467746 Oct 2004 EP
1509326 Mar 2005 EP
1670315 Jun 2006 EP
1716901 Nov 2006 EP
1406492 Dec 2009 EP
2186877 May 2010 EP
2968412 Jan 2016 EP
854715 Nov 1960 GB
60053845 Mar 1985 JP
60250014 Dec 1985 JP
2036872 Feb 1990 JP
02071747 Mar 1990 JP
02129224 May 1990 JP
2000189407 Jul 2000 JP
2000199760 Jul 2000 JP
2004305439 Nov 2004 JP
2005013783 Jan 2005 JP
2005098704 Apr 2005 JP
2005524451 Aug 2005 JP
2006305365 Nov 2006 JP
2006527025 Nov 2006 JP
2008104798 May 2008 JP
2009155234 Jul 2009 JP
WO-8400905 Mar 1984 WO
WO-8802259 Apr 1988 WO
WO-9010031 Sep 1990 WO
WO-9108285 Jun 1991 WO
WO-9222312 Dec 1992 WO
WO-9305067 Mar 1993 WO
WO-9308904 May 1993 WO
WO-9407548 Apr 1994 WO
WO-9617871 Jun 1996 WO
WO-9848938 Nov 1998 WO
9905989 Feb 1999 WO
9967277 Dec 1999 WO
WO-0061256 Oct 2000 WO
WO-0074713 Dec 2000 WO
WO-0103756 Jan 2001 WO
WO-0183068 Nov 2001 WO
WO-0238610 May 2002 WO
WO-02060925 Aug 2002 WO
WO-02098566 Dec 2002 WO
WO-03015800 Feb 2003 WO
WO-03024215 Mar 2003 WO
WO-03053362 Jul 2003 WO
03063799 Aug 2003 WO
03080104 Oct 2003 WO
03088905 Oct 2003 WO
WO-03092894 Nov 2003 WO
WO-03099412 Dec 2003 WO
2004009207 Jan 2004 WO
WO-2004104553 Dec 2004 WO
WO-2005034843 Apr 2005 WO
2006043972 Apr 2006 WO
WO-2006041406 Apr 2006 WO
2007121538 Nov 2007 WO
2007128973 Nov 2007 WO
WO-2007127834 Nov 2007 WO
WO 2007142908 Dec 2007 WO
2008021237 Feb 2008 WO
WO-2008127639 Oct 2008 WO
WO-2009021257 Feb 2009 WO
WO-2009111338 Sep 2009 WO
WO-2010115190 Oct 2010 WO
WO-2011008836 Jan 2011 WO
2011031553 Mar 2011 WO
WO-2011031524 Mar 2011 WO
WO-2011031553 Mar 2011 WO
2012030593 Mar 2012 WO
WO-2012030593 Mar 2012 WO
WO2012030593 Mar 2012 WO
WO 2012030593 Mar 2012 WO
WO-2014144505 Sep 2014 WO
WO-2014144505 Sep 2014 WO
WO-2014149266 Sep 2014 WO
WO-2014149266 Sep 2014 WO
WO-2014149270 Sep 2014 WO
WO-2014149300 Sep 2014 WO
WO-2014149301 Sep 2014 WO
WO-2014149979 Sep 2014 WO
WO-2014150375 Sep 2014 WO
WO-2014150375 Sep 2014 WO
Non-Patent Literature Citations (337)
Entry
O'Shaughnessey et al. Blood-derived anti-inflammatory protein solution blocks the effect of IL-1 beta on human macrophages in vitro. Inflammation Research. Published online Jun. 18, 2011; 60(10): 929-36.
Vangsness et al. Stimulation of IL-1ra production from platelet-rich plasma. Poster No. 488 presented at 54th Annual Meeting of the Orthopedic Research Society, Mar. 2-5, 2008.
Carpenter et al. Long-term storage of proteins. Current Protocols in Protein Science. 2002; 4.6.1-4.6.6.
Carpenter et al. 2002. Rationale design of stable lyophilized protein formulations: theory and practice. in “Rationale Design of stable protein formulations—theory and practice” (J.F. Carpenter and M.C. Manning eds.) Kluwer Academic/Plenum publishers, New York, pp. 109-133.
Nakajima et al. Bioactivity of freeze-dried platelet-rich plasma in an adsorbed form on a biodegradable polymer material. Platelets 2012; 23(8):594-603).
Alford, J. et al. “Cartilage Restoration, Part 1” The American Journal of Sports Medicine, vol. 33, No. 2 (2005) p. 295-306.
Anitua, E. et al. “Autologous platelets as a source of proteins for healing and tissue regeneration” Thromb Haemost, vol. 91 (pp. 4-15) 2004.
Anonymous: “Update for veterinarians” Dec. 2012. vet.osu.edu/sites/default/files/documents/pdf/news/vmc/ovmaVeternarianUp/date/20121112.pdf.
Arend, W. et al. “Interleukin-1 Receptor Antagonist: Role in Biology” Annu. Rev. Immunol., vol. 16 (pp. 27-55) 1998.
Baltzer AW, et al. Autologous conditioned serum (Orthokine) is an effective treatment for knee osteoarthritis. Osteoarthritis Cartilage Feb. 1, 2009; 17(2):152-60.
Becker C. et al. Efficacy of epidural perineural injections with autologous conditioned serum for lumbar radicular compression: an Investigator-initiated, prospective, double-blind, reference-controlled study. Spine Aug. 1, 2007; 32 (17):1803-8.
Bendele et al. “Combination benefit of treatment with the cytokine inhibitors interleukin-1 receptor antagonist and PEGylated soluble tumor necrosis factor receptor type I in animal models of rheumatoid arthritis” Arthritis & Rheumatism, vol. 43, No. 12, Dec. 2000, pp. 2648-2659.
Bielecki, T. et al, “Antibacterial effect of autologous platelet gel enriched with growth factors and toher acive substances” J Bone Joint Surg, vol. 89-B, No. 3 (p. 417-420) Mar. 2007.
Bio-Rad Laboratories. Bio-Gel P Polyacrylamide Gel Instruction Manual, Obtained from www.bio-rad.com/webmaster/pdfs/9154_Bio-Gel_P.pdf on Jun. 20, 2012 (14 pages).
Biomet Biologics, Inc. “GPS® II Platelet Concentrate System: The New Gold Standard” Product Brochure (14 pages) Sep. 2006.
Biomet Biologics, Inc. “GPS® III Platelet Separation System” Product Brochure (8 pages) 2007.
Biomet Biologics, Inc. “Plasmax Plasma Concentrate” Product Brochure (6 pages) 2006.
Biomet Biologics, Inc. “Vortech Concentration System Product” Product Brochure (16 pages) Aug. 2005.
Biomet Biologics, Inc. “GPS System Shoulder Recovery with the GPS Platelet Concentrate System” Product Brochure (6 pages) 2004.
Burnouf, T. “Blood-derived, tissue engineering biomaterials” Biomedical Engineering—Applications, Basis & Communications, vol. 16, No. 6, Dec. 2004 (pp. 294-304).
Cell Factor Technologies, Inc. “GPS® Platelet Concentrate System” Product Brochure (9 pages) 2004.
Cell Factor Technologies, Inc., Biomet Europe. “GPS® II System, Gravitational Platelet Separation System” User Manual (13 pages), http://www.cellfactortech.com/global_products.cfm, printed Sep. 16, 2005.
Cell Factor Technologies, Inc., Biomet Europe. “GPS® II System, Gravitational Platelet Separation System, Accelerating the Body's Natural Healing Process” Product Bruchure (16 pages) 2005.
Dallari et al. “Enhanced Tibial Osteotomy Healing with Use of Bone Grafts Supplemented with Platelet Gel or Platelet Gel and Bone Marrow Stromal Cells” The Journal of Bone and Joint Surgery, vol. 89 (2007) pp. 2413-2420.
Dinarello, C. “Interleukin-1 and Interleukin-1 Antagonism” Blood, vol. 77, No. 8 (pp. 1627-1652) Apr. 1991.
Dinarello, C. A. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood, 2011, vol. 117 (14), p. 3720-3732.
Eppley, et al. “Platelet Quantification and Growth Factor Analysis from Platelet-Rich Plasma: Implications for Wound Healing,” Plastic and Reconstructive Surgery, 114(6):1502-1508 (Nov. 2004).
Evans, C.H.Novel biological approaches to the intra-articular treatment of osteoarthritis. BioDrugs 2005; 19(6):355-62.
Fiotti et al. “Atherosclerosis and Inflammation. Patterns of Cytokine Regulation in Patients with Peripheral Arterial Disease” Atherosclerosis, Elsevier Ireland Ltd, IE, vol. 145, No. 1, pp. 51-60. Jul. 1, 1999.
Floryan, K. et al. “Home Study Program: Intraoperative use of Autologous Platelet-Rich and Platelet-Poor Plasma for Orthopedic Surgery Patients” vol. 80, No. 4 (Oct. 2004) p. 667-674.
Hou, WH et al. “Microfluidic Devices for Blood Fractionation” Micromachines (2011) 2, 319-343.
Juge-Aubry, C. et al. “Adipose Tissue is a Major Source of Interleukin-1 Receptor Antagonist” Diabetes, vol. 52, May 2003 (pp. 1104-1110).
Kaufman, A. et al. “Human macrophage response to UHMWPE, TiAlV, CoCr, and alumina particles: Analysis of multiple cytokines using protein arrays” Journal of Biomedical Materials Research Part A, published online in Wiley InterScience DOI: 10.1002/jbm.a.31467 (pp. 464-474) Jul. 2007.
Kim, Seon Hee et al. “Ex vivo gene delivery of II-1Ra and soluble TNF receptor confers a distal synergistic therapeutic effect in antigen-induced arthritis”, Molecular Therapy, vol. 6, No. 5, Nov. 1, 2002 (pp. 591-600).
King, W. et al. “A simple method to Correlate the Concentration of an Anti-Inflammatory Cytokine with White Blood Cells in an Autologous Protein Solution” Feb. 24, 2014.
Klingenberg et al. “Treating inflammation in Atherosclerotic Cardiovascular Disease: Emerging Therapies” European Heart Journal., vol. 30, No. 23, pp. 2838-2844, Dec. 2009.
Lavi, G. et al. “Sustained delivery of IL-1Ra from biodegradable microspheres reduces the number of murine B16 melanoma lung metastases” Journal of Controlled Release. 123, 123-130 (2007).
Lucarelli, E. et al. “Platelet-derived growth factors enhance proliferation of human stromal stem cells” Biomaterials, vol. 24 (2003) pp. 3095-3100.
Matthews, J. et al. “Comparison of the response of primary human peripheral blood mononuclear phagocytes from different donors to challenge with model polyethylene particles of known size and dose” Biomaterials, vol. 21 (pp. 2033-2044) 2000.
Meijer, H. et al. “The production of antiinflammatory cytokines in whole blood by physico-chemical induction” Inflamm. Res., vol. 52 (pp. 404-407) Oct. 2003.
Miltenyi Biotec GmbH, Isolation of Granulocytes From Human Peripheral Blood by Density Gradient Centrifugation (2008) 2 pages.
Morizaki et al. “The Effects of Platelet-Rich Plasma on Bone Marrow Stromal Cell Transplants for Tendon Healing in Vitro” J. Hand Surg. Am., vol. 35, No. 11 (Nov. 2010) pp. 1833-1841.
Murphy et al. “Autologous Bone Marrow Mononuclear Cell Therapy is Safe and Promotes Amputation-free Survival in Patients with Critical Limb Ischemia” Journal of Vascular Surgery, C.V. Mosby Co., vol. 53, No. 6, Jan. 28, 2011.
Muzio, M. et al. “Interleukin-13 Induces the Production of Interleukin-1 Receptor Antagonist (IL-1ra) and the Expression of the mRNA for the Intracellular (Keratinocyte) Form of IL-1ra in Human Myelomonocytic Cells” Blood, vol. 83, No. 7 (pp. 1738-1743) Apr. 1994.
Nursen Düzgün et al. “Cytokine inhibitors: soluble tumor necrosis factor receptor 1 and interleukin-1 receptor antagonist in Behçet's disease” Rheumatology International ; Clinical and Experimental Investigations, Springer, Berlin, DE vol. 25, No. 1, Jan. 2005. p. 1-5.
O'Shaughnessey, K.M. et al. Blood-derived anti-inflammatory protein solution blocks the effect of IL-1beta on human macrophages in vitro. Inflamm Res Oct. 2011; 60(10):929-36.
Plasmax® Plasma Concentration System. 2007. Biomet Biologics. p. 1-20.
Rader, C. et al.“Cytokine Response of Human Macrophage-like Cells After Contact With Polyethylene and Pure Titanium Particles” The Journal of Arthroplasty, vol. 14, No. 7 pp. 840-848 (Oct. 1999).
Sorbera L A “Pegsunercept. Pegylated Soluble Tumor Necrosis Factor Receptor Type 1 PEG-STNF-RI” Drugs of the Future, Prous Science, ES, vol. 28, No. 12. Jan. 1, 2003. p. 1182-1188.
Swift, M. et al. “Characterization of Growth Factors in Platelet Rich Plasma” Cell Factor Technologies, Inc. Printed Sep. 16, 2005 from www.cellfactortech.com/global_products.cfm.
Tateishi-Yuyama, E. et al. “Therapuetic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-barrow cells: a pilot study and randomised controlled trial” The Lancet 2002; 360:427-435.
Ulich, T.R. et al. “Intratrachael Administration of Endotoxin and Cytokines: IV. The Soluble Tumor Necrosis Factor Receptor Type 1 Inhibits Acute Inflammation” American Journal of Pathology; vol. 142, No. 5, May 1993.
Vangsness, T. et al. “Stimulation of IL-1ra Production from Platelet-Rich Plasma” Poster No. 488 presented at 54th Annual Meeting of the Orthopeadic Research Society in San Francisco, CA (1 page) Mar. 2-5, 2008.
Woodell-May, J. et al. “Effect of Incubation Time on Production of IL-1ra and sTNF-RI from Platelet-Rich Plasma” Paper No. 200, 55th Annual Meeting of the Orthopaedic Research Society (1 page) Feb. 2009.
Woodell-May, J. et al. “Elution of IL-1ra from a concentrated-plasma matrix—An in vitro study” Poster Presentation at 8th World Congress of the International Cartilage Repair Society, Miami, FL (1 page) May 2009.
Woodell-May, J. et al. “Producing Accurate Platelet Counts for Platelet Rich Plasma: Validation of a Hematology Analyzer and Preparation Techniques for Counting” Scientific Foundation, Journal of Carniofacial Surgery, vol. 16, No. 5 (pp. 749-756) Sep. 2005.
Woodell-May, J. et al. Autologous protein solution inhibits MMP-13 production by IL-1beta and TNFalpha-stimulated human articular chondrocytes. J Orthop Res Sep. 15, 2011; 29:1320-6.
Wright-Carpenter, T. “Treatment of Muscle Injuries by Local Administration of Autologous Conditioned Serum: A Pilot Study on Sportsmen with Muscle Strains” Int J Sports Med, vol. 25 (pp. 588-593) Oct. 2004.
Yang, S. et al. “Protective effects of IL-1Ra or vIL-10 gene transfer on a murine model of wear debris-induced osteolysis” Gene Therapy, vol. 11 (pp. 483-491) 2004.
Yang, T. et al. “Recent Applications of Polyacrylamide as Biomaterials” Recent Patents on Materials Science, vol. 1 (pp. 29-40) 2008.
Yoshida S. et al. “Elevation of serum soluble tumour necrosis factor (TNF) receptor and IL-1 receptor antagonist levels in bronchial asthma” Clinical and Experimental Immunology, Wiley-Blackwell Publishing Ltd. vol. 106, No. 1, Oct. 1996.
Zhang et al.“IL-1ra alleviates inflammatory hyperalgesia through preventing phosphorylation of NMDA receptor NR-1 subunit in rats” Pain. vol. 135, No. 3, Mar. 5, 2008, pp. 232-239.
“International Application Serial No. PCT/US2014/016900, International Search Report dated May 12, 2014”, 5 pgs.
“International Application Serial No. PCT/US2014/016900, Written Opinion dated May 12, 2014”.
“A phase I safety study of combination treatment with pegylated soluble tumor necrosis factor receptor type I (PET STNF-RI) and anakinra (interleukin-1 receptor antagonist, IL-1RA) in patients with rheumatoid arthritis”, Prous integrity, (Jun. 12, 2002), 1-1.
“U.S. Appl. No. 12/101,586, Final Office Action dated Feb. 3, 2011”, 11 pgs.
“U.S. Appl. No. 12/101,586, Non Final Office Action dated Sep. 20, 2010”, 12 pgs.
“U.S. Appl. No. 12/101,586, Notice of Allowance dated Mar. 24, 2011”, 5 pgs.
“U.S. Appl. No. 12/101,594, Final Office Action dated Mar. 18, 2010”, 8 pgs.
“U.S. Appl. No. 12/101,594, Non Final Office Action dated Oct. 16, 2009”, 8 pgs.
“U.S. Appl. No. 12/101,594, Notice of Allowance dated May 27, 2010”, 7 pgs.
“U.S. Appl. No. 12/897,401, Non Final Office Action dated Nov. 16, 2010”, 9 pgs.
“U.S. Appl. No. 12/897,401, Notice of Allowance dated Oct. 18, 2011”, 6 pgs.
“U.S. Appl. No. 13/837,005, Final Office Action dated Dec. 5, 2014”, 9 pgs.
“U.S. Appl. No. 13/837,005, Non Final Office Action dated Feb. 17, 2016”, 13 pgs.
“U.S. Appl. No. 13/837,005, Non Final Office Action dated May 13, 2014”, 10 pgs.
“U.S. Appl. No. 13/837,005, Non Final Office Action dated Jun. 9, 2015”, 13 pgs.
“U.S. Appl. No. 13/837,005, Response filed Jan. 3, 2014 to Restriction Requirement dated Dec. 3, 2013”, 4 pgs.
“U.S. Appl. No. 13/837,005, Response filed Mar. 5, 2015 to Final Office Action dated Dec. 5, 2014”, 11 pgs.
“U.S. Appl. No. 13/837,005, Response filed Aug. 13, 2014 to Non Final Office Action dated May 13, 2014”, 13 pgs.
“U.S. Appl. No. 13/837,005, Response filed Nov. 9, 2015 to Non Final Office Action dated Jun. 9, 2015”, 11 pgs.
“U.S. Appl. No. 13/837,005, Restriction Requirement dated Dec. 3, 2013”, 9 pgs.
“U.S. Appl. No. 13/837,480, Non Final Office Action dated Aug. 11, 2015”, 10 pgs.
“U.S. Appl. No. 13/837,480, Response filed Jan. 11, 2016 to Non Final Office Action dated Aug. 11, 2015”, 14 pgs.
“U.S. Appl. No. 13/837,480, Response filed Nov. 5, 2014 to Restriction Requirement dated Sep. 16, 2014”, 3 pgs.
“U.S. Appl. No. 13/837,480, Restriction Requirement dated Sep. 16, 2014”, 6 pgs.
“U.S. Appl. No. 13/839,280, Final Office Action dated Apr. 10, 2015”, 17 pgs.
“U.S. Appl. No. 13/839,280, Non Final Office Action dated Apr. 7, 2016”, 16 pgs.
“U.S. Appl. No. 13/839,280, Non Final Office Action dated Jul. 17, 2014”, 12 pgs.
“U.S. Appl. No. 13/839,280, Response filed Mar. 17, 2014 to Restriction Requirement dated Jan. 15, 2014”, 5 pgs.
“U.S. Appl. No. 13/839,280, Response filed Oct. 12, 2015 to Final Office Action dated Apr. 10, 2015”, 9 pgs.
“U.S. Appl. No. 13/839,280, Response filed Oct. 17, 2014 to Non Final Office Action dated Jul. 17, 2014”, 19 pgs.
“U.S. Appl. No. 13/839,280, Restriction Requirement dated Jan. 15, 2014”, 6 pgs.
“U.S. Appl. No. 13/840,129, Final Office Action dated Jun. 18, 2015”, 9 pgs.
“U.S. Appl. No. 13/840,129, Non Final Office Action dated Oct. 23, 2014”, 8 pgs.
“U.S. Appl. No. 13/840,129, Response filed Feb. 23, 2015 to Non Final Office Action dated Oct. 23, 2014”, 15 pgs.
“U.S. Appl. No. 13/840,129, Restriction Requirement dated Mar. 14, 2014”, 6 pgs.
“U.S. Appl. No. 13/841,083, Examiner Summary Received Jan. 29, 2016”, 1 pg.
“U.S. Appl. No. 13/841,083, Final Office Action dated Sep. 9, 2016”, 10 pgs.
“U.S. Appl. No. 13/841,083, Non Final Office Action dated Jan. 29, 2016”, 11 pgs.
“U.S. Appl. No. 13/841,083, Non Final Office Action dated Jul. 15, 2015”, 8 pgs.
“U.S. Appl. No. 13/841,083, Non Final Office Action dated Dec. 10, 2014”, 12 pgs.
“U.S. Appl. No. 13/841,083, Response filed Apr. 10, 2015 to Non Final Office Action dated Dec. 10, 2014”, 17 pgs.
“U.S. Appl. No. 13/841,083, Response filed Apr. 28, 2016 to Non Final Office Action dated Jan. 29, 2016”, 11 pgs.
“U.S. Appl. No. 13/841,083, Response filed Aug. 27, 2014 to Restriction Requirement dated Jul. 21, 2014”, 3 pgs.
“U.S. Appl. No. 13/841,083, Response filed Oct. 13, 2015 to Non Final Office Action dated Jul. 15, 2015”, 10 pgs.
“U.S. Appl. No. 13/841,083, Response filed Nov. 29, 2016 to Final Office Action dated Sep. 9, 2016”, 12 pgs.
“U.S. Appl. No. 13/841,083, Restriction Requirement dated Jul. 21, 2014”, 6 pgs.
“U.S. Appl. No. 13/841,103, Final Office Action dated Aug. 13, 2015”, 13 pgs.
“U.S. Appl. No. 13/841,103, Non Final Office Action dated Dec. 4, 2014”, 10 pgs.
“U.S. Appl. No. 13/841,103, Response filed Jan. 13, 2016 to Final Office Action dated Aug. 13, 2015”, 11 pg.
“U.S. Appl. No. 13/841,103, Response filed Apr. 18, 2016 to Restriction Requirement dated Feb. 19, 2016”, 8 pgs.
“U.S. Appl. No. 13/841,103, Response filed May 4, 2015 to Non Final Office Action dated Dec. 4, 2014”, 18 pgs.
“U.S. Appl. No. 13/841,103, Response filed Aug. 27, 2014 to Restriction Requirement dated Jul. 21, 2014”, 3 pgs.
“U.S. Appl. No. 13/841,103, Restriction Requirement dated Feb. 19, 2016”, 7 pgs.
“U.S. Appl. No. 13/841,103, Restriction Requirement dated Jul. 21, 2014”, 6 pgs.
“U.S. Appl. No. 14/050,950, Final Office Action dated Jun. 17, 2016”, 9 pgs.
“U.S. Appl. No. 14/050,950, Non Final Office Action dated Nov. 19, 2015”, 13 pgs.
“U.S. Appl. No. 14/050,950, Notice of Allowance dated Oct. 6, 2016”, 12 pgs.
“U.S. Appl. No. 14/050,950, Response filed Feb. 19, 2016 to Non Final Office Action dated Nov. 19, 2015”, 11 pgs.
“U.S. Appl. No. 14/050,950, Response filed Jun. 23, 2015 to Restriction Requirement dated Apr. 23, 2015”, 1 pgs.
“U.S. Appl. No. 14/050,950, Response filed Aug. 17, 2016 to Final Office Action dated Jun. 17, 2016”, 8 pgs.
“U.S. Appl. No. 14/050,950, Restriction Requirement dated Apr. 23, 2015”, 7 pgs.
“U.S. Appl. No. 14/841,086, Examiners Interview Summary dated Nov. 7, 2016”, 3 pgs.
“U.S. Appl. No. 14/973,913, Preliminary Amendment filed Mar. 2, 2016”, 10 pgs.
“U.S. Appl. No. 13/840,129, Response filed May 14, 2014 to Restriction Requirement dated Mar. 14, 2014”, 3 pgs.
“Arthritis”, [Online]. Retrieved from the Internet: Wayback Machine <URL:http://www.mayoclinic.org/diseases-conditions/arthritis/basics/treatment/con-20034095 >, (2014), 5 pgs.
“Bio-Rad Laboratories. Bio-Gel P Polyacrylamide Gel Instruction Manual”, [Online]. Retrieved from the Internet: <www.bio-rad.com/webmaster/pdfs/9154 Bio-Gel P.pdf>, (Jun. 20, 2012), 14 pgs.
“BioCUE™ Platelet Concentration System”, (Jun. 2010), 2 pgs.
“Canadian Application Serial No. 2,905,552, Voluntary Amendment filed Sep. 11, 2015”.
“Canadian Application Serial No. 2,906,310, Voluntary Amendment filed Sep. 14, 2015”, 2 pgs.
“Caps for Corning® and Costar®, Plastic Labware”, Technical Bulletin, (Dec. 2008), 2 pgs.
“Cell Isolation Techniques, Methods and Materials, Working with Enzymes”, Worthington Biochemical Corp, (2004), 9 pgs.
“Cell Isolation Theory, Tissue Types”, Worthington Biochemical Corp, (2004), 5 pgs.
“Centrifuge Tubes”, Corning Costar, (1996/1997), 76-77.
“Chinese Application Serial No. 201080019707.7, Office Action dated Jun. 30, 2014”, in English, 7 pgs.
“Chinese Application Serial No. 201280030026.X, Office Action dated Nov. 21, 2014”, w/ English Translation, 27 pgs.
“Chinese Application Serial No. 201480027157.1, Voluntary Amendment filed Jun. 6, 2016”, w/English Claims, 63 pgs.
“Clotalyst® Autologous Clotting Factor”, “Would you like to have an autologous thrombin for rapid clotting and haemostasis?” Biomet Biologics, (Jan. 2007), 16 pgs.
“Corning® 15 and 50 ml Centrifuge Tubes”, Life Sciences. Corning Incorporated., (Jun. 2005), 2 pgs.
“Cytori Celution Cell Concentrate Device”, Exhibit 14, 501(k) Summary, FDA approval K060482, (Sep. 28, 2006), 7 pgs.
“European Application Serial No. 10712677.3, Examination Notification Art. 94(3) dated Jun. 5, 2013”, 5 pgs.
“European Application Serial No. 14709014.6, Office Action dated Nov. 19, 2015”, 2 pgs.
“European Application Serial No. 14709803.2, Communication Pursuant to Article 94(3) EPC dated Jul. 8, 2016”, 7 pgs.
“European Application Serial No. 14709803.2, Response filed May 16, 2016 to Communication pursuant to Rules 161(1) and 162 EPC dated Nov. 5, 2015”, 14 pgs.
“European Application Serial No. 14709803.2, Response filed Nov. 15, 2016 to Communication Pursuant to Article 94(3) EPC dated Jul. 8, 2016”, 18 pgs.
“European Application Serial No. 14724817.3, Office Action dated Oct. 27, 2015”, 2 pgs.
“Fibrostik™ Plasma Concentrator”, Attention Operating Surgeon, Cell Factor Technologies, Inc., (Jul. 2003), 2 pgs.
“Frequently Asked Questions, 1. Kits, 2. Enzymes”, Worthington Biochemical Corp, (2003), 3 pgs.
“GPS® III Platelet Separation System, Leadership through Technology”, Biomet Biologics, Inc, (Jul. 2007), 8 pgs.
“Hemocor HPH® Hemoconcentrator”, Minntech® Filtration Technologies Group, Minntech Corporation (2004), <http://www.minntech.com/ftg/products/hph/index.html>, (Jul. 15, 2004), 2 pgs.
“Increasing bone graft bioactivity through reproducible concentrations of natural growth factors”, Symphony II Platelet Concentrate System/PCS brochure, (Jan. 2003), 8 pgs.
“International Application Serial No. PCT/US2003/016506, International Search Report dated Oct. 13, 2003”, 2 pgs.
“International Application Serial No. PCT/US2007/012587, International Search Report dated Nov. 6, 2007”, 2 pgs.
“International Application Serial No. PCT/US2008/004687, International Preliminary Report on Patentability dated Aug. 13, 2009”, 19 pgs.
“International Application Serial No. PCT/US2008/004687, International Search Report dated Jul. 2, 2008”, 3 pgs.
“International Application Serial No. PCT/US2008/004687, Written Opinion dated Mar. 17, 2009”, 5 pgs.
“International Application Serial No. PCT/US2008/004687, Written Opinion dated Jul. 2, 2008”, 5 pgs.
“International Application Serial No. PCT/US2009/035564, International Preliminary Examination Report dated Aug. 31, 2010”, 6 pgs.
“International Application Serial No. PCT/US2009/035564, International Search Report dated Jul. 3, 2009”, 3 pgs.
“International Application Serial No. PCT/US2009/035564, Written Opinion dated Jul. 3, 2009”, 5 pgs.
“International Application Serial No. PCT/US2010/029957, International Preliminary Report on Patentability dated Oct. 13, 2011”, 10 pgs.
“International Application Serial No. PCT/US2010/029957, International Search Report dated Jul. 30, 2010”, 6 pgs.
“International Application Serial No. PCT/US2010/029957, Written Opinion dated Jul. 30, 2010”, 9 pgs.
“International Application Serial No. PCT/US2010/041942, International Preliminary Report on Patentability dated Jan. 26, 2012”, 9 pgs.
“International Application Serial No. PCT/US2010/041942, International Search Report dated Oct. 8, 2010”, 3 pgs.
“International Application Serial No. PCT/US2010/041942, Written Opinion dated Oct. 8, 2010”, 8 pgs.
“International Application Serial No. PCT/US2011/031954, International Search Report dated Aug. 9, 2011”, 4 pgs.
“International Application Serial No. PCT/US2011/031954, Written Opinion dated Aug. 9, 2011”, 9 pgs.
“International Application Serial No. PCT/US2011/045290, International Search Report dated Nov. 7, 2011”, 5 pgs.
“International Application Serial No. PCT/US2011/045290, Written Opinion dated Nov. 7, 2011”, 5 pgs.
“International Application Serial No. PCT/US2012/034104, International Preliminary Report on Patentability dated Oct. 31, 2013”, 7 pgs.
“International Application Serial No. PCT/US2012/034104, International Search Report dated Oct. 29, 2012”, 5 pgs.
“International Application Serial No. PCT/US2012/034104, Written Opinion dated Oct. 29, 2012”, 6 pgs.
“International Application Serial No. PCT/US2013/056793, International Preliminary Report on Patentability dated Mar. 12, 2015”, 8 pgs.
“International Application Serial No. PCT/US2013/056793, International Search Report dated Dec. 5, 2013”, 3 pgs.
“International Application Serial No. PCT/US2013/056793, Written Opinion dated Dec. 5, 2013”, 6 pgs.
“International Application Serial No. PCT/US2014/016384, International Preliminary Report on Patentability dated Sep. 24, 2015”, 16 pgs.
“International Application Serial No. PCT/US2014/016384, International Search Report dated Oct. 9, 2014”, 10 pgs.
“International Application Serial No. PCT/US2014/016384, Written Opinion dated Oct. 9, 2014”, 14 pgs.
“International Application Serial No. PCT/US2014/016421, International Preliminary Report on Patentability dated Sep. 24, 2015”, 16 pgs.
“International Application Serial No. PCT/US2014/016421, International Search Report dated Jul. 24, 2014”, 8 pgs.
“International Application Serial No. PCT/US2014/016421, Written Opinion dated Jul. 24, 2014”, 14 pgs.
“International Application Serial No. PCT/US2014/016895, International Preliminary Report on Patentability dated Sep. 24, 2015”, 16 pgs.
“International Application Serial No. PCT/US2014/016895, International Search Report dated Jul. 24, 2014”, 9 pgs.
“International Application Serial No. PCT/US2014/016895, Written Opinion dated Jul. 24, 2014”, 14 pgs.
“International Application Serial No. PCT/US2014/021707, International Preliminary Report on Patentability dated Sep. 24, 2015”, 16 pgs.
“International Application Serial No. PCT/US2014/021707, International Search Report dated Sep. 24, 2014”, 8 pgs.
“International Application Serial No. PCT/US2014/021707, Written Opinion dated Jul. 24, 2014”, 14 pgs.
“International Application Serial No. PCT/US2014/023091, International Preliminary Report on Patentability dated Sep. 24, 2015”, 11 pgs.
“International Application Serial No. PCT/US2014/023091, International Search Report dated Oct. 9, 2014”, 7 pgs.
“International Application Serial No. PCT/US2014/023091, Written Opinion dated Oct. 9, 2014”, 9 pgs.
“International Application Serial No. PCT/US2014/028942, International Preliminary Report on Patentability dated Sep. 24, 2015”, 15 pgs.
“Japanese Application Serial No. 2010-503066, Office Action dated Jan. 22, 2013”, w/ English Translation, 17 pgs.
“Japanese Application Serial No. 2012-503768, Office Action dated May 20, 2014”, w/ English Translation, 5 pgs.
“Japanese Application Serial No. 2012-520742, Office Action dated Sep. 9, 2014”, w/ English Translation, 6 pgs.
“Japanese Application Serial No. 2014-024420, Preliminary Notice of Reasons for Rejection dated Feb. 24, 2015”, w/ English Translation, 15 pgs.
“Knee Cartilage Implantation Carticel™, Autologous Cultured Chondrocyte Implantation”, The Sports Medicine Center, [Online]. Retrieved from the Internet: <http://www.orthoassociates.com/carticel.htm>, (Apr. 6, 2006), 7 pgs.
“Letter CryoSeal FS System. Vaccines, Blood & Biologics”, FDA U.S. Food and Drug Administation., http://www.fda.gov/BiologicsBloodVaccines/BioodBioodProducts/ApprovedProducts/Premarket ApprovalsPMAs/ucm091631.htm, (Jul. 26, 2007), 21 pgs.
“MarrowStim™ Concentration Kit Peripheral Arterial Disease (PAD) Study”, Retriewed From Intenet : <http://www.biomet.com/patients/clinical recruitment padstudy.cfm>, (Jul. 2, 2009), 2 pgs.
“MarrowsTim™ Concentration System”, Biomet Biologics, Inc, (Feb. 15, 2008), 20 pgs.
“Medical Applications: Blood Filtration”, Minntech® Filtration Technologies Group, Minntech Corporation (2004), <http://www.minntech.com/ftg/industries/medical/blood_filter.html>, (Jul. 15, 2004), 1 pg.
“Minivalve international: duckbill valves—du 054.001 sd”, [Online]. Retrieved from the Internet: <http://www.minivalve.com/htm/DV054.htm>, 1 pg.
“Momentive Silopren*LSR 2050”, (Jun. 30, 2014), 3 pg.
“Platelet Rich Plasma (PRP)”, The Stone Clinic, (May 2006), 2 pgs.
“Prosys PRP Kit”, Tozai Holdings, Inc. EC21 Global B2B Marketplace, Retrieved From Internet : <http://www.ec21.com/product-details/Prosys-PRP-Kit--5467061.html Printed from Web>, (Jul. 18, 2011), 5 pgs.
“Renaflo® II Hemofilter”, Minntech® Filtration Technologies Group, Minntech Corporation (2004), <http://www.minntech.com/ftg/products/renaflo/index.html>, (Jul. 15, 2004), 2 pgs.
“Sigma-Aldrich® Alkaline Phosphatase (Procedure No. 85), drug fact sheet”, Sigma-Aldrich, (2003), 1-2.
“SmartPrep PRP-20 Procedure Pack—Instructions for Use”, Harvest, 12 pgs.
“ThermoGenesis Corp. to Supply Autologous Thrombin Kits to Biornet, Inc”, noblood: Transfusion Alternatives Patient Blood Mangement, [Online]. Retrieved from the Internet: <URL: http://noblood.org/forum/threads/2128-ThermoGenesis-Corp-to-Supply-Autologous-Thrombin-Kits-to-Biomet-Inc>, (Apr. 5, 2005), 3 pgs.
“Vernay Product Information Sheet, Umbrella Check Valve”, Part No. V251010200, (Jul. 2013), 2 pgs.
Andia, Isabel, et al., “Platelet-rich plasma for managing pain and inflammation in osteoarthritis”, Nature Reviews Rheumatology, vol. 9. No. 12., (Oct. 1, 2013), 721-730.
Badiavas, Evangelos V., et al., “Treatment of Chronic Wounds With Bone Marrow-Derived Cells”, Arch Dermatol. 139, (Apr. 2003), 510-516.
Bang, N U, et al. “Plasma Protein Requirements for Human Platelet Aggregation”, Acad Sci, 201, (1972), 280-299.
Bendele, Alison M, et al., “Combination Benefit of Treatment With the Cytokine Inhibitors Interleukin-1 Receptor Antagonist and Pegylated Soluble Tumor Necrosis Factor Receptor Type I in Animal Models of Rheumatoid Arthritis”, Arthritis & Rheumatism, J.B. Lippincott vol. 43, No. 12, (Dec. 1, 2000), 2648-2659.
Berguer, R, et al., “Warning: fatal reaction to the use of fibrin glue in deep hepatic wounds. Case reports”, J Trauma 31, (1991), 408-411.
Berruyer, M, et al., “Immunization by bovine thrombin used with fibrin glue during cardiovascular operations. Development of thrombin and factor V inhibitors”, J Thorac Cardiovasc Sura 105, (1993), 892-7.
Boomgaard, et al., “Pooled Platelet Concentrates Prepared by the Platelet-Rich-Plasma Method and Filtered with Three Different Filters and Stored for 8 Days”, Vox Sanq, vol. 68, (Feb. 1995), 82-89.
Brodke, et al., “Bone Grafts Prepared with Selective Cell Retention Technology Heal Canine Segmental Defects as Effectively as Autograft”, SCR-Enriched Bone Grafts Heal Canine Segmental Defects, Journal of Orthopaedic Research, (May 2006), 857-866.
Casali, B, et al., “Fibrin glue from single-donation autologous plasmapheresis”, Transfusion 32, (1992), 641-643.
Clayden, J D, et al., “Improved segmentation reproducibility in group tractography using a quantitative tract similarity measure”, Neuroimage, Academic Press, Orlando, FL, US vol. 33, No. 2, (Nov. 1, 2006), 482-492.
Collier, B S, et al., “The pH Dependence of Quantitative Ristocetin-induced Platelet Aggregation: Theoretical and Practical Implications—A New Device for Maintenance of Platelet-Rich Plasma pH”, Hematology Service, Clinical Pathology Department, Clinical Center, National Institutes of Health, Bethesda Blood, vol. 47, No. 5, (May 1976).
Connolly, John, et al., “Development of an Osteogenic Bone-Marrow Preparation”, The Journal of Bone and Joint SurQery, Incorporated. vol. 71-A, No. 5, (Jun. 1989), 684-691.
Connolly, John F., “Injectable Bone Marrow Preparations to Stimulate Osteogenic Repair”, Clinical Orthopaedics and Related Research 313, (Apr. 1995), 8-18.
Dallari, et al., “In Vivo Study on the Healing of Bone Defects Treated with Bone Marrow Stromal Cells, Platelet-Rich Plasma, and Freeze-Dried Bone Allografts, Alone and in Combination”, Healing of Bone Defects, Journal of Orthopaedic Research, (May 2006), 877-888.
Dawson, J, et al., “Effects of soluble interleukin-1 type II receptor on rabbit antigen-induced arthritis: Clinical, biochemical and histological assessment”, Rheumatology (Oxford) vol. 38, No. 5, (May 5, 1999), 401-406.
De Ugarte, et al., “Comparison of Multi-Lineage Cells from Human Adipose Tissue and Bone Marrow”, Cells Tissues Organs 174, (2003), 101-109.
De Ugarte, et al., “Differential Expression of Stem Cell Mobilization-Associated Molecules on Multi-Lineage Cells from Adipose Tissue and Bone Marrow”, Immunology Letters 89, (2003), 267-270.
De Wit, et al., “Experiments on the Preparation of Blood Components with the IBM 2991 Blood Cell Processor”, Vox Sang. 29, (Feb. 10, 1975), 352-362.
Delrossi, A, et al., “Platelet-rich plasma reduces postoperative blood loss after cardiopulmonary bypass”, J Thorac Cardiovasc Sura 100, (Aug. 1990), 281-285.
Depalma, L, “The preparation of fibrinogen concentrate for use as fibrin glue by four different methods”, Transfusion vol. 33, No. 9, (1993), 717-720.
Deugarte, M D, et al., “Future of Fat as Raw Material for Tissue Regeneration”, Lippincott Williams & Wilkins, Inc., (2007), 215-219.
Dimuzio, Paul, et al., “Development of a Tissue-Engineered Bypass Graft Seeded with Stem Cells”, Vascular, vol. 14, No. 6, (2006), 338-342.
Dinarello, C, “Interleukin-1 and Interleukin-1 Antagonism”, Blood, vol. 77, No. 8, (Apr. 1991), 1627-1652.
Edlich, Richard F, et al., “Surgical Devices in Wound Healing Management”, In Wound Healing: Biochemical & Clinical Aspects 1st ed., vol. Philadelphia: W.B. Saunders Company, (1992), 581-601.
Ehricke, H H, et al., “Visualizing MR diffusion tensor fields by dynamic fiber tracking and uncertainty mapping”, Computers and Graphics, Elsevvier vol. 30, No. 2, (Apr. 1, 2006), 255-264.
Epstein, G H, et al., “A new autologous fibrinogen-based adhesive for otologic surgery”, Ann Otol Rhinol Laryngol 95, (May 25-26, 1985), 40-45.
Feige, U, et al., “Anti-interleukin-1 and anti-tumor necrosis factor-alpha synergistically inhibit adjuvant arthritis in Lewis rats”, Cmls Cellular and Molecular Li Fe Sciences, Bi Rkhauser Verlag, Heidelberg, DE, vol. 57, No. 10, (Sep. 1, 2000), 1457-1470.
Fraser, John K, et al., “Plasticity of human adipose stem cells toward endothelial cells and cardiomyocytes”, Nature Clinical Practice Cardiovascular Medicine, vol. 3, Supplement 1, (Mar. 2006), S33-S37.
Friesen, Robert, et al., “Blood Conservation During Pediatric Cardiac Surgery: Ultrafiltration of the Extracorporeal Circuit Volume After Cardiopulmonary Bypass”, Anesth, Analg, (1993), 702-707.
Galois, et al., “Cartilage Tissue Engineering: State-of-the-Art and Future Approaches”, Pathol Bioi (Paris), 53—Abstract only, (Dec. 2005), 2 pgs.
Gerald, Marx, “Mechanism of Fibrin Coagulation Based on Selective, Cation-Driven, Protofibral Association”, Biopolymers, vol. 27, (1988), 763-774.
Gibble, et al., “Fibrin glue: the perfect operative sealant”, Transfusion, 1990, vol. 30, No. 8., (1990), 741-747.
Gimble, Jeffrey M, “Adipose-Derived Stem Cells for Regenerative Medicine”, Circulation Research American Heart Association, Inc., (May 11, 2007), 1249-1260.
Gomillion, Cheryl T, et al., “Stem cells and adipose tissue engineering”, Biomaterials 27, Science Direct Elsevier, (2006), 6052-6063.
Grove, et al., “Plasticity of Bone Marrow-Derived Stem Cells”, Stem Cells: Concise Review, (Jan. 2004), 487-500.
Guilak, Farshid, et al., “Adipose-derived adult stem cells for cartilage tissue engineering”, Biorheology 41, (2004), 389-399.
Harris, E. L.V, et al., “Protein Purification Methods—A Practical Approach”, Clarification and Extraction, (1989), 7 pgs.
Hartman, A. R, et al., “Autologous whole plasma fibrin gel. Intraoperative procurement”, Arch Surg 127, (Mar. 1992), 357-359.
Hattori, et al., “Osteogenic Potential of Human Adipose Tissue-Derived Stromal Cells as an Alternative Stem Cell Source”, Cells Tissues Organs, (2004), 2-12.
Haynesworth, S E, et al., “Mitogenic Stimulation of Human Mesenchymal Stem Cells by Platelet Releasate Suggests a Mechanism for Enhancement of Bone Repair by Platelet Concentrate”, 48th Annual Meeting of the Orthopaedic Research Society Poster No. 0462, (2002), 1 pg.
Hennis, H L, et al., “Infectious disease risks of fibrin glue [letter]”, Ophthalmic Sura 23, (Sep. 1992), 1 pg.
Hernigou, et al., “Percutaneous Autologous Bone-Marrow Grafting for Nonunions. Influence of the Number and Concentration of Progenitor Cells”, Journal of Bone & Joint Surgery, (Jul. 2005), 1430-1437.
Hiromasa, Mitsuhata, et al., “An Anaphylactic Reaction to Topical Fibrin Glue”, Anesthesiology, vol. 81, No. 4, (Oct. 1994), 1074-1077.
Hom, D, et al., “Promoting Healing with Recombinant Human Platelet-Derived Growth Factor-BB in a Previously Irradiated Problem Wound”, The Laryngoscope, vol. 113, (Sep. 2003), 1566-1571.
Hood, Andrew G. et al., “Perioperative Autologous Sequestration III: A New Physiologic Glue with Wound Healing Properties”, (Jan. 1993), 126-129.
Ishida, et al., “Platelet-Rich Plasma With Biodegradable Gelatin Hydrogel Promotes Rabbit Meniscal Tissue Regeneration”, 52nd Annual Meeting of the Orthopaedic Research Society Paper No. 1035, (2006), 1 pg.
Jackson, C M, et al., “Blood coagulation”, Annu Rev Biochem 49: 765-811, (1980), 22 pgs.
Jayadev, Suprya, “Trypsinization of Adherent Cells”, (Aug. 8, 1991), 1 pg.
Johnstone, et al., “Autologous Mesenchymal Progenitor Cells in Articular Cartilage Repair”, Clinical Orthopaedics and Related Research 367S:, (Oct. 1999), S156-S162.
Jones, D K, et al., “Confidence mapping in diffusion ensor magnetic resonance imaging tractography using a bootstrap approach”, Magnetic Resonance in Medicine Wiley USA, vol. 53 , No. 5, (May 2005), 1143-1149.
Jorgensen, et al., “Stem Cells for Repair of Cartilage and Bone: The Next Challenge in Osteoarthritis and Rheumatoid Arthritis”, Annals of Rheumatic Diseases, (Aug. 2000), 5 pgs.
Karpatkin, S., “Heterogeneity of Human Platelets. VI., Correlation of Platelet Function with Platelet Volume”, Blood, vol. 51, No. 2, (Feb. 1978), 307-316.
Kim, Seon Hee, et al., “Ex vivo gene delivery of II-1 Ra and soluble TNF receptor confers a distal synergistic therapeutic effect in antigen-induced arthritis”, Molecular Therapy, vol. 6, No. 5, (Nov. 1, 2002), 591-600.
Kim, Sun Jin, et al., “Development of a novel sustained release formulation of recombinant human growth homrone using sodium hyaluronate microparticles”, Journal of Controlled Release, 2005, vol. 104 (2005), 323-335.
Kimble, Robert B, et al., “Simultaneous block of interleukin-1 and tumor necrosis factor is required to completely prevent bone loss in the early postovariectomy period”, Endocrinology, The Endocrine Society, US, vol. 136, No. 7—Abstract, (Jul. 1, 1995), 1 pg.
Kitazawa, R, et al., “Interleukin-1 Receptor Antagonist and Tumor Necrosis Factor Binding Protein Decrease Osteoclast Formation and Bone Resorption in Ovariectomized Mice”, Journal of Clinical Investigation, American Society for Clinical Investigation, US, vol. 94, No. 6, (Dec. 1, 1994), 2397-2406.
Kjaergard, H. K, et al., “A simple method of preparation of autologous fibrin glue by means of ethanol”, Surg Gynecol Obstet 175, (1992), 72-3.
Kjaergard, H. K, “Preparation of autologous fibrin glue from pericardial Blood”, Ann Thorac Sur 55, (1993), 543-4.
Kuderma, H, et al., “Die klinische Anwendung der Klebung van Nervenanastomosen mit Gerinnungssubstanzen bei der Rekonstruction verletzter peripherer Nerven”, Wein Klin Wochenschr 87—Not in English, (Aug. 15, 1975), 6 pgs.
Kumar, Vijay, et al., “Autologous Thrombin: Intraoperative Production From Whole Blood”, Journal of American Society of Extra-Corporeal Technology, JECT, 40, (2008), 94-98.
Kumar, Vijay, et al., “Stability of Human Thrombin Produced From 11 ml of Plasma Using the Thrombin Processing Device”, Journal of American Society of Extra-Corporeal Technology JECT, 37, (Mar. 2005), 390-395.
Kumar, Vijay, et al., “Whole Blood Thrombin: Development of a Process for Intra-Operative Production of Human Thrombin”, Journal of American Society of Extra-Corporeal Technology JECT, 39, (Jan. 1, 2007), 18-23.
Kyosti Laitakari, M D, et al., “Autologous and Homologous Fibrinogen Sealants: Adhesive Strength”, Laryngoscope vol. 99, (Sep. 1989), 974-976.
Lasher, Lisa, “My Experience with PRP”, PowerPoint presentation, <http://www.cellfactortech.com/global_products.cfm>, (Sep. 16, 2005), 35 pgs.
Lendeckel, Stefan, et al., “Autologous stem cells (adipose) and fibrin glue used to treat widespread traumatic calvarial defects: case report”, Journal of Cranio-Maxillofacial Surgery (2004) European Association for Cranio-Maxillofacial Surgery, (2004), 370-373.
Lerner, R, et al., “Current status of surgical adhesives”, J Surg Res 48, (Feb. 1990), 165-80.
Longas, Maria O, “An Improved Method for the Purification of Human Fibrinogen”, J. Biochem vol. 11, (1980), 559-564.
Lori, N F, et al., “Diffusion tensor fiber tracking of human brain connectivity: acquisition methods, reliability analysis and biological results”, NMR in Biomedicine Wiley UK, vol. 15, No. 7-8, (Nov. 2002), 493-515.
Lu, X, et al., “Bone Marrow Mesenchymal Stem Cells: Progress in Bone/Cartilage Defect Repair”, 19(1) Abstract, (Jan. 2002), 2 pgs.
Marx, Gerard, et al., “Heat Denaturation of Fibrinogen to Develop a Biomedical Matrix”, Journal of Biomedical Materials Research Part B: Applied Biomaterials, (Apr. 2007), 49-57.
Masri, Marwan A, et al., “Isolation of Human Fibrinogen of High Purity and in High Yield Using Polyethylene Glycol 1000”, Thromb Haemostas (Struttgart) vol. 49 (2), (1983), 116-119.
Matras, Helene, “Fibrin Seal: The State of the Art”, Journal of Oral Maxillofacial Surgery, vol. 43, (1985), 605-611.
Matras, Helene, et al., “Zur nahtlosen interfaszikularen Nerventransplantation im Tierexperiment”, Wein Med Woschtr 122:37—Not in English, (1972), 517-523.
Matuska, et al., “Autologous Solution Protects Bovine Cartilage Explants from IL-1a and STFa-Induced Cartilage Degradation”, Journal of Orthopaedic Research, (Jul. 16, 2013), 7 pgs.
Mehmet, C, et al., “Autologous Fibrin Glue From Intraoperatively Collected Platelet-Rich Plasma”, Ann Thorac Surg, vol. 53, (1992), 530-531.
Mehta, Sanjay, et al., “Gentamicin distribution from a collagen carrier”, Journal of Orthopaedic Research, vol. 14, No. 5—Abstract, (Sep. 1, 1996), 749-754.
Molnar, Amy, “Stem Cells from Muscles Can Repair Cartilage, Study Finds Genetically Engineered Muscle-Derived Stem Cells Improved Cartilage Repair in Rats”, American College of Rheumatology, (2005), 2 pgs.
Moretz, W., et al., “A simple autologous fibrinogen glue for otologic surgery”, Otolarvnaol Head Neck Surg 95, (Jul. 1986), 122-4.
Nakagami, Hironori, et al., “Novel Autologous Cell Therapy in Ischemic Limb Disease Through Growth Factor Secretion by Cultured Adipose Tissue-Derived Stromal Cells”, Angiogenesis by Adipose Tissue-Derived Cells, American Heart Association, Inc., (Dec. 2005), 2542-2547.
Nathan, Suresh, et al., “Cell-Based Therapy in the Repair of Osteochondral Defects: A Novel Use for Adipose Tissue”, Tissue Engineering, vol. 9, No. 4, Mary Ann Liebert, Inc., (2003), 733-744.
Nilsson, et al., “Bone Repair Induced by Bone Morphogenetic Protein in Ulnar Defects in Dogs”, The Journal of Bone and Joint Surgery, vol. 68 B., No. 4, (Aug. 1986), 635-642.
Okamoto, Y, et al., “Determination of soluble tumor necrosis factor-alpha receptor type (TNFRI) and II (TNFRII) in the urine of healthy Japanese subjects”, Journal of Immunoassay and Immunochemistry, 2011, vol. 32,. (2011), 145-155.
Okunishi, K, et al., “Hepatocyte Growth Factor Significantly Suppresses Collagen-Induced Arthritis in Mice”, The Journal of Immunology, vol. 179, No. 8, (Oct. 15, 2007), 15 pgs.
Orphardt, Charles E, “Denaturation of Proteins”, Virtual Chembook, Elmhurst College, <http://www.elmhurst.edu/chm/vchembook/568denaturation.html> (web accessed Mar. 9, 2011), (2003), 3 pgs.
O'Shaughnessey, Krista, et al., “Autologous Protein Solution Prepared From the Blood of Osteoarthritic Patients Contains an Enhanced Profile of Anti-Inflammatory Cytokines and Anabolic Growth Factors”, Journal of Orthopaedic Research, (Jun. 1, 2014), 1349-1355 pgs.
Parchment, et al., “Roles for in vitro myelotoxicity tests in preclincial drug development and clinical trial planning, Toxicology Pathology, Society of Toxicological Pathologists”, vol. 21, No. 2, (1993), 241-250.
Parker, Anna M, et al., “Adipose-derived stem cells for the regeneration of damaged tissues”, Expert Opinion, Cell- & Tissue-based Therapy, Expert Opin. Bioi. Ther., Informa UK Ltd, (2006), 567-578.
Planat-Benard, V., et al,, “Spontaneous Cardiomyocyte Differentiation From Adipose Tissue Stroma Cells”, Adipose-Derived Cell Cardiomyocyte, American Head Association, Inc., (Feb. 6, 2004), 223-229.
Pommer, et al., “Dielectrophoretic separation of platelets from whole blood in microfluidic channels”, Electrophoresis, (2008), 1213-1218.
Ponticiello, Michael S, “A Rapid Technique for the Isolation and Concentration of Stem Cells from Human Bone Marrow”, Cell Factor Technologies, Inc., (2006), 1 pg.
Rangappa, Sunil, et al., “Transformation of Adult Mesenchymal Stem Cells Isolated From the Fatty Tissue Into Cardiomyocytes”, Adult Stem Cells Transformed into Cardiomyoctyes, Ann Thorac Surg, (2003), 775-779.
Rigotti, M D, et al., “Clinical Treatment of Radiotherapy Tissue Damage by Lipoaspirate Transplant: A Healing Process Mediated by Adipose-Derived Adult Stem Cells”, Plastic and Reconstructive Surgery, Breast, PRS Journal vol. 119, No. 5, Stem Cell Therapy for Angiogenesis, (Apr. 15, 2007), 1409-1422.
Robert, Quigley L, et al., “Intraoperative Procurement of Autologous Fibrin Glue”, Ann Thorac Surg, vol. 56, (1993), 387-389.
Rubin, M. D., et al., “Clinical Treatment of Radiotherapy Tissue Damage by Lipoaspirate Transplant: A Healing Process Mediated by Adipose-Derived Adult Stem Cells”, Plastic and Reconstructive Surgery, Discussion vol. 119, No. 5, Stem Cell Therapy for Angiogenesis, (Apr. 15, 2007), 1423-1424.
Sadeghi, M, et al., “Strikingly higher interleukin (IL)-1a, IL-1b and soluble interleukin-1 receptor antagonist (sIL-1RA) but similar IL-2, sII-2R, IL-3, IL-4, IL-6, sII-6R, IL-10, tumour necrosis factor (TNF)-a, transforming growth factor (TGF)-B2, (cont.)”, (Title cont. “transforming growth factor (TGF)-(32 and interferon IFN-y urine Levels in healthy females compared to healthy males: protection against urinary tract injury?”) Clinical and Experimental Immunology, vol. 142, (2005), 312-317.
Sanal, M, et al., “Does fibrin glue cause foreign body reactions?”, Eu r J Pediatr Sura 2, (1992), 285-6.
Schaffler, Andreas, et al., “Concise Review: Adipose Tissue-Derived Stromal Cells-Basic and Clinical Implications for Novel Cell-Based Therapies”, Tissue-Specific Stem Cells, Stem Cells®, (Apr. 10, 2007), 12 pgs.
Schmidt, K G, “Labelling of Human and Rabbit Platelets with Indium-Oxine Complex”, (1979), 97-106.
Schmidt, K G, et al., “Preparation of Platelet Suspensions from Whole Blood in Buffer”, Scand. J. Hoemato, 23, (1979), 88-96.
Semple, Elisabeth, et al., “Quality of Thrombin Produced From the Patient's Own Plasma Using the TPD™, a New Thrombin-Processing Device”, Journal of American Society of Extra-Corporeal Technology, 37(2), (2005), 196-200.
Sevenoaks, Martin J., et al., “Chronic Obstructive Pulmonary Disease, inflammation and co-morbidity—a common inflammatory phenotype?”, respiratory Research vol. 7:70, (2006), 1-9.
Siedentop, Karl H, et al., “Autologous Fibrin Tissue Adhesive”, Laryngoscope, vol. 95, (Sep. 1985), 1074-1076.
Siedentop, Karl H, et al., “Extended Experimental and Preliminary Surgical Findings with Autologous Fibrin Tissue Adhesive Made from Patient's Own Blood”, Laryngoscope, vol. 96, (Oct. 1986), 1062-1064.
Sierra, D H, “Fibrin sealant adhesive systems: a review of their chemistry, material properties and clinical applications”, J Biomater Appl 7, (Apr. 1993), 309-52.
Silver, Frederick H, et al., “Review Preparation and use of fibrin glue in surgery”, Biomaterials 16 (1995), (1995), 891-903.
Solchaga, Luis A., et al., “Hyaluronic Acid-Based Polymers as Cell Carriers for Tissue-Engineered Repair of Bone and Cartilage”, Journal of Orthopaedic Research, Orthopaedic Research Society, US, vol. 17, (Jan. 1, 1999), 205-213.
Solem, Jan Otto, et al., “Hemoconcentration by Ultrafiltration During Open-Heart Surgery”, Scand J Thor Cardiovasc Surg 22, (1988), 271-274.
Spotnitz, William D, et al., “Successful Use of Fibrin Glue During 2 Years of Surgery at a University Medical Center”, The American Surgeon, vol. 55,, (Mar. 1989), 166-168.
Sutton, Robin G, et al., “Comparison of Three Blood-Processing Techniques During and After Cardiopulmonary Bypass”, Ann Thorac Surg (1993) vol. 56, (1993), 6 pgs.
Takahashi, Kazutoshi, et al., “Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors”, Cell, Elsevier Inc., (Nov. 30, 2007), 1-12.
Tawes, Jr., Roy L, et al., “Autologous Fibrin Glue: The Last Step in Operative Hemostatis”, The American Journal of Surgery, vol. 168, (Aug. 1994), 120-122.
Thompson, et al., “Fibrin Glue: A Review of Its Preparation, Efficacy, and Adverse Effects as a Topical Hemostat”, Drug Intelligence and Clinical Pharmacy, vol. 22, (Dec. 1988), 946-952.
Toriumi, Dean M, et al., “Surgical Tissue Adhesives in Otolaryngology—Head and Neck Surgery”, Otolaryngologic Clinics of North America, vol. 27, No. 1, (Feb. 1994), 203-209.
Wang, “Cell separation by dielectrophoretic field-flow-fractionation”, Analytical Chemistry, (2000), 832-839.
Weis-Fogh, U S, “Fibrinogen prepared from small blood samples for autologous use in a tissue adhesive system”, Eur Surg Res 20, (1988), 381-9.
Weisman, M D, “Biochemical Characterization of Autologous Fibrinogen Adhesive”, Laryngoscope 97, (Oct. 1987), 1186-1190.
Wiseman, David M, et al., “Wound Dressings: Design and Use”, In Wound Healing: Biochemical & Clinical Aspects 1st ed., vol., (1992), 562-580.
Yoon, Eulsik, et al., “In Vivo Osteogenic Potential of Human Adipose-Derived Stem Cells/Poly Lactide-Co-Glycolic Acid Constructs for Bone Regneration in a Rat Critical-Sized Calvarial Defect Model”, Tissue Engineering, vol. 13, No. 3, (2007), 619-627.
Zhang, Duan-Zhen, et al., “Transplantation of autologous adipose-derived stem cells ameliorates cardiac function in rabbits with myocardial infarction”, Chinese Medical Journal, vol. 120, No. 4 General Hospital of Shenyang Military Region, (2007), 300-307.
Zuk, P. A, et al., “Multilineage cells from human adipose tissue: Implications for cellbased therapies”, Tissue Engineering, 7(2), XP00219871 0, ISSN: 1076-3279, (Apr. 1, 2001), 211-228.
“International Application Serial No. PCT/US2014/016900, International Preliminary Report on Patentability dated Sep. 24, 2015”, 9 pgs.
“U.S. Appl. No. 13/841,083, Examiner Interview dated Feb. 24, 2017”, 1 pg.
“U.S. Appl. No. 13/841,083, Non Final Office Action dated Feb. 24, 2017”, 12 pgs.
“U.S. Appl. No. 13/841,083, Non Final Office Action dated Feb. 24, 2017”, Examiner Interview Summary from Feb. 24, 2017 included, 14 pgs.
“U.S. Appl. No. 13/841,083, Response filed May 24, 2017 to Non Final Office Action dated Feb. 24, 2017”, 18 pgs.
Agu, R. U., et al., “The lung as a route for systemic delivery of therapeutic proteins and peptides”, Respir Res.; vol. 2, (2001), pp. 198-209.
Related Publications (1)
Number Date Country
20140274895 A1 Sep 2014 US