throbber
(19) United States
`(12) Patent Application Publication (10) Pub. No.: US 2014/0030315 A1
`JOHNSON
`(43) Pub. Date:
`Jan. 30, 2014
`
`US 20140030315A1
`
`(54) NANOFIBER SCAFFOLDS FOR
`BOLOGICAL STRUCTURES
`
`(71) Applicant: Jed K. JOHNSON, Columbus, OH (US)
`
`(72) Inventor: Jed K. JOHNSON, Columbus, OH (US)
`(21) Appl. No.: 13/740.913
`(22) Filed:
`Jan. 14, 2013
`
`Related U.S. Application Data
`(60) Provisional application No. 61/585,869, filed on Jan.
`12, 2012.
`Publication Classification
`
`(51) Int. Cl.
`A 6LX 9/70
`A6 IK 47/32
`A6 IK35/12
`
`(2006.01)
`(2006.01)
`(2006.01)
`
`(52) U.S. Cl.
`CPC. A61 K9/70 (2013.01); A61 K35/12 (2013.01);
`A61K47/32 (2013.01)
`USPC ....... 424/444; 424/400; 424/93.7: 514/772.1;
`435/396: 118/621
`s
`
`57
`(57)
`
`ABSTRACT
`
`- Y
`-
`A system for manufacturing an artificial construct suitable for
`transplantation into a biological organism that includes a two
`or three three-dimensional preform that is based on the actual
`two or three-dimensional structure of a native mammalian
`tissue; and an electrospinning apparatus, wherein the electro
`spinning apparatus is operative to depositat least one layer of
`polymer fibers on the preform to form a polymer scaffold, and
`wherein the orientation of the fibers in the scaffold relative to
`one another is Substantially parallel.
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`Patent Application Publication
`
`Jan. 30, 2014 Sheet 1 of 6
`
`US 2014/0030315 A1
`
`8
`
`383
`
`cs: a
`
`
`
`w
`
`s
`s
`
`
`
`
`
`
`
`
`
`
`
`wife
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`Patent Application Publication
`
`Jan. 30, 2014 Sheet 2 of 6
`
`US 2014/003O31S A1
`
`
`
`
`
`
`
`
`
`
`
`
`
`S:
`
`.
`
`8
`
`
`
`s
`
`3. :
`
`2 s
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`&
`
`&
`
`S &S
`
`
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`Patent Application Publication
`
`Jan. 30, 2014 Sheet 3 of 6
`
`US 2014/003O31S A1
`
`
`
`:
`
`
`
`&
`
`F.G. S
`
`
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`Patent Application Publication
`
`Jan. 30, 2014 Sheet 4 of 6
`
`US 2014/003O31S A1
`
`
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`Patent Application Publication
`
`Jan. 30, 2014 Sheet 5 of 6
`
`US 2014/003O31S A1
`
`
`
`is v
`838. 388
`
`kxx &cs: 8:8:
`88 & 3. 3.
`
`83
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`Patent Application Publication
`
`Jan. 30, 2014 Sheet 6 of 6
`
`US 2014/003O31S A1
`
`F.G. 2
`
`
`
`it.
`N
`28
`wis
`
`F.G. 3
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`US 2014/003O315 A1
`
`Jan. 30, 2014
`
`NANOFIBER SCAFFOLDS FOR
`BOLOGICAL STRUCTURES
`
`CROSS-REFERENCE TO RELATED
`APPLICATIONS
`0001. This patent application claims the benefit of U.S.
`Provisional Patent Application Ser. No. 61/585,869 filed on
`Jan. 12, 2012, and entitled “Biocompatible Nanofiber Mate
`rials for Biological Structures, the disclosure of which is
`hereby incorporated by reference herein in its entirety and
`made part of the present U.S. utility patent application for all
`purposes.
`
`BACKGROUND OF THE INVENTION
`0002 Tissue engineering involves the synthesis of bio
`logically relevant tissue for a wide range of applications
`including wound healing and the replacement or Support of
`damaged organs. A common strategy is culturing target spe
`cific cells in vitro in a scaffold followed by implantation of the
`scaffold in a biological organism. As a logical cellular source
`for tissue engineering, stem cells have attracted a great deal of
`attention due to their relatively fast proliferation rate and
`diverse differentiation potential to various phenotypes. These
`include cells derived from several origins: induced pluripo
`tent stem cells from fibroblasts, mesenchymal stem cells from
`bone marrow and adult stem cells from adipose tissue. Stem
`cells distinctively self-renew and their terminal differentia
`tion depends on the influence of soluble molecules (e.g.,
`growth factors, cytokines) as well as physical and biochemi
`cal interactions with scaffolds.
`0003 Cellular behavior and subsequent tissue develop
`ment at the cell—scaffold interface therefore involve adhe
`Sion, motility, proliferation, differentiation and functional
`maturity. The physicochemical properties of a scaffold. Such
`as bulk chemistry, Surface chemistry, topography, three-di
`mensionality and mechanical properties, all influence cellular
`response. Bulk chemistry can control cytotoxicity, as most
`scaffolds are made of biodegradable materials and must even
`tually release the by-products of their degradation. The effect
`of surface chemistry is often mediated by instantly adsorbed
`proteins such as fibronectin, collagen, fibrinogen, vitronectin,
`and immunoglobulin that affect phenotype, viability, and
`morphology, as well as proliferation and differentiation.
`0004 Studies regarding the effect of surface topography
`and texture on cellular response have been conducted. Stem
`cells are known to recognize topographical features of the
`order of hundreds of nanometers to several micrometers, and
`exhibit distinctive genomic profiles in the absence of bio
`chemical differentiation cues and a commitment to terminal
`differentiation. Electrospun scaffolds are ideal matrices for
`two dimensional or three dimensional culture of the cells
`providing non-woven nano- to micro-sized fibrous micro
`structures typically having relative porosities of 70-90%.
`Natural biodegradable materials such as collagen, gelatin,
`elastin, chitosan, and hyaluronic acid, as well as Synthetic
`biodegradable polymers such as poly(e-caprolactone) (PCL),
`poly(glycolic) acid (PGA) and poly(lactic) acid (PLA), have
`been electrospun for chondral and Osseous applications.
`0005. In general, the broad utility of electrospun scaffolds
`for tissue engineering, wound healing, and organ replacement
`is clear (see Modulation of Embryonic Mesenchymal Pro
`genitor Cell Differentiation via Control Over Pure Mechani
`cal Modulus in Electrospun Nanofibers, Nama et al., Acta
`
`Biomaterialia 7, 1516-1524 (2011), which is incorporated by
`reference herein in its entirety, for all purposes) and the
`present invention provides polymer fiber constructs for these
`and other applications. Alignment of fibers produced during
`electrospinning has previously been achieved by various
`methods including, for example, high Velocity collection of
`fibers (e.g., on the Surface of a high Velocity rotating mandrel)
`and alternating collection of fibers from one grounded elec
`trode to another on an immobile surface or in the air. Current
`methods of electrospinning aligned fibers are not known to
`achieve the ideal alignment of fibers observed in the human
`body, such as, for example, in brain tissue. Therefore,
`improvements in alignment must be made in order to obtain
`the high degree of alignment necessary for an in vitro model
`of human tissue.
`
`SUMMARY OF THE INVENTION
`0006. The following provides a summary of certain exem
`plary embodiments of the present invention. This Summary is
`not an extensive overview and is not intended to identify key
`or critical aspects or elements of the present invention or to
`delineate its scope.
`0007. In accordance with one aspect of the present inven
`tion, a synthetic construct Suitable for transplantation into a
`biological organism is provided. This construct includes a
`synthetic construct Suitable for transplantation into a biologi
`cal organism, comprising: a two-dimensional or three-dimen
`sional polymer scaffold, wherein the shape and dimensions of
`the polymer scaffold are based on a native biological struc
`ture, wherein the polymer scaffold further includes at least
`one layer of polymer fibers that have been deposited by elec
`trospining, and wherein the orientation of the fibers in the
`scaffold relative to one another is substantially parallel; and
`wherein, optionally, the polymer scaffold has been preseeded
`with at least one type of biological cell prior to implantation
`into a biological organism, and wherein the at least one type
`of biological cell is operative to facilitate integration of the
`polymer scaffold into the organism so that the polymer scaf
`fold may function in a manner significantly similar to or the
`same as the native structure.
`0008. In accordance with another aspect of the present
`invention, a system for manufacturing an artificial construct
`Suitable for transplantation into a biological organism is pro
`vided. This system includes: a two or three three-dimensional
`preform that is based on the actual two or three-dimensional
`structure of a native mammalian tissue; and an electrospin
`ning apparatus, wherein the electrospinning apparatus is
`operative to depositat least one layer of polymer fibers on the
`preform to form a polymer scaffold, and wherein the orien
`tation of the fibers in the scaffold relative to one another is
`Substantially aligned or parallel.
`0009. In yet another aspect of this invention, a system for
`manufacturing an artificial construct suitable for transplanta
`tion into a biological organism for wound healing purposes is
`provided. This system includes a two or three three-dimen
`sional preform that is based on the actual two or three-dimen
`sional structure of a native mammalian tissue; an electrospin
`ning apparatus, wherein the electrospinning apparatus is
`operative to depositat least one layer of polymer fibers on the
`preform to form a polymer scaffold, and wherein the orien
`tation of the fibers in the scaffold relative to one another is
`Substantially parallel; and a least one type of biological cell
`for preseeding onto the polymer Scaffold, and wherein the at
`least one type of biological cell further includes autologous
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`US 2014/003O315 A1
`
`Jan. 30, 2014
`
`cells or allogeneic cells, and wherein the autologous cells or
`allogeneic cells further include cord blood cells, embryonic
`stem cells, induced pluripotent cells, mesenchymal cells, pla
`cental cells, bone marrow derived cells, hematopoietic cell,
`epithelial cells, endothelial cells, fibroblasts and chondro
`cytes.
`0010 Additional features and aspects of the present inven
`tion will become apparent to those of ordinary skill in the art
`upon reading and understanding the following detailed
`description of the exemplary embodiments. As will be appre
`ciated by the skilled artisan, further embodiments of the
`invention are possible without departing from the scope and
`spirit of the invention. Accordingly, the drawings and associ
`ated descriptions are to be regarded as illustrative and not
`restrictive in nature.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`0.011 The accompanying drawings, which are incorpo
`rated into and form a part of the specification, Schematically
`illustrate one or more exemplary embodiments of the inven
`tion and, together with the general description given above
`and detailed description given below, serve to explain the
`principles of the invention, and wherein:
`0012 FIG. 1 depicts electrospun PCL fibers spun without
`an anti-static bar for 10 minutes at 0.5 ml/h, +4 kV on the
`needle, and -4 kV on the mandrel and the plot shows the angle
`of the fibers from horizontal;
`0013 FIG. 2 depicts electrospun PCL fibers spun with an
`anti-static bar for 10 minutes at 0.5 ml/h, +4 kV on the needle,
`and -4 kV on the mandrel and the plot shows the angle of the
`fibers from horizontal.
`0014 FIG.3 depicts electrospun PCL fibers spun without
`an anti-static bar at 1.0 ml/h and the plot shows the angle of
`the fibers from horizontal;
`0015 FIG. 4 depicts electrospun PCL fibers spun with an
`anti-static bar at 1.0 ml/h and the plot shows the angle of the
`fibers from horizontal;
`0016 FIG. 5 depicts electrospun PCL fibers spun using a
`dual syringe setup with two anti-static bars, wherein each
`needle has a flow rate of 1.0 ml/h and the plot shows the angle
`of the fibers from horizontal;
`0017 FIG. 6 depicts a dual syringe electrospinning setup,
`wherein the triangles indicate the relative positions of the first
`and second anti-static bars, wherein the straight arrows indi
`cate relative locations of the Syringe pump with polymer
`Solution, and wherein the clockwise arrows indicate the rota
`tion direction of the fiber collection mandrel at the center of
`the image:
`0018 FIG. 7 depicts electrospun PCL fibers spun with an
`anti-static bar at 1.0 ml/h, wherein an alternating ground was
`not applied during electrospinning;
`0019 FIG. 8 depicts electrospun PCL fibers spun with an
`anti-static bar at 1.0 ml/h, wherein an alternating ground was
`applied during electrospinning.
`0020 FIG. 9 is a scanning electromicrograph of the poly
`mer nanofibers used in the present invention;
`0021
`FIGS. 10-11 are scanning electromicrographs of the
`polymer nanofibers used in the present invention with a coat
`ing of marrow Stromal cells;
`0022 FIG. 12 is a scanning electromicrograph of the poly
`mer nanofibers used in the prior art; and
`0023 FIG. 13 is a scanning electromicrograph of the poly
`mer nanofibers used in the prior art with a coating of marrow
`stromal cells.
`
`DETAILED DESCRIPTION OF THE INVENTION
`0024 Exemplary embodiments of the present invention
`are now described with reference to the Figures. Although the
`following detailed description contains many specifics for the
`purposes of illustration, a person of ordinary skill in the art
`will appreciate that many variations and alterations to the
`following details are within the scope of the invention.
`Accordingly, the following embodiments of the invention are
`set forth without any loss of generality to, and without impos
`ing limitations upon, the claimed invention.
`0025. With reference to the Figures, this invention relates
`generally to the construction of implantable artificial tissues
`for humans and/or animals, and more specifically to a process
`or method for manufacturing two-dimensional polymer
`microScale and nanoscale structures for use as Scaffolds in the
`growth of biological structures. The use of these scaffolds in
`creating or repairing numerous and multiple biological tis
`Sues and structures, is contemplated by and included in this
`invention. Exemplary versions of the manufacturing process
`of this invention include preparing a preform or Substrate that
`is based on an actual native tissue and/or organ; electrospin
`ning one or more layers of nanoscale (less than 1000 nanom
`eters) or microscale (less than 50 microns) polymer fibers on
`the preform to form a nanofiber-based scaffold. The fibers are
`typically formed by electrospinning by extruding a polymer
`Solution from a fiberization tip; creating an electronic field
`proximate to the fiberization tip; and positioning a ground or
`opposite polarity within the preform. The preform may be
`rotated to align the fibers on the preform or a second ground
`or polarity may be placed in the preform and rapidly Switch
`ing the electric field to align the fibers. The microscale and
`nanoscale polymer fibers may be randomly aligned or maybe
`substantially parallel or both. These nanofiber structures may
`be seeded with one or more types of biological cells prior to
`implantation in the body to increase the rate of tissue growth
`into the scaffold. The polymer scaffold may include autolo
`gous or allogeneic cells Such as cord blood cells, embryonic
`stem cells, induced pluripotent cells, mesenchymal cells, pla
`cental cells, bone marrow derived cells, hematopoietic cell,
`epithelial cells, endothelial cells, fibroblasts, chondrocytes or
`combinations thereof. These biological cells may be applied
`to the surface of the scaffold or distributed throughout the
`scaffold matrix utilizing perfusion within a bioreactor. The
`polymer fibers may also be coated or otherwise treated with at
`least one compound that is operative to promote cellular
`attachment to the scaffold or promote engraftment of the
`scaffold into the biological organism. The at least one com
`pound may be selected from the group consisting of proteins,
`peptides, cytokines, growth factors, antibiotic compounds,
`anti-inflammatory compounds, and combinations thereof.
`0026 Choosing a material that accurately mimics the
`mechanical properties of the native tissue or organ may pro
`mote proper stem cell differentiation and facilitate normal
`function of the replacement tissue or organ. Included materi
`als may be non-resorbable for permanent implantation or may
`be designed to slowly degrade while the host body rebuilds
`the native tissue. In the latter case, the implanted prosthesis
`will eventually be completely resorbed. Permanent (i.e., non
`resorbable) polymers may include polyurethane, polycarbon
`ate, polyester terephthalate and degradable materials may
`include polycaprolactone, polylactic acid, polyglycolic acid,
`gelatin, collagen, or fibronectin. The fibers may be electro
`spun onto a preform with the desired prosthesis shape. An
`exemplary mandrel (preform) may be coated with Teflon or
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`US 2014/003O315 A1
`
`Jan. 30, 2014
`
`similar material to facilitate removal of the scaffold after
`deposition or a slight taper (e.g., about 1°) can be manufac
`tured into the mandrel. Nearly any size or shape can be pro
`duced from the electrospun fibers by using a pre-shaped form
`and the fiber deposition methods of the present invention.
`0027 Closely mimicking the structural aspects of the tis
`Sue or organ is important with regard to replicating the func
`tion of the native tissue or organ. By controlling the orienta
`tion of the fibers and assembling a composite structure of
`different materials and/or different fiber orientations it is pos
`sible to control and direct cell orientation and differentiation.
`Fiber orientation can be altered in each layer of a composite or
`sandwich scaffold in addition to the material and porosity to
`most closely mimic the native tissue. A properly constructed
`scaffold will permit substantially complete cellular penetra
`tion and uniform seeding for proper function and prevention
`of necrotic areas developing. If the fiber packing is too dense,
`then cells may not be able to penetrate or migrate from the
`exposed surfaces into the inner portions of the scaffold. How
`ever, if the fiber packing is not close enough, then attached
`cells may not be able to properly fill the voids, communicate
`and signal each other and a complete tissue or organ may not
`be developed. Controlling fiber diameter can be used to
`change scaffold porosity as the porosity Scales with fiber
`diameter. Alternatively, blends of different polymers may be
`electrospun together and one polymer preferentially dis
`solved to increase scaffold porosity. The properties of the
`fibers can be controlled to optimize the fiber diameter, the
`fiber spacing orporosity, the morphology of each fiber such as
`the porosity of the fibers or the aspect ratio, varying the shape
`from round to ribbon-like. The precursor solution described
`below may be controlled to optimize the modulus or other
`mechanical properties of each fiber, the fiber composition,
`and/or the degradation rate (from rapidly biosoluable to bio
`persitent). The fibers may also be formed as drug eluting
`fibers, anti-bacterial fibers or the fibers may be conductive
`fibers, radio opaque fibers to aid in positioning or locating the
`fibers in an X-ray, CT or other scan.
`0028. In accordance with this invention, the process of
`electrospinning is driven by the application of a high Voltage,
`typically between 0 and 30 kV, to a droplet of a polymer
`solution or meltata flow rate between 0 and 50 ml/h to create
`a condition of charge separation between two electrodes and
`within the polymer Solution to produce a polymer jet. A
`typical polymer Solution would consist of a polymer Such as
`polycaprolactone, polystyrene, or polyetherSulfone and a sol
`vent such as 1,1,1,3,3,3-Hexafluoro-2-propanol, N,N-Dim
`ethylformamide, acetone, or tetrahydrofuran in a concentra
`tion range of 1-50 wt %. As the jet of polymer solution travels
`toward the electrode it is elongated into small diameter fibers
`typically in the range of 0.1-100 um.
`0029. In preparing an exemplary scaffold, a polymer
`nanofiber precursor solution is prepared by dissolving 2-30
`wt % polyethylene terephthalate (PET) (Indorama Ventures)
`in a mixture of 1,1,1,3,3,3-hexafluoroisopropanol (HFIP) and
`trifluoroacetic acid and the solution is heated to 60° C. fol
`lowed by continuous stirring to dissolve the PET. The solu
`tion may be cooled to room temperature and the Solution
`placed in a syringe (e.g., 60 cc) with a blunt tip needle (e.g., 20
`gauge). The nanofibers are formed by electrospinning using a
`high Voltage DC power Supply (Glassman High Voltage, Inc.,
`High Bridge, N.J.) set to 1 kV-40kV (e.g., +15 kV) positive or
`negative polarity, a 5-30 cm (e.g., 15 cm) tip-to-substrate
`distance, and a 1 ul/hr to 100 mL/hr (e.g., 10 ml/hr) flow rate.
`
`It is possible to use a needle array including a large number of
`needles (e.g., >1000) to increase system output. Fiber diam
`eter may be controlled by the viscosity of the precursor solu
`tion and the solvent used and suitable exemplary fibers are in
`the range of 100 nanometer 30 microns. Approximately 0.2-3
`mm (e.g., 1 mm) thickness of randomly oriented and/or
`highly-aligned fibers may be deposited onto the form, and
`polymer rings added, followed by an additional approxi
`mately 0.2-3.0 mm (e.g., 2 mm) of fiber added while the form
`is rotated. The scaffold may be placed in a vacuum overnight
`to ensure removal of residual solvent (typically less than 10
`ppm) and treated using a radio frequency gas plasma for 1
`minute to make the fibers more hydrophilic and promote cell
`attachment. Samples may be storied in re-closeable polyeth
`ylene bags, or the like.
`0030. In accordance with this invention, an exemplary
`preparation of electrospinning solution typically includes
`polyethylene terephthalate (PET), polycaprolactone (PCL),
`polylactic acid (PLA), polyglycolic acid (PGA), polyether
`ketoneketone (PEKK), polyurethane (PU), polycarbonate
`(PC), polyamide (Nylon), natural polymers such as fibronec
`tin, collagen, gelatin, hyaluronic acid or combinations thereof
`that are mixed with a solvent and dissolved. Suitable solvents
`include acetone, dimethylformamide, trifluoroacetic acid,
`hexafluoroisopropanol, acetic acid, dimethylacetamide, chlo
`roform, dichloromethane, water, ionic compounds, or com
`binations thereof. A form is prepared for the deposition of
`nanofibers. Optionally, simulated cartilage or other Support
`ive tissue may be applied to the form and the fibers are then
`sprayed onto or transferred onto a form to build up the scaf
`fold.
`0031. During electrospinning, polymer fibers are driven
`toward a collector by charge separation caused by applied
`Voltage. The collector typically is a conductive Surface. Such
`as, for example, aluminum or copper, and in this disclosure
`the collector is covered by a thin layer of plastic, ranging, for
`example, between about 0.001-0.1 inches thick. The charge
`that drives electrospinning toward the collector is derived
`from mobile ions within the polymer solution or melt. The jet
`of polymer that is produced has a net positive or negative
`charge, depending upon the polarity of the Voltage applied to
`the electrode(s). When the jet solidifies on the collector Sur
`face, a charge builds up as Subsequent fiber layers are col
`lected. As the charge builds up on the surface, fiber with
`similar charge is repelled leading to irregularly arranged
`fibers and thus a lower degree of alignment. To reduce the
`effects of surface charge, an anti-static device (e.g., bar) may
`be incorporated into the process to improve fiberalignment.
`The anti-static bar bombards the surface of a sample with
`positively and negatively charged ions in the form of for
`example, a plasma or corona discharge to neutralize the
`charge on the Substrate. Therefore, as fiber builds up. Succes
`sive layers of fibers will deposit more uniformly side-by-side
`(parallel relationship) to increase the alignment. The position
`of the anti-static bar is generally parallel to the surface of the
`collection mandrel, wheel, device, plate, etc. and is for
`example, about 0.5-3 inches away from the surface.
`0032 Experimental results demonstrate that fiber align
`ment was improved significantly with the addition of an anti
`static bar (or a device having similar functional properties),
`when compared to samples spun under the same conditions
`without the anti-static bar. FIG. 1 shows the alignment of a
`sample to be 83%, when electrospun without a static bar;
`while FIG. 2 shows a 12% increase in fiberalignment to 95%,
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`US 2014/003O315 A1
`
`Jan. 30, 2014
`
`when a static bar treats the Surface during spinning. An addi
`tional benefit of anti-static bars is the ability to electrospin
`using multiple needles. FIG.3 shows a sample spun at 1 ml/h
`without a static bar; while FIG. 4 shows a sample spun at 1
`ml/h with an anti-static bar, and FIG. 5 shows a sample spun
`using a dual syringe configuration and two anti-static bars
`treating a wheel surface where the fibers are being deposited.
`When fibers are spun without an anti-static bar (see FIG. 3),
`the fiberalignment is low, with only 74% of fibers collected in
`a low angle orientation. With an anti-static bar, under the
`same spinning conditions, the alignment becomes 92% (see
`FIG. 4). Additionally, alignment is maintained at 92% (see
`FIG. 5) when multiple needles are used for electrospinning,
`and when two anti-static bars treat the wheel surface as illus
`trated in FIG. 6. This demonstrates the ability to scale-up the
`production rate by incorporating additional needles and anti
`static devices. Antistatic bars (or, alternately, one or more
`ionizing guns) may also be used to create continuous (i.e.,
`very long) fibers that are continuously aligned.
`0033. As previously indicated, the alternating collection
`offibers from one ground to the next will create aligned fiber.
`High velocity collection of fibers, such as on the surface of a
`rotating mandrel, will achieve similar results. When the two
`methods are combined, alternating grounds on a rotating
`mandrel, the fiberalignment is enhanced beyond what either
`method typically achieves. The combined method of fiber
`alignment is highly effective when the surface of the mandrel
`is coated in a thin insulating layer, such as, for example,
`polystyrene. An alternating ground is established by securing
`a continuous plastic sheet and wrapping conductive tape
`around the surface of the wheel, or a similar method that
`creates alternating layers of conductive and non-conductive
`Surface material. The conductive tape generally is made of
`for example, carbon or copper and ranges between about
`0.1-2 inches wide and is spaced uniformly around the wheel
`circumference. This tape should be connected to the charged/
`grounded wheel for the alternating ground effect to be
`obtained. FIGS. 7-8 illustrate the alternating ground effect.
`FIG. 7 shows a sample that was electrospun without the
`alternating ground and with an anti-static bar. FIG. 8 shows a
`sample that was electrospun with both the alternating ground
`and anti-static bar. These images demonstrate the dramatic
`improvement in alignment when using the alternating ground
`and anti-static bar on a high-speed mandrel.
`0034. With reference to FIGS. 9-13, the polymer fiber
`scaffolds of the present invention may be used to manufacture
`two-dimensional biocompatible patches of varying thickness
`for use in humans or animals (e.g., primates, cats, dogs,
`horses and cattle) as an aid in wound healing involving
`muscles, internal organs, bones, cartilage, and/or external
`tissues. Biocompatible materials, which are suitable for use in
`medical applications within the body or on external Surfaces,
`typically elicit little or no immune response in human or
`Veterinary applications. In one or more exemplary embodi
`ments, these patches include Substantially parallel electro
`spun nanoscale and microscale polymer fibers. These patches
`may be seeded with biological cells prior to use to increase the
`rate of tissue growth into the patch. Such biological cells may
`include autologous or allogenic cells such as cord blood cells,
`embryonic stem cells, induced pluripotent cells, mesenchy
`mal cells, placental cells, bone marrow derived cells, hemato
`poietic cells, epithelial cells, endothelial cells, fibroblasts and
`chondrocytes. Examples of internal uses include tissue, ocu
`lar tissue (lens, cornea, optic nerve or retina), intestinal tissue,
`
`internal organs such as the liver, kidney, spleen, pancreas,
`esophagus, trachea, uterus, stomach, bladder, muscles, ten
`dons, ligaments, nerves, dura matter and other brain struc
`tures, dental structures, blood vessels and other bodily struc
`tures. Examples of external uses may include wound
`dressings, burn and abrasion coverings, and recovery aides to
`inhibit the formation of scar tissue. External structures are
`typically the skin but may include the cornea or surface of the
`eye, the ear canal, the mouth and nasal passages or the nail
`bed.
`0035 An exemplary method for making the biocompat
`ible patches of this invention includes depositing a layer of
`Substantially parallel electrospun polymer fibers on a preform
`(i.e., a wheel or similar structure) to form a fiber patch or gap
`filling material; and applying donor cells to the patch. Pref
`erably, the fibers are formed byelectrospinning by extruding a
`polymer Solution from a fiberization tip; creating an elec
`tronic field proximate to the fiberization tip; and positioning a
`ground within the deposition surface. The preform may be
`rotated to align the fibers on the Surface or a second ground or
`opposite polarity may be placed in the preform and rapidly
`Switching the ground. To speed the growth of human tissue
`into the fiber preform, the fibers are aligned by rapidly spin
`ning the preform so that alignment of the structure produced
`by standard electrospinning while the fibers are drawn into a
`Substantially parallel ordering by the tension created by spin
`ning the form. See generally, Preparation and anisotropic
`mechanical behavior of highly-Oriented electrospun poly(bu
`tylene terephthalate) fibers, Journal of Applied Polymer Sci
`ence Volume 101, Issue 3, pages 2017-2021 (August 2006),
`which is incorporated by reference herein, in its entirety. A
`split ground technique, in which fiber deposition rapidly
`alternates between two separate grounding plates within the
`preform or by alternating the electric field is also an effective
`method of forming parallel fibers on the preform. See, gen
`erally, Electrospinning Nanofibers as Uniaxially Aligned
`Arrays and Layer-by-Layer Stacked Films, Advanced Mate
`rials Volume 16, Issue 4, pages 361-366 (February 2004),
`which is incorporated by reference herein, in its entirety.
`Fiber alignment can be further enhanced by controlling cer
`tain variables Such as humidity, Solvents, flow rates, and rota
`tional speeds. Lower relative humidity (e.g., 20%) results in
`better overall alignment than higher relative humidity (e.g.,
`50%). However, deposition efficiency is increased in higher
`humidity (e.g., deposition on plastic). The use of certain
`Solvents such as acetone typically reduce fiber alignment,
`while others such as 1,1,1,3,3,3-Hexafluoro-2-propanol typi
`cally increase alignment. Decreasing the flow rate at which
`the polymer Solution is being pumped typically increases
`fiberalignment, while increasing the flow rate decreases fiber
`alignment. Finally, increasing the rotational speed of the
`wheel upon which the fiber is being deposited typically
`increases fiberalignment.
`0036. The thickness of the patch may be from a few
`microns for application to Surfaces to speed cellular growth
`and inhibitscarring to several centimeters for use as a plug for
`insertion into a wound or to speed the growth of structures in
`a specific direction. High thickness patches are useful in
`repairing infracted cardiac tissue, esophageal or tracheal tis
`Sue or Supporting the growth of nerve in a predetermined
`direction. Depending on the material used in preparing the
`fibers the patch may dissolve within the body after a prede
`termined time or may be relatively permanent for longer term
`applications. It is also possible to fabricate a multipart struc
`
`NANOFIBER 1014
`Nanofiber v Acera
`IPR2021-01016
`
`

`

`US 2014/003O315 A1
`
`Jan. 30, 2014
`
`ture which includes one or more layers

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket