WO2013028573A1 - Séparation par diélectrophorèse de gradients (grads) avec thérapie lumineuse concomitante - Google Patents

Séparation par diélectrophorèse de gradients (grads) avec thérapie lumineuse concomitante Download PDF

Info

Publication number
WO2013028573A1
WO2013028573A1 PCT/US2012/051484 US2012051484W WO2013028573A1 WO 2013028573 A1 WO2013028573 A1 WO 2013028573A1 US 2012051484 W US2012051484 W US 2012051484W WO 2013028573 A1 WO2013028573 A1 WO 2013028573A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
dielectrophoresis
chamber
flow
top plate
Prior art date
Application number
PCT/US2012/051484
Other languages
English (en)
Other versions
WO2013028573A4 (fr
Inventor
Allan Yang Wu
David Martin Morrow
Original Assignee
Allan Yang Wu
David Martin Morrow
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Allan Yang Wu, David Martin Morrow filed Critical Allan Yang Wu
Priority to CA2845713A priority Critical patent/CA2845713A1/fr
Priority to JP2014527210A priority patent/JP2014531199A/ja
Publication of WO2013028573A1 publication Critical patent/WO2013028573A1/fr
Publication of WO2013028573A4 publication Critical patent/WO2013028573A4/fr
Priority to IL230985A priority patent/IL230985A0/en

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C5/00Separating dispersed particles from liquids by electrostatic effect
    • B03C5/02Separators
    • B03C5/022Non-uniform field separators
    • B03C5/026Non-uniform field separators using open-gradient differential dielectric separation, i.e. using electrodes of special shapes for non-uniform field creation, e.g. Fluid Integrated Circuit [FIC]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N5/067Radiation therapy using light using laser light
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B03SEPARATION OF SOLID MATERIALS USING LIQUIDS OR USING PNEUMATIC TABLES OR JIGS; MAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03CMAGNETIC OR ELECTROSTATIC SEPARATION OF SOLID MATERIALS FROM SOLID MATERIALS OR FLUIDS; SEPARATION BY HIGH-VOLTAGE ELECTRIC FIELDS
    • B03C5/00Separating dispersed particles from liquids by electrostatic effect
    • B03C5/005Dielectrophoresis, i.e. dielectric particles migrating towards the region of highest field strength
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N13/00Treatment of microorganisms or enzymes with electrical or wave energy, e.g. magnetism, sonic waves
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N2005/065Light sources therefor
    • A61N2005/0651Diodes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N2005/0658Radiation therapy using light characterised by the wavelength of light used
    • A61N2005/0659Radiation therapy using light characterised by the wavelength of light used infrared
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/06Radiation therapy using light
    • A61N2005/073Radiation therapy using light using polarised light

Definitions

  • the present invention relates the field of dielectrophoresis and more particularly to a method and device using dielectrophoresis to separate fractions of particles/cells from a heterogeneous population of particles/cells and simultaneously allow light or electrical therapy to the cells.
  • the cells may be derived from liposuctioned adipose tissue.
  • Elution by frequency modulation is one method of separating cell fractions from a heterogeneous population. See “Enrichment of putative stem cells from adipose tissue using dielectrophoresis field-flow fractionation", Jody Vykoukal, Daynene M. Vykoukal, Susanne Freyberg, Eckhard U. Alt, and Peter R. C. Gascoynea, Lab Chip. 2008 August; 8(8): 1386-1393 (Published online 2008 May 28. doi: 10.1039/b717043b). This is done by starting with a high "entrapping" frequency of 200 kHz and slowly stepping down the frequency. Each unique fraction is collected as the current is stepped down. However, in this method it is difficult to adjust the timing so as to collect the different elution fractions.
  • This invention is an improvement of the inventor's prior US Patent Applications Nos. 61/545,015, 61/136,932 and 12/578,549; and PCT Patent Application No. PCT/US09/6071 .
  • the non-provisional and PCT applications were published as US Patent Application Publication No. 2010/01 12084 and WIPO Publication No. WO/2010/045389.
  • the entire specifications, claims and drawings of these applications and publications are hereby incorporated into this application by reference.
  • the improvement also comprises modification of the prior invention apparatus to detect differing electrical "signatures" of each different phase (i.e. oil vs. fat vs. aqueous solution) as they pass through a channel and actuate a diverting port at the end of the channel to collect and separate the different phases of lipoaspirate.
  • the electrical signatures may be measured by means of: current, current signal shift, conductivity, resistance, impedance, phase shift, magnetic shift with either direct or alternating current.
  • the complex dielectric constant is, where ⁇ is the dielectric constant, ⁇ is the_electrical conductivity, ⁇ is the field frequency, and j is the imaginary number.
  • This equation is accurate for highly elongated ellipsoids when the electric field gradients are not very large (e.g., close to electrode edges).
  • the equation only takes into account the dipole formed and not higher order polarization. When the electric field gradients are large, higher order terms become relevant, and result in higher forces.
  • the time-dependent equation only applies to lossless particles, because loss creates a lag between the field and the induced dipole. When averaged, the effect cancels out and the equation holds true for lossy particles as well.
  • An equivalent time-averaged equation can be easily obtained by replacing E with the root mean square of E (E fms ), or, for sinusoidal voltages by dividing the right hand side by 2.
  • the (time-averaged) DEP force is 1 :
  • curly brackets The factor in curly brackets is known as the complex Clausius-Mossotti function and contains all the frequency dependence of the DEP force.
  • Particles are levitated above the planar electrode array by negative dielectrophoresis.
  • the levitating negative DEP force, FDEPz (which has been shown to fall exponentially with height above the electrode array) and hydrodynamic lift force are balanced against a sedimentation force, Fgrav, to establish particle equilibrium position, heq within the parabolic flow profile.
  • Fgrav sedimentation force
  • the prior invention apparatus may be modified to detect differing electrical "signatures" of each different phase (i.e. oil vs. fat vs. aqueous solution) as they pass through a channel and actuate a diverting port at the end of the channel to collect and separate the different phases of lipoaspirate.
  • the electrical signatures may be measured by means of: current, current signal shift, conductivity, resistance, impedance, phase shift, capacitance , magnetic shift and use of either direct or alternating current.
  • the problem with the prior art is that cells can clump as they pass through the dielectrophoresis (DEP) equipment and become contaminated.
  • DEP dielectrophoresis
  • the present invention is an improved micro-electrode for DEP.
  • the present invention is used to separate fractions of particles/cells from a heterogeneous population of particles/cells.
  • the cells may be derived from liposuctioned adipose tissue.
  • ITO indium tin oxide
  • ITO surface to allow electrical conditioning of the cells. 3.
  • ITO surface to allow electrical conditioning of the cells. 3.
  • the invention requires less processing time in that cells do not have to be eluted after successive frequency/DEP field drops.
  • the invention can use flanking borderflow chambers or lane barriers to create repulsive DEP fields which create "compartments" that keep collected cells from each array separated and confined for final elution into separate vials.
  • Figure 1 Plan view of a prior art planar DEP electrode.
  • Figure 1 A Partial cross section of a prior art DEP flow chamber.
  • Figure 2 A conceptual representation of the instant invention having flow chambers with successively increasing dielectrophoresis fields represented by increasing shades of serially aligned rectangles in parallel to the direction of flow (arrow) and path of the cell (sphere).
  • Figure 3 Magnified view of planar microelectrode surface and orientation of electrode lines.
  • Figure 4 Conceptual representation of how different DEP fields are generated by connecting separate waveform generators to each flow chamber.
  • Figure 4A Conceptual representation of an alternative embodiment.
  • the flow cells can be connected to one waveform generator and fashioned such that the microelectrodes linearly become closer together to create the gradient DEP field.
  • Figure 4B is a plan view of one of the arrays in Figure 4A illustrating again that the distance between electrodes may be successively reduced.
  • Figure 5 Typical dimensions of the planar microelectrode array of this invention in cross section.
  • Figure 6 Cross sectional view of active 2-D DEP field lines represented in dotted arcs between electrodes. Cells represented in circles are trapped within the DEP fields. Undesired parasitic cell entrapment is noted with aberrant field on far right with "X" through it.
  • Figure 7 Cross sectional view. Typical alternating "+” and "-" charge arrangement of planar microelectrode with overlying ITO surface electrode in off position (no charge).
  • Figure 8 Electrical arrangement of third or 3D electrode connected to power source and ground to waveform generator to provide DEP fields in the vertical dimension.
  • Figure 9 Electrical arrangement of third or 3D electrode connected to power source and ground to measure for impedance and used for diagnostic purposes such as regional saturation of cells.
  • Figure 10 Conceptual cross sectional view of charged/deployed electrodes in the 3D configuration.
  • Figure 1 1 Conceptual representation of how 3D DEP fields are generated by connecting upper ITO surface to waveform generator and using both electrodes on the planar surface as one electrode. DEP fields are represented as dotted lines.
  • Figure 12 Conceptual representation of how a therapeutic laser used in attempting to ablate a trapped cell, will damage the nearby microelectrode if a 3D configuration or third electrode is not used.
  • Figure 13 Representation of how a therapeutic laser used in trying to ablate a trapped cell, will avoid damage to the nearby microelectrode if a 3D configuration or third electrode is used. At the far right the laser is incorrectly focused and delivers energy past the cell and onto the microelectrode beneath which is undesired and represented with an "X".
  • Figure 14 Ideal configuration with microscope (represented by the eye) above the apparatus and an intervening half mirror placed at 45 degrees to allow observation but also passage of light or therapeutic lasers or light (noted at right with the sunburst and dotted line).
  • Figure 15 Ideal configuration of planar electrodes placed in a sequential format relative to flow (black arrow), wherein electrodes 1 and 2 are off (missing generator symbol) and the device is in procurement or "trapping" mode.
  • FIG 16 The same configuration of planar electrodes as in Figure 15, however electrodes 1 and 2 are now “on” producing a cell repulsing DEP confining the cells within the central electrode. The remaining electrodes are "off' (absent generator symbol). Elution fluid may be flooded into the central electrode region and confined cells may be collected separate from cells previously trapped on far left and far right electrode.
  • Figure 17 Shows an alternate configuration in which lane barriers may be skewed to allow cells to elute with only one flow through direction.
  • Figure 18 shows that indigo/methylene blue could be used to stain cells then run it out on the electrode and document with a video (CCD) camera.
  • CCD video
  • Figure 20 shows the same "gradient" like electrode could be used with an overlying set of skewed lane barriers.
  • Figure 19 is an exploded view showing construction of a flow cell of this invention.
  • Figure 20 is a sketch of standard, straight planar array used for DEP. This is prior art.
  • Figure 21 is a sketch of the improved, two-dimensional, interdigitating micro electrode embodiment of this invention for DEP capture of cells.
  • Figure 22 is a sketch of the improved electrode embodiment of Figure 21 with a trailing, mirror electrode.
  • Figure 23 illustrates leading edge separation
  • Figure 24 illustrates the trailing edge collection area.
  • Figure 25 illustrates how captured cells are held in the trailing edge collection area.
  • Figure 26A-7C are representational drawings of the workflow for determining the optimum electrode gap for collecting the cells of interest.
  • FIG. 1 and 1A illustrate prior art 2-D flow chambers 10.
  • Such flow chambers 10 have a top plate 14 and a bottom plate 18.
  • the bottom plate 18 has a microelectrode 22 adhered to its inner surface 26.
  • This microelectrode 22 comprises a plurality of fingers 30a, 30b half of which are electrically connected together at one end 34a and, the other half of which are electrically connected together at the other end 34b.
  • the differently connected fingers 34a, 34b alternate. They could be described as interdigitized. In use one set of fingers 34b is connected to a positive lead and the other set 34a is connected to a negative lead. Thus cells 38 flowing through the chamber 10 in the direction D are exposed to alternating positive and negative electrical fields.
  • FIGs 2 and 3 show that the instant invention may be made of flow chambers 42 similar to those of the prior art with microelectrodes 46 similar to those of the prior art. However, in the instant invention each successive flow chamber 42 has successively increasing dielectrophoresis field in the direction of cell flow D.
  • Figure 4 shows that different DEP fields can be produced by connecting separate waveform generators 50 to each microelectrode 46.
  • Figures 4A and 4B show that, alternatively, the flow chamber 42 can be connected to one waveform generator 50 but are fashioned such that the fingers 54 of their microelectrodes 58 linearly become closer together to create the gradient DEP field.
  • Figure 5 provides typical dimensions of the planar microelectrode array 58 and flow chamber 42 of this invention in cross section.
  • the flow chamber 42 has a top plate 56 and bottom plate 60.
  • Figure 6 shows the 2-D DEP fields, represented by dotted lines, produced by the microelectrode arrays 58 of flow chambers 22, 42 of this and prior art inventions.
  • Cells 38 are trapped within the DEP fields.
  • Undesired parasitic cell entrapment is noted with aberrant field on far right with "X" through it.
  • Figure 7 shows typical alternating "+" and "-" charge arrangement of planar microelectrode 58 with overlying ITO electrode 62 in off position (no charge).
  • the ITO electrode 62 is coated on a substrate 66 which is preferably glass.
  • ITO is electrically conductive yet transparent, which makes it the preferred material for electrodes of this invention.
  • Figure 8 shows the electrical arrangement of third or 3-D electrode 62 connected to power source and waveform generator 50 to provide DEP fields in the vertical dimension.
  • Figure 9 shows the electrical arrangement of third or 3-D electrode 62 connected to power source and ground to measure for impedance and used for diagnostic purposes such as regional saturation of cells.
  • Figure 10 shows the electrical configuration of charged/deployed electrodes in the 3D configuration. In this configuration the bottom plate 60 and the ITO upper surface electrode 62 are connected together and thus have the same polarity while the fingers 54 of the microelectrode array 58 are connected together and have the opposite polarity.
  • Figure 1 1 shows the 3-D DEP fields generated by connecting the upper ITO surface electrode 62 to the waveform generator 50 and using both electrodes on the planar surface as one electrode as shown in Figure 10. DEP fields are represented as dotted lines.
  • Figure 12 shows how a therapeutic laser 70 used in attempting to ablate a trapped cell 38, will damage the nearby microelectrode 54 if a 2D configuration is used or when the third electrode 62 is not used.
  • Figure 13 shows how a therapeutic laser 70 used in trying to ablate a trapped cell 38, will avoid damage to the nearby microelectrode 54 if a 3D configuration or third electrode 62 is used.
  • the laser 70 is incorrectly focused and delivers energy past the cell 38 and onto the microelectrode 54 beneath which is undesired and represented with an "X".
  • Figure 14 shows the ideal configuration of the instant invention with microscope 74 above the apparatus 42 and an intervening half mirror 78 placed at 45 degrees to allow observation and passage of light or therapeutic lasers.
  • Figure 15 shows another ideal configuration of this invention with flow chambers 42 having planar electrodes 58 placed sequentially relative to flow D.
  • the electrodes 58 in flow chambers 1 and 2 are off (missing generator symbol) and the device is in procurement or "trapping" mode.
  • Figure 16 shows the same configuration of as in Figure 15, however the electrodes 58 in flow chambers 1 and 2 are now “on” producing a cell repulsing DEP confining the cells 38 within the central flow chambers 42.
  • the electrodes 58 in chambers 1 and 2 are functioning as lane barriers 58a.
  • the remaining electrodes 58 are "off (absent generator symbol).
  • Elution fluid may be flooded into the central flow chamber 42 and confined cells 38 may be collected separately from cells 38 previously trapped on far left and far right flow chambers 42.
  • Figure 17 shows an alternate configuration in which lane barriers 58a may be skewed to allow cells 38 to be eluted at the ends of the lane barriers 58a.
  • Figure 18 shows that a flow cell 42 with a gradient DEP field could be used with an overlying set of skewed lane barriers 58a.
  • Figure 19 is an exploded view showing construction of a flow chamber 42 of this invention.
  • a gasket 82 In between the top 56 and bottom 60 plates there is a gasket 82.
  • Two holes 86 in the top plate 56 allow for flow of cells through the flow chamber 42.
  • the inlet and outlet ports 86 could be placed elsewhere in the device - for example on the ends, through the gasket 82.
  • cells 1 10 can clump at the center where the sample is being injected. Flow is shown by the arrow 1 14. Cells clump towards the beginning of the DEP field with a standard straight planar array 1 18. This causes debris and unwanted cells to become physically trapped in the clump of cells 1 10. In other words, clumping impedes efficient separation of debris and unwanted cells from the desired cells.
  • Figure 21 shows an improved micro-electrode embodiment 122 for DEP capture of cells which may be alternatively used with the previous invention disclosed in US Patent Application No. 61/525,573.
  • the 2-D, planar, interdigitating, micro-electrode array 122 shown on Figure 21 has a zigzag shape thus facilitating leading edge 132 separation and trailing edge 134 collection.
  • By creating a wedge or angle shaped electrode 124 cells attracted to the electrode array 122 (by F dep ) will be captured, but dissipate laterally (by way of sample flow force) from the center pathway 1 14 of sample injection. This allows debris and unwanted cells to pass down the middle 150 unimpeded.
  • the lateral trailing edge collection area will hold the specimen out of the midplane 150 until all sample is processed and final flush of captured cells is required.
  • the gap G between electrodes 124 may vary.
  • the gap G at the center 136 (within the leading edge 132) may be wider than that at the trailing edge 134, such that the gap starting at the center 136 (leading edge 132) tapers as it approaches the trailing edge 134 (lateral border of array) or vice versa.
  • the microelectrodes 124 may be made of any suitable material such ITO (indium tin oxide), gold, copper, platinum or any electrically conducting surface and placed on a suitable substrate such as (but not limited to) glass, plastic, acrylic, PET, PMMS or PDMS.
  • the electrical signal applied to the array 122 may be direct or alternating current and may be modified to include standard and complex wave forms such as sine wave, triangular, stacked square and progressive sinusoidal, with or without periodic current interruption.
  • Figure 22 shows he same planar interdigitating micro electrode array 122 with leading edge 132 separation and trailing edge 134 collection, adjacent a second electrode 122a which is a mirror image of the first electrode 122, to allow further processing.
  • Figure 23 shows the leading edge 132 separation area 126 of the electrode array 122.
  • Figure 24 shows the trailing edge 134 collection areas 138 of the electrode array 122.
  • Figure 25 shows how captured cells are held in the trailing edge 134 collection area 138 and out of the midline sample flow line 150.
  • Figures 7A-7C are representational drawings of workflow for using a diagnostic modification of the planar 2-D array with widening gap between the electrodes to determine the optimum separation for collecting the cells 1 10 of interest.
  • a small sample of the mixed population of cells 1 10 is first labeled with a visibly detectable dye under microscopy. See Figure 26A. Then the sample mixture of cells 1 10 and labeled cells 1 10a are processed through a single array with multiple regions of electrode gap. In this example there are three progressively narrowing regions of 200, 100 and 50 microns. After the cells 1 10 are captured, an observer 130 is able to see that the pre- labeled cells 1 10a of interest have been captured to a specific gap width. See Figure 26B. In this example the cells of interest 1 10a have been captured on the region with 100 micron gap width. The practitioner then uses a larger filter with the same gap for larger scale processing of the entire specimen 1 10. See Figure 26C.
  • the gap width can be widened to obtain a spectrum of F dep .
  • the cells can be labeled with pan- dye or specific flourochrome labeled Ab, which can be visualized under fluorescence microscopy to determine where that particular band of cells rests. Then the F de p or energy within that segment using the top ITO electrode. In this way the machine can be set to the specific frequency, Vp-p and cartridge gap width.
  • Capacitance can be used to separate raw phases (i.e. aqueous, fatty tissue, oil) using the apparatus and method of US Patent Application Publication No. 2010/01 12084
  • Dielectrophoresis force can vary as a function of distance between two electrodes or by the frequency that is generated between them.
  • F dep capture force in general, the wider the gap of the electrodes the less force is generated.
  • the crossover threshold In other words if a certain cell population is captured with a known electrode gap (e.g. 100 microns) and a certain frequency, the same cell population can also be captured using a larger gap (e.g. 125 microns) but with a higher frequency. But the limit to this is the crossover threshold.
  • Antioxidants may be added to the samples to dielectrophoresis buffer.
  • Antioxidants may be ascorbic acid (vitamin C), glutathione, lipoic acid, uric acid, carotenes, alpha tocopherol (vitamin E), and ubiquinol (coenyme Q).
  • the cells 1 10 may be stimulated in the wavelengths of above 760, and 400 to 450nm (in the infrared range) and light and polarized light (400-2000nm).
  • the electrode arrays 122 described herein can be fashioned as a syringe insert or as a submersible bioreactor.
  • This invention is a method and device for separating charged particles and/or cells 38 in a dielectophoretic liquid medium using gradient array dielectrophoresis.
  • This invention provides the ability to simultaneously provide light therapy while separating the particles and/or cells 38.
  • Flanking microelectrodes or lane barriers 58a may be used to create repulsive DEP fields which create "compartments” that keep collected cells from each array 58 separated and confined for final elution into separate vials.
  • Lipoaspirate fluid may be processed by passage through serial "combs” to mechanically dissociate ADSC from the fat and tissue matrix using two bags of equal volume (similar to an IV bag but tougher) connected together by an intervening adapter.
  • the adapter houses a number of combs in narrowing passages
  • One bag is filled with unprocessed lipoaspirate and compressed manually or automatically from the exterior so that the specimen is moved through the combs into the other bag. The process is repeated until the material has a smooth consistency and there is minimal resistance during fluid transfer between bags. The mixture is then transferred sterilely into a column/receptacle and allowed to settle into oil/fatty tissue/aqueous phases. Then the phases of the lipoaspirate are separated.
  • microcurrent detection wherein: a. A small microcurrent is run between two leads at the base of the column/receptable's exit port; b. Upon contact with the different phases (i.e. oil/fatty tissue/aqueous) differing conductivities and/or resistivities are detected; and c. A computer is programmed to detect this difference and switch a diverting valve at the base of the exit port to respective collecting receptacles/containers.
  • aqueous phase or Stromal Vascular Fraction cells and erythrocytes may be completed by free flow dielectrophoresis wherein: a. a chamber composed of two non-conducting plates, with at least one being transparent, are separated by a small gap (0 to 5mm); b. two electrodes are spaced apart within the gap; c. a current is applied to the two electrodes creating a dieletrophoretic field of between
  • the chamber is filled with physiologic buffer; e. the buffer is continually renewed from the top opening of the chamber; f. the lower chamber has an exit port with a regulator valve to adjust flow through the chamber; g. a sample injection port is placed somewhere between the buffer ports and electrical leads; h. the buffer port drip rates are regulated by drip regulators (similar to an iv or drip irrigation); i. the drip rate of the sample port can be regulated in a similar fashion and rate as the buffer ports; and j. the chamber also has multiple sample collection ports wherein cells may be collected prior to exit from the buffer spill port.
  • This apparatus may be manufactured of disposable materials. This apparatus may be formed in a "closed system" to minimize risk of contamination and comply with GMP standards.
  • the chamber may be modified to include: i. a diffraction gradient to accelerate separation which is composed of microscopic barriers or posts which separate cells by size; ii. LEDs placed in front or in back of the panels to irradiate and optimize ADSC so that they are actively being separated thus minimizing processing time; and/or iii. cooling elements added in the front and in back of the panels to prevent overheating from the electrical leads.
  • Additional electrodes may also be placed to compensate for dielectrophoresis field unbalance due to non-rectangular internal shape of the chamber 42.
  • the triangulated shape of the upper and lower aspects of the buffer chamber causes the electric field strength to be weaker at the apical and lower pole of the electrodes.
  • multiple electrodes with greater voltage or current can be applied towards the distal ends of the dielectrophoresis field.
  • Residual may be further processed using the same apparatus as above, but this time used for separating stem cell related proteins.
  • Residual non cellular fluid
  • this time used for separating stem cell related proteins.
  • planar micro-electrodes 58 which create a 2-D dielectrophoresis field:
  • a lateral laser 78 or IPL or LEDs optically focused to reflect off the half mirror 78 and onto middle of flow cell 42 thus not on the 2D electrodes 54;
  • a central processing unit attached to all electrodes to facilitate switching between 2- D, 3D and impedance testing mode
  • frequencies for all electrodes 58 may be in a range from 1 Hz to 100MHz.
  • chambers 42 connected to separate collection ports. 13. flow cell 42 and chamber flooded with suspension of cells/particles 38 in dielectrophoresis buffer and delivered at a constant rate to minimize cell/particle 38 shearing when cells/particles 38 are trapped on electrodes 58.
  • the final collection of cells/particles 38 may be done with physiologic buffer or solvent.
  • the microelectrodes 58 may be comprised of any suitable material such ITO (indium tin oxide), gold, copper, platinum or any electrically conducting surface and placed on a suitable substrate such as (but not limited to) glass, plastic, acrylic, PET, PMMS or PDMS.
  • ITO indium tin oxide
  • gold gold
  • copper gold
  • platinum any electrically conducting surface and placed on a suitable substrate such as (but not limited to) glass, plastic, acrylic, PET, PMMS or PDMS.
  • the electrode 58 pitch may be modified such that the space between microelectrodes 54 becomes progressively wider or narrower to create a gradient DEP field.
  • the electrical signal or waveform may be modified to utilize complex forms (i.e. triangular, stacked square, progressive sinusoidal) other than a standard sine wave.
  • the microelectrode array 58 may be modified or adapted to fit within a syringe.
  • the microelectrode array 58 may be modified to fit within a bioreactor. While not prior art to this invention an example of an analytical device fitted into a bioreactor is the BioSep, manufactured by Applikon Biotechnology B.V. , with headquarters at Nieuwpoortweg 12, P.O. Box 101 , 3100 AC Schiedam, The Netherlands.
  • FFE free flow dielectrophoresis
  • the lane barriers 58a may be skewed to allow cells 38 to elute with only one flow through direction.
  • the distance between electrodes 54 may be successively expanded to create a gradient of DEP field. This could be used potentially as a quick and easy diagnostic test to cellularly "profile" the SVF mixture.
  • a series of three flow chambers were connected in series by flow (not electrically). Each chamber was made of an ITO coated glass plate, laser etched to produce interdigitating microelectrodes with lines having a width of 50 microns and the gap or pitch of 100 microns. A third ITO coated glass plate was used as a cap for each flow chamber thus allowing for creation of a 3D dielectrophoresis field. A barrier of 350 microns was placed between the ITO coated glass plates.
  • Each chamber was sealed with a polydimethylsiloxane (PDMS) gasket which circumscribed a 2.5cm x 7cm x 350 micron flow cell.
  • PDMS polydimethylsiloxane
  • the entire apparatus was mounted on clear Plexiglas ® seated atop an inverted microscope.
  • a flat panel of LEDs to stimulate the collagen was placed directly over each flow cell when the microscope was not in use.
  • Infranatant from lipoaspirate was centrifuged at 300 relative centrifugal force (RCF). The resulting pellet was washed and re-suspended in dielectrophoresis buffer containing sucrose, dextrose, tissue culture media and balanced with phosphate buffer solution (PBS) to achieve a conductivity of 30 milli Sieverts per meter (mS/m). Final concentration was approximately 1 x10 6 cells/mL
  • the flow cells were flooded with DEP buffer and allowed to soak 10 minutes prior to starting the waveform generators.
  • the 2-D waveform generator was set at 30, 60, 200 kHz for the 1 st, 2 nd and 3 rd flow chamber electrode set respectively.
  • the 3D waveform generator was set to turn on every 10 minutes, for 2 minutes at X frequency, during which time the 2-D waveform generators were off.
  • the re-suspended pellet (sample) was injected by controlled syringe pump at a rate of 1 cc/min. Impedance measurements were obtained at the end of each 3-D cycle. A soak cycle in which only clean DEP buffer was injected for 30 minutes. Direct visualization of entrapped cells was achieved light microscopy on an inverted stage.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Pathology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Electrochemistry (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Optics & Photonics (AREA)
  • Microelectronics & Electronic Packaging (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Electrostatic Separation (AREA)

Abstract

La présente invention concerne un procédé et un dispositif selon lesquels une population hétérogène de cellules ou de particules est introduite dans une série de chambres, dotées de champs de diélectrophorèse croissant successivement produits par des réseaux de microélectrodes planaires, et séparée de cette façon selon la charge et la taille. Une troisième électrode claire ou translucide recouvrant le réseau planaire définit le plafond de la chambre de flux et permet l'introduction simultanée de thérapie lumineuse aux cellules tandis qu'elles sont séparées. Une adjonction latérale d'électrodes planaires aux électrodes de capture peut être employée à des fins de diélectrophorèse répulsive afin de confiner et garder des cellules piégées séparées durant la phase finale de collecte et d'élution. Le processus complet peut être surveillé électroniquement par un test d'impédance ou visuellement par une microscopie in situ et en temps réel sans perturber le processus de séparation.
PCT/US2012/051484 2011-08-19 2012-08-17 Séparation par diélectrophorèse de gradients (grads) avec thérapie lumineuse concomitante WO2013028573A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA2845713A CA2845713A1 (fr) 2011-08-19 2012-08-17 Separation par dielectrophorese de gradients (grads) avec therapie lumineuse concomitante
JP2014527210A JP2014531199A (ja) 2011-08-19 2012-08-17 並行光線療法を伴う傾斜アレイ誘電泳動分離(GrADS)
IL230985A IL230985A0 (en) 2011-08-19 2014-02-13 Dielectrophoresis separation in a gradient array (grads) with simultaneous light treatment

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161525573P 2011-08-19 2011-08-19
US61/525,573 2011-08-19
US201161545015P 2011-10-07 2011-10-07
US61/545,015 2011-10-07

Publications (2)

Publication Number Publication Date
WO2013028573A1 true WO2013028573A1 (fr) 2013-02-28
WO2013028573A4 WO2013028573A4 (fr) 2013-04-25

Family

ID=47746791

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/051484 WO2013028573A1 (fr) 2011-08-19 2012-08-17 Séparation par diélectrophorèse de gradients (grads) avec thérapie lumineuse concomitante

Country Status (4)

Country Link
JP (1) JP2014531199A (fr)
CA (1) CA2845713A1 (fr)
IL (1) IL230985A0 (fr)
WO (1) WO2013028573A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104148180A (zh) * 2014-08-19 2014-11-19 阮海生 一种高效介电电泳净化单元
CN104174503A (zh) * 2014-08-19 2014-12-03 阮海生 一种新型dep电极结构、形成的电极板和电极阵列
CN105683745A (zh) * 2013-08-29 2016-06-15 阿波赛尔公司 用于目标颗粒的分离、捕获和分子分析的方法和装置
WO2020245275A1 (fr) * 2019-06-05 2020-12-10 Jaguar Land Rover Dispositif de manipulation d'une substance, véhicule et ensemble le comprenant, et procédé d'utilisation du dispositif
US11662332B2 (en) 2016-12-05 2023-05-30 Georg-August-Universitaet Goettingen Stiftung Oeffentlichen Rechts, Universitaetsmedizin Method of producing transparent biological preparations for examination by light microscopy

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040058423A1 (en) * 2002-05-03 2004-03-25 Nancy Albritton Fast electrical lysis of cells and rapid collection of the contents thereof using capillary electrophoresis
US20040226819A1 (en) * 2003-05-13 2004-11-18 Talary Mark Stuart Dielectrophoresis apparatus
US7029564B1 (en) * 1999-07-20 2006-04-18 University Of Wales, Bangor Dielectrophoretic apparatus and method
US20060289341A1 (en) * 2003-03-17 2006-12-28 Evotec Ag Methods and devices for separting particles in a liquid flow
US20070128686A1 (en) * 2001-03-22 2007-06-07 Gaoshan Jing Cell isolation method and uses thereof
US20110108422A1 (en) * 2008-04-03 2011-05-12 The Regents Of The University Of California Ex vivo multi-dimensional system for the separation and isolation of cells, vesicles, nanoparticles and biomarkers

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7029564B1 (en) * 1999-07-20 2006-04-18 University Of Wales, Bangor Dielectrophoretic apparatus and method
US20070128686A1 (en) * 2001-03-22 2007-06-07 Gaoshan Jing Cell isolation method and uses thereof
US20040058423A1 (en) * 2002-05-03 2004-03-25 Nancy Albritton Fast electrical lysis of cells and rapid collection of the contents thereof using capillary electrophoresis
US20060289341A1 (en) * 2003-03-17 2006-12-28 Evotec Ag Methods and devices for separting particles in a liquid flow
US20040226819A1 (en) * 2003-05-13 2004-11-18 Talary Mark Stuart Dielectrophoresis apparatus
US20110108422A1 (en) * 2008-04-03 2011-05-12 The Regents Of The University Of California Ex vivo multi-dimensional system for the separation and isolation of cells, vesicles, nanoparticles and biomarkers

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105683745A (zh) * 2013-08-29 2016-06-15 阿波赛尔公司 用于目标颗粒的分离、捕获和分子分析的方法和装置
EP3039416A4 (fr) * 2013-08-29 2017-04-26 Apocell, Inc. Procédé et appareil pour l'isolement, la capture et l'analyse moléculaire de particules cibles
CN104148180A (zh) * 2014-08-19 2014-11-19 阮海生 一种高效介电电泳净化单元
CN104174503A (zh) * 2014-08-19 2014-12-03 阮海生 一种新型dep电极结构、形成的电极板和电极阵列
US11662332B2 (en) 2016-12-05 2023-05-30 Georg-August-Universitaet Goettingen Stiftung Oeffentlichen Rechts, Universitaetsmedizin Method of producing transparent biological preparations for examination by light microscopy
WO2020245275A1 (fr) * 2019-06-05 2020-12-10 Jaguar Land Rover Dispositif de manipulation d'une substance, véhicule et ensemble le comprenant, et procédé d'utilisation du dispositif

Also Published As

Publication number Publication date
IL230985A0 (en) 2014-03-31
JP2014531199A (ja) 2014-11-27
CA2845713A1 (fr) 2013-02-28
WO2013028573A4 (fr) 2013-04-25

Similar Documents

Publication Publication Date Title
US11519877B2 (en) Devices and methods for contactless dielectrophoresis for cell or particle manipulation
Wu et al. High-throughput separation, trapping, and manipulation of single cells and particles by combined dielectrophoresis at a bipolar electrode array
JP3586279B2 (ja) ダイエレクトロホリティクによる分離装置及び分離方法
Pethig Dielectrophoresis: An assessment of its potential to aid the research and practice of drug discovery and delivery
Goater et al. Electrorotation and dielectrophoresis
WO2013028573A1 (fr) Séparation par diélectrophorèse de gradients (grads) avec thérapie lumineuse concomitante
US8222014B2 (en) Planar electroporation apparatus and method
Das et al. A microfluidic device for continuous manipulation of biological cells using dielectrophoresis
JP2000505545A (ja) 誘電電気泳動とフィールドフローフラクショネーションを用いる分別方法と装置
WO2012048230A2 (fr) Dispositifs et procédés de diélectrophorèse
WO2020014264A1 (fr) Micro dispositifs d'électroporation à circulation continue et procédés de transfection de cellules
JP2009539096A (ja) 単細胞及びその小集合体を操作するための方法及び装置
Kirschbaum et al. Highly controlled electrofusion of individually selected cells in dielectrophoretic field cages
KR100738071B1 (ko) 농도구배발생부가 구비된 유전영동 장치, 그를 이용한물질의 분리방법 및 물질 분리의 최적 조건을 탐색하는 방법
Nascimento et al. Dielectrophoretic sorting on a microfabricated flow cytometer: Label free separation of Babesia bovis infected erythrocytes
BR112019012738A2 (pt) dispositivo de matriz de micro-elétrodo multiplanar e métodos de fabricação e uso do mesmo
Fuhr et al. Cell motion in time-varying fields: principles and potential
WO2018009892A1 (fr) Diélectrophorèse isomotrice pour analyse diélectrique de sous-populations de particules
Yuan et al. A resettable in-line particle concentrator using AC electrokinetics for distributed monitoring of microalgae in source waters
Trainito Study of cell membrane permeabilization induced by pulsed electric field–electrical modeling and characterization on biochip
Heida Electric field-induced effects on neuronal cell biology accompanying dielectrophoretic trapping
Salmanzadeh et al. Isolation of rare cancer cells from blood cells using dielectrophoresis
Deivasigamani et al. A correlation of conductivity medium and bioparticle viability on dielectrophoresis‐based biomedical applications
WO2009027927A2 (fr) Dispositif diélectrophorétique destiné à la manipulation de particules
Cemazar et al. Electrical manipulation and sorting of cells

Legal Events

Date Code Title Description
DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12826491

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 230985

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2845713

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014527210

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12826491

Country of ref document: EP

Kind code of ref document: A1