EP4143292A2 - Dispositif et procédé de liposuccion et de traitement de tissus intégrés automatisés - Google Patents

Dispositif et procédé de liposuccion et de traitement de tissus intégrés automatisés

Info

Publication number
EP4143292A2
EP4143292A2 EP21735386.1A EP21735386A EP4143292A2 EP 4143292 A2 EP4143292 A2 EP 4143292A2 EP 21735386 A EP21735386 A EP 21735386A EP 4143292 A2 EP4143292 A2 EP 4143292A2
Authority
EP
European Patent Office
Prior art keywords
compartment
processing
tissue
processing unit
side wall
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21735386.1A
Other languages
German (de)
English (en)
Inventor
Christopher ALT
Andreas Mues
Jakob Schmid
Eckhard U. Alt
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
SciCoTec GmbH
Original Assignee
SciCoTec GmbH
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 SciCoTec GmbH filed Critical SciCoTec GmbH
Publication of EP4143292A2 publication Critical patent/EP4143292A2/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M45/00Means for pre-treatment of biological substances
    • C12M45/09Means for pre-treatment of biological substances by enzymatic treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/71Suction drainage systems
    • A61M1/76Handpieces
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/84Drainage tubes; Aspiration tips
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/89Suction aspects of liposuction
    • A61M1/892Suction aspects of liposuction with treatment of the collected fat
    • A61M1/893Suction aspects of liposuction with treatment of the collected fat with extraction of specific components, e.g. of stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/89Suction aspects of liposuction
    • A61M1/895Suction aspects of liposuction with means for reinjection of collected fat
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/32Surgical cutting instruments
    • A61B17/320068Surgical cutting instruments using mechanical vibrations, e.g. ultrasonic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B17/00Surgical instruments, devices or methods, e.g. tourniquets
    • A61B17/32Surgical cutting instruments
    • A61B17/320068Surgical cutting instruments using mechanical vibrations, e.g. ultrasonic
    • A61B2017/32007Surgical cutting instruments using mechanical vibrations, e.g. ultrasonic with suction or vacuum means
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B2217/00General characteristics of surgical instruments
    • A61B2217/002Auxiliary appliance
    • A61B2217/005Auxiliary appliance with suction drainage system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2202/00Special media to be introduced, removed or treated
    • A61M2202/08Lipoids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/10General characteristics of the apparatus with powered movement mechanisms
    • A61M2205/106General characteristics of the apparatus with powered movement mechanisms reciprocating

Definitions

  • the disclosure generally relates to a centrifuge-based processing method and device, and more particularly to a multistage centrifuge-based processing method and device for automated processing of cell extraction and recovery in a single processing unit.
  • the present disclosure describes a tissue processing unit and method for processing tissues to extract cells with minimum human handling.
  • the processing unit comprises a processing compartment having a processing side wall; at least one wash fluid compartment having a washing side wall, wherein the wash fluid compartment is located radially outside of the processing compartment and separated by the processing side wall, wherein a first opening is formed at the top of the processing side wall to allow fluidic communication between the processing compartment and the wash fluid compartment; and at least one concentration compartment, wherein the concentration container is located radially outside of the wash fluid compartment and separated by the washing side wall, wherein a second opening is formed at the top of the washing side wall to allow fluidic communication between the wash fluid compartment and the concentration compartment.
  • the different slopes and shapes of the sidewalls of the chambers facilitate processing, breaking down and separation of the initial tissue into its component.
  • a part of the liquid previously present in the processing compartment can initially be separated from the processed tissue and flows into a waste or washing compartment.
  • the separation and recovery of different components of tissues in the processing chamber is achieved through repetitive stop-and-go cycles and subsequent application of different cycles of centrifugation forces.
  • the processed tissues and cells can be further washed and the remaining enzymatic activity can be diluted.
  • Separation and recovery of cells from debris, washing fluid including cell, erythrocyte and collagen fragments and from liquid fat is based on the different specific density and gravity of the substances and based on application of different acceleration forces applied to the tissue components in the respective chambers.
  • the amount of clean washing fluid is preferably calculated such that after dilution the concentration of enzymatic rest activity from collagenases and proteases will be reduced to a level safe and acceptable for immediate use.
  • the sidewalls of the bowls are designed to have different slopes with openings placed on the sidewalls. Based on the specific mass of the processed tissue and cells and the processing fluid in the inner processing chamber, we can calculate the centrifugal g- force such that only the washing fluid would travel along the sidewall high enough to reach the openings and could enter the concentration chamber.
  • additional sealable openings are provided on the lid.
  • One of these sealable openings can be connected with a liposuction syringe for extracting adipose tissues directly from a subject for a contactless transfer of the adipose tissues into the processing chamber, thus minimizing the chance of contamination.
  • Additional openings can be used to deposit the washing fluid into the washing and concentration chambers.
  • the plurality of openings comprises a first array of perforations and a second array of perforations or filters, wherein the first array of perforations are formed on the side wall at a location lower than the second array of perforations.
  • the size of the first array of openings allows liquid to pass through, but not the cells in order to keep the cells inside the processing chamber.
  • Second array of openings further up the sidewall of the processing chamber have a diameter that the cells can pass through, and centrifugation at a higher g-force can spin the cells to reach the secondary openings, where the cells will travel pass the secondary openings into the dilution chamber.
  • the first array of openings has a diameter of 1-5 pm. In one embodiment, the second array of openings have a diameter of 40-200 pm.
  • the processing compartment is generally triangular in shape, wherein a first distance from the bottom recess to a vertex of the triangular shape is greater than a second distance from the bottom recess to a side of the triangular shape.
  • the processing compartment has bottom baffles to improve tissue breakdown during agitation.
  • the concentration compartment is a detachable concentration container. In one embodiment, the detachable concentration containers are threaded to the processing unit.
  • the tissue processing unit further comprises a lid that covers and seals the first, second and third bowls.
  • the lid further comprises a first inlet that allows the injection of tissues to the processing chamber, and a second inlet that allows the introduction of washing fluid into the wash fluid chamber.
  • a filter is provided to cover the second opening between the wash fluid compartment and the concentration compartment.
  • the filter can be a filter mesh having mesh openings of 40-300 pm.
  • a siphon structure is provided between the second opening and the concentration compartment to prevent fluid backflow.
  • a concentrated cellular suspension can be recovered from a delivery device such as a syringe, for injection into the recipient by, for example, subcutaneous, intravenous, intramuscular, or intraperitoneal techniques.
  • the regenerative cells can be injected into blood vessels for systemic or local delivery, into tissue (e.g., cardiac muscle or skeletal muscle), into the dermis (subcutaneous), into tissue space (e.g., pericardium or peritoneum), or other location. Injection of the regenerative platform can result in an area near the injection site being augmented, repaired, having reduced inflammation, reduced pain, and combinations thereof.
  • one or more additives are added to the cells before administration.
  • the cells can be mixed with other cells, biologically active compounds, biologically inert compounds, demineralized bone, a matrix or other resorbable scaffold, one or more growth factors, or other additive that can enhance the delivery, efficacy, tolerability, or function of the cell population.
  • the recovery of a cellular preparation such as from tumor or other tissues, can be used for further diagnostic measures.
  • a method for automated extraction of adipose- derived stem cells from adipose tissue comprises the steps of: providing a centrifuge processing unit discussed above and a centrifuge machine; loading adipose tissue, enzymatic reagents and a fluid such as saline or calcium containing ringer solution into the processing chamber; loading washing fluid into the middle washing chamber; placing the centrifuge processing container into the centrifuge processing unit; agitating the tissue together with an enzyme-containing fluid in an inner processing compartment by applying repetitive acceleration and deceleration cycles of a first g-force of 0.5 to 3 g in order to dissociate the tissue; accelerating the processed tissue from step a) with a second g-force greater than the first g-force sufficient to move the processed tissue from the processing compartment into a washing compartment placed radially outside the processing compartment, wherein the washing compartment is prefilled with a washing fluid; allowing the processed tissue to be diluted and was
  • a vibrating liporator comprises: a housing having a cannula channel, a cylindrical piston space, an outlet, and a vacuum port, wherein the vacuum port is fluidically connected to the cylindrical piston space; a piston located in the cylindrical piston space; and a cannula located within the cannula channel and sealingly connected to the outlet through the piston such that a flow path from the cannula to the outlet is sealed, wherein the piston engages with a spring such that when a vacuum creates a negative pressure inside the cylindrical piston space through the vacuum port, the piston moves to compress the spring.
  • the vibrating liporator is disposable.
  • a closed system for processing and recovering cellular component from mammalian tissue, without contacting ambient environment.
  • the system comprises a processing unit having: a processing compartment having a processing side wall; at least one wash fluid compartment having a washing side wall, wherein the wash fluid compartment is located radially outside of the processing compartment and separated by the processing side wall, wherein a first opening is formed at the top of the processing side wall to allow fluidic communication between the processing compartment and the wash fluid compartment; at least one concentration compartment, wherein the concentration container is located radially outside of the wash fluid compartment and separated by the washing side wall, wherein a second opening is formed at the top of the washing side wall to allow fluidic communication between the wash fluid compartment and the concentration compartment; a top cover having a first inlet and a second inlet, wherein the first inlet provides fluidic communication with the processing compartment, and the second inlet provides fluid communication with the wash fluid compartment.
  • the closed system further comprises a tissue-extraction device operatively coupled to the first inlet on
  • the tissue extraction device is powered by vacuum to induce a vibrating cannula that is connected to a moving piston.
  • the piston is located within a cylinder and the vacuum retracts the piston and the cannula against a spring. By releasing the vacuum the spring moves the piston and canula forward again, thus vibrating the device. Movements of the cannula at a frequency between 5 and 50 Hz can be achieved.
  • the same vacuum source that also applied the vacuum suction to the tip of the canula is used to drive the vibration of the piston and cannula.
  • the tissue-extraction device is the vibrating liporator described herein.
  • the vacuum-driven liposuction device preferably comprises a handheld piece formed from polycarbonate, and the cannula made from stainless steel. Due to its simplified construction, material and external power source, the device can be made as a single use disposable device.
  • a power-assisted disposable tissue extraction device comprises: a housing having a cannula channel, a cylindrical piston space, an outlet, and a vacuum port, wherein the vacuum port is fluidically connected to the cylindrical piston space; a piston located in the cylindrical piston space; and a cannula located within the cannula channel and through the piston, said cannula having a distal end and a proximal end, wherein the proximal end is sealingly connected to the outlet such that a flow path from the distal end of the cannula to the outlet is sealed, wherein the piston engages with the spring such that when a vacuum creates a negative pressure inside the cylindrical space on the piston through the vacuum port, the piston moves and compresses the spring, wherein the spring decompresses to push the piston and cannula back when the vacuum releases, thereby creating a vibrational movement of the cannula with repetitive cycles of vacuum and vacuum release to facilitate the recovery of tissue from
  • a method of recovering cells from a mammalian tissue using a single disposable device comprises: filling the mammalian tissue into a processing chamber; processing the mammalian tissue in the processing chamber into cells and non-cell tissue fragments; separating the cells from the non-cell tissue fragments using multi-stage centrifugation of different centrifugal forces; and recovering the cells in a sterile container in a concentrated low fluid volume of 2 to 10 ml.
  • vial or “bowl” refers to a bowl-like container that is capable of holding liquid with a sealed bottom.
  • processing refers to the step of treating a tissue with an enzymatic fluid and/or mechanical force to digest and/or breakdown the tissue.
  • Enzymes may include protease, collagenase, and mixtures thereof. Heating may also be applied to facilitate processing.
  • FIG. 1 A-C Perspective view of the processing unit of this disclosure.
  • FIG. 2 Top view of the processing unit without the lid.
  • FIG. 3 Cross-sectional view of the processing unit at the sidewall.
  • FIG. 4A Alternative view of the lid.
  • FIG. 4B The lid in conjunction with a liposuction syringe.
  • FIG. 5 Flow diagram of the method of this disclosure.
  • FIG. 6A Cross-sectional view of the processing unit according to another embodiment of this disclosure.
  • FIG. 6B Top view of the processing unit with the cover on according to another embodiment of this disclosure.
  • FIG. 6C Top view of the processing unit without the cover according to another embodiment of this disclosure.
  • FIG. 7A Cross-sectional view of the processing unit illustrating step 1 of the method of processing: preparation.
  • FIG. 7B Cross-sectional view of the processing unit illustrating step 2 of the method of processing: processing cells.
  • FIG. 7C Cross-sectional view of the processing unit illustrating step 3 of the method of processing: washing and concentrating.
  • FIG. 8A Perspective view of an extraction device for extracting adipocytes.
  • FIG. 8B Cross-sectional view of the extraction device of FIG. 8 A.
  • FIG. 9 Illustration of the method of this disclosure to extract adipose tissue and transfer it into the processing unit of this disclosure without contacting the atmosphere.
  • the disclosure provides novel centrifuge processing container to be used in an automated system to extract stem cells from adipose tissue.
  • This disclosure describes methods and apparatus for recovery of cells from tissues, including human and animal tissues such as canine, feline, equine, bovine, ovine, or porcine tissues.
  • the methods and apparatus described herein are particularly useful for recovery of cells from adipose tissue obtained from, for example, liposuction (i.e., lipoaspirate), including suction assisted, laser or pressure assisted, or ultrasound assisted liposuction, and combinations thereof.
  • the methods and apparatus described herein can be used to isolate stem cells, progenitor cells, hematopoietic cells, or fully differentiated cells from adipose tissue.
  • the method and apparatus of this disclosure can extract adipose tissue from a subject and transfer it into the processing unit of this disclosure in a completely sterile fashion, thereby ensuring the thus-extracted adipose tissue is free of contamination.
  • the extracted tissue can be processed, washed, and concentrated all within the same unit through sequential introduction/removal of buffers and automated centrifugation. The resulting cells are thus ready for further therapeutic use on site.
  • the methods and apparatus described herein can be used on site to prepare cellular compositions for administration to a patient (e.g., autologous administration).
  • the methods and apparatus described herein can be used to recover regenerative cells from a patient, referred to herein as a regenerative platform, that can be prepared for administration and then administered (e.g., injected or surgically implanted) back to the patient from which the cells were recovered.
  • the methods and apparatus also can be used to prepare cellular compositions for growth studies, gene expression studies, differentiation studies, or other research purposes.
  • the methods and apparatus described herein can be used to recover regenerative cell populations (e.g., stem cells) such that the cells can be banked, for example, by cry opreserving the cells with an appropriate medium.
  • regenerative cell populations e.g., stem cells
  • the single container for processing and separating mammal tissue is shown in Figure 1A.
  • the single container 1 for processing consists of three separate bowls 11, 12, 13 aligned in a concentric fashion, with bowl 11 in the inner most surrounded by intermediate bowl 12 that is in turn surrounded by the outermost bowl 13.
  • multiple chambers are created individual chambers, 2, 3, 4 in between each bowl, including the processing chamber 2 inside the innermost bowl 11, the washing chamber 3 between the outer wall of the innermost bowl 11 and the inner wall of the intermediate bowl 12, and the dilution/concentration chambers 4 between the intermediate bowl 12 and the outermost bowl 13.
  • the concentric configuration puts all three bowls 11, 12, 13 revolving around a rotational axis 8 that may adapt to a shaft of a centrifugation machine (not shown).
  • the processing chamber 2 is surrounded by the washing chamber 3, and the washing chamber 3 is surrounded by the dilution/concentration chamber 4.
  • the inner wall of the processing chamber 2 has uneven surface, or ribs, such that when the processing unit is being rotated during centrifugation, the uneven surface can facilitate breaking up of tissues.
  • a predetermined amount of washing fluid is deposited in the washing chamber 3.
  • a predetermined amount of dilution fluid is deposited in the dilution/concentration chambers 4.
  • the overall shape of the integrated chamber and unit is triangular with three distal ends. Each of the distal ends has a point of maximal acceleration upon centrifugation, such that the openings 9 are experiencing the highest gravitational force based on the distance to the rotational axis 8.
  • the processing unit is firmly covered by a lid 5 that covers the walls of the intermediate bowl 12, the washing chamber number 3, the walls of the outermost bowl 13 and the dilution/concentration chamber 4.
  • the upper wall of the inner most bowl 11 and the processing chamber 2 are covered by and connected to the lid 5.
  • openings 9 which are closed by a valve or a swappable luer lock, to which a syringe can be connected in order to remove the cells after the cellular preparation further detailed below.
  • the inner center of the processing unit is connected to the unsterile outside by an opening 15 in lid 5.
  • the flow of operations occurs in a way that processing chamber number 2 is filled through the luer locked entry 16, and the washing chamber is filled through an opening 17.
  • Each of those openings can be initially closed by a luer stopcock.
  • Filling of the inner processing chamber 2 occurs through opening number 16 with tissue material, preferably liposuction of adipose tissue through the opening, the liposuction material is agitated together with an enzyme and fluid within the chamber number by repetitive low g force accelerations in a range from 0.5 to 3 g.
  • FIG. 4B shows the lid 5 used in conjunction with a liposuction syringe and a three-way valve.
  • the three-way valve is turned to allow vacuum suction between the needle and the syringe.
  • the three-way valve is turned to allow direct transfer of the adipose tissue from the syringe into the processing chamber through opening 16 on the lid 5.
  • a calcium containing fluid is added to the processing chamber 2 together with an enzyme, preferably consisting of an enzyme mixture of collagenase I, collagenase II, and a protease.
  • the processing unit is placed within a centrifugation system that is contained in a controlled heated enclosure, such as previously described in the U.S. Pat. No. 8951513.
  • the processing of tissue by agitation can be effected in about 20 to 40 minutes.
  • elements 85 are helpful for better mixing and agitation of the processed fluid and tissue that are helpful to create vertical forces that complement the differential horizontal accelerations as represented by the distances di between 82-83 and d2 between 82-84.
  • Figure 3 is a side view cut through the unit depicting the left half part of the processing chamber 2, the washing chamber 3, and the dilution chamber 4 on top.
  • the side walls and the whole unit is made preferably of polycarbonate, however other materials may also be used, as long as they are easy to manufacture and/or biocompatible.
  • In the grey area in the middle is the unsterile outside field, through which the unit is firmly attached to the motor.
  • the sidewall 21 of the processing chamber 2 has a slope that changes from the bottom to the top.
  • This slope at sidewall 21 is different comparing to the slope at sidewall 31 of the washing chamber 3 that allows a higher acceleration force; first due to the greater distance to the axis and second due to the lower or less steep incline of the wall, resulting in a relatively higher vertical vector as part of the centrifugal vector.
  • FIGS. 6A-C depict an alternative processing unit of this disclosure.
  • FIG. 6A is a cross-sectional view of the processing unit
  • FIGS. 6B, 6C are top views of the processing unit with and without the cover, respectively.
  • FIG. 6 A the processing unit 100 is engaged on a rotor 700 of a centrifuge as discussed above.
  • the processing unit 100 of this embodiment is different from the one shown in FIGS. 1-4 above.
  • the processing unit 100 has a processing compartment 101 for holding a process fluid and tissues for processing, a wash fluid compartment 200 for holding wash fluid, and a cover 600 that complete covers the processing unit 100 to provide an isolated space within the processing unit 100, except for two inlets.
  • a first inlet 601 is located on the cover 600 corresponding to the processing compartment 101, and a second inlet 602 is located on the cover 600 corresponding to the wash fluid compartment 200. See also FIG. 6B.
  • the first inlet 601 enables a sterile connection with a lipoaspirator (further discussed below with regard to FIGS. 8A-B) for transferring the extracted tissue into the processing compartment 101.
  • the second inlet 602 enables a sterile injection of wash fluid into the wash fluid compartment 200.
  • the processing compartment 101 has multiple baffles 103 at the bottom of the space to increase the efficiency of physically breaking down the tissue by shear force. See also FIG. 6C.
  • the processing compartment 101 has a first wall 105 of a first slope, and a second wall 107 of a second slope, wherein the first slope is steeper than the second slope.
  • the wash fluid compartment 200 is separated from the processing compartment 101 by the first wall 105, and an opening 109 is provided at the top of the first wall 105 to allow fluidic communication between the two compartments 101, 200.
  • a concentration container 500 is removably attached to the processing unit 100 through, for example, thread 501.
  • other mechanism to connect the concentration container 500 to the processing unit 100 may also be used without deviating from this disclosure.
  • the concentration container 500 has a fluid path from the wash fluid compartment 200 through an opening 201, a siphon structure 400, and a one-way valve 502.
  • a filter mesh 300 is provided at the opening 201, wherein the filter mesh 300 has mesh openings large enough for cells to pass through and small enough to block the digested tissue and debris.
  • a filter mesh having mesh opening of about 40 to 200 microns can be used.
  • the one-way valve 502 serve to prevent leakage from the processing unit 100 after the concentration container 500 is detached.
  • the detachable concentration container is advantageous in that once the processing/washing/concentrating steps are completed, the resulting cells are collected within the concentration container 500 and is ready for the next procedure or being stored away for future use. There is no further need for the processing unit 100, as it is intended for single-use only.
  • the slope of the first wall 105 is steeper than the slope of the second wall 107 such that during the processing step with lower centrifugal speed, the processing fluid and tissue will stay in the processing compartment 101 without overflowing into the wash fluid compartment 200 through the opening 109. See FIG. 6C. It is only when sufficient centrifugal speed is reached can the processing fluid as well as the tissue reach high enough on the first wall 105 to pass through the opening 109 into the wash fluid compartment 200.
  • the following discussion describes the method of extracting adipose tissue from a subject, processing the adipose tissue to release cells from the tissue, washing the cells, and increasing the concentration of the cells.
  • the method will be illustrated with reference to FIGS. 5-9.
  • the adipose tissues are extracted by a syringe or liposuction handpiece 800, as shown in FIG. 8A.
  • the liposuction handpiece is disposable.
  • FIG. 8B shows the cross section of the liposuction handpiece 800, where a liposuction cannula 802 is connected to a lipoaspirate outlet 806 through a piston 803 and tubing 805.
  • the piston 803 sits inside a cylindrical space 810 and engages a spring 804.
  • the cylindrical space 810 that is fluidically connected to a vacuum port 807 that can be connected to a vacuum 809 through a vacuum modulating valve 808 (see FIG. 9).
  • the vacuum 809 creates a negative pressure that pulls the piston 803 towards the vacuum port 807 and compresses the spring 804, which then pushes the piston 803 back to its original position inside the cylindrical space 810.
  • the back-and -forth motion of the piston 803 creates a vibration onto the liposuction handpiece 800 and the cannula 802, thereby allowing easier extraction of adipose tissue from the patient.
  • the bodily fluid and tissues extracted from a subject will first flow through the lipoaspirate outlet 806 and into a fat harvesting apparatus 900, such as LipiVage® by Genesis Biosystems, Inc.
  • the fat harvesting apparatus separates fat tissue from unwanted fluid.
  • the fat tissue is then transferred into the processing unit 100 through the first inlet 601 discussed above.
  • the unwanted fluid portion is transferred into a waste container 901 for proper disposal. This creates a disposable closed system to effectively extract tissues and cells from a patient, process the tissues and wash the cells, followed by concentrating the cells in a suspension that can then be readily administered to the patient, or conserved for further diagnostic measures.
  • the entire process is performed in the closed system such that the chance of contamination is greatly reduced or minimized.
  • the disposable aspect makes it easy to use without having to clean and sterilize the system every time after use.
  • the processing chamber 101 has been pre-filled with processing fluid that contains the enzyme mixture discussed above.
  • tissue, enzymes and processing fluid are introduced inside the processing compartment 101 through the first inlet 601. Additionally, enzyme-free washing fluid is also introduced into the washing fluid compartment 200 through the second inlet 602.
  • the content of the washing fluid chamber 200 is initially filled to a level reflecting not more than the height of the upper limit of wall 105.
  • the amount of washing fluid can vary depending on the tissues and amount thereof being processed.
  • step 503 by repetitive application of rotational low g-force (1 st g-force), the fluid is moved within and around the processing compartment 101. Repetitive cycles of subsequent accelerations and decelerations agitates the to be processed tissue and fluid inside the heated or pre-heated processing compartment 101.
  • the elevated temperature facilitates the breaking down of the solid tissue and dissociation of cells from the tissue.
  • the sidewalls of processing unit 100 are not round but triangularly shaped as show in FIG. 6C, resulting in different distances from the sidewall to the middle axis of the driving axis of the centrifuge, the acceleration and deceleration at different sites inside the processing compartment 101 is not homogeneous, in contrast to a fully round shaped chamber.
  • the distance, for example, dl from the perimeter of the middle axis to one of the three vertices is greater than the distance d2 from the perimeter of the middle axis to the wall 105. Therefore, different acceleration forces act on the tissue that don’t allow a concentric vortex, but effect a better and more intense agitation. This in turns facilitates processing and action of the enzyme to better break down of the tissues.
  • step 505 also referring to FIG. 7B, with pulsatile acceleration at a 2 nd g-force, clean washing fluid from wash fluid compartment 200 is driven through opening 201 and the siphon structure 400 into the concentration container 500 to nearly fill up the concentration container. This is done before the processed tissue and cells enter the wash fluid compartment 200 and mix with the washing fluid.
  • the siphon structure 400 prevents washing fluid from spontaneously flowing back into the wash fluid chamber 200.
  • step 507 also referring to FIG. 7C, the processing unit is accelerated at a 3 rd g-force higher than the 1 st processing force but lower than the 2 nd pulsatile acceleration force, thereby driving processed tissue and cells through the opening 109 into the wash fluid compartment 200.
  • the initial dissociated tissue and the enzyme is diluted and washed by the washing fluid.
  • the concentration of enzymes can be diluted to a level that is clinically acceptable to use on a patient.
  • step 509 by subsequent applying a 4 th , higher centrifugal forces, the washed tissue and fluid are driven from the wash fluid compartment 200 toward the opening 201, where the filter mesh 300 filters out the large tissue fragment and debris, while the smaller and heavier cells pass through the mesh opening and the siphon structure 400, and eventually enter the concentration container 500. This way, cells can be removed from each of the three concentration containers 500 .
  • the amount of the washing fluid in both the washing fluid compartment 200 and the concentration container 500 is calculated such that the concentration of any remaining processing fluid (enzymes and buffers) can be reduced below the threshold value, and that the diluted cells can be readily injected into a subject if needed, or used for further diagnostic purposes, or conserved by cryofreezing.
  • any remaining processing fluid enzymes and buffers
  • the diluted cells can be readily injected into a subject if needed, or used for further diagnostic purposes, or conserved by cryofreezing.
  • the concentration of any remaining processing fluid enzymes and buffers
  • the diluted cells can be readily injected into a subject if needed, or used for further diagnostic purposes, or conserved by cryofreezing.
  • the centrifugation can be controlled to transfer 100 mL of the mixture into the washing chamber, and in step 509 the washed cells are further diluted by 10X, resulting in a final concentration of less than 4 unit of enzymes.
  • the centrifuge machine can be programmed to automate a consecutive multi stage centrifugation, and the inventive three-chamber configuration requires no or minimal human handling between each stage; with the lid ensuring sterile inner conditions.
  • Conventional method and system on the contrary, requires at least one, often repetitive human handlings between each stage, which significantly increase the complexity or chance of contamination.
  • the processing unit can be easily used with existing a programmable centrifuge to enhance processing of adipose tissue and extract stem cells with high efficiency and low contamination.
  • Prantl L Peripheral motor and sensory nerve conduction following transplantation of undifferentiated autologous adipose tissue-derived stem cells in a biodegradable u.s. food and drug administration- approved nerve conduit. PlastReconstr Surg 2016; 138(1): 132- 139.
  • Biedermann T Therapy of ulcus curis of venous and mixed venous arterial origin with autologous, adult, native progenitor cells from subcutaneous adipose tissue: a prospective clinical pilot study. J Eur Acad Dermatol Venerol 2017; 31(12): 2104-2118.
  • Adipose stem cells display higher regenerative capacities and more adaptable electro- kinetic properties compared to bone marrow- derived mesenchymal stromal cells. Sci Rep 2016; 6, 37801.
  • Van Dongen JA Tuin AJ, Spiekman M, Jansma J, van der Lei B, Harmsen MC. Comparison of intraoperative procedures for isolation of clinical grade stromal vascular fraction for regenerative purposes: a systematic review. J Tissue Eng Regen Med 2018; 12(1): e261-e274. [00122] Winnier G, Valenzuela N, Alt C, Alt EU. Isolation of adipose tissue derived regenerative cells from human subcutaneous tissue with or without the use of enzymatic reagent. PLoS One 2019; 14(9): e0221457.
  • Domenis R Lazzaro L, Calabrese S, Mangoni D, Gallelli A, Bourkoula E, Manini I, Bergamin N, Toffoletto B, Beltrami CA, Beltrami AP, Cesselli D, Parodi PC.
  • Adipose tissue derived stem cells in vitro and in vivo analysis of a standard and three commercially available cell-assisted lipotransfer techniques. Stem Cell Res Ther 2015; 6: 2.
  • Boetel T Safety of adipose-derived regenerative cells injection for treatment of osteoarthritis of the facet joint.
  • ClinicalTrials.gov Identifier: NCT03513731, IDE 17991. https://www.clinicaltrials.gov/ct2/show/NCT03513731 (zuêt am 14.10.2019 aufrick).
  • Vandermark R healing osteoarthritic joints in the wrist with adult ADRCs.
  • ClinicalTrials.gov Identifier: NCT03503305, IDE 17984. https://www.clinicaltrials.gov/ct2/show/NCT03503305 (zuêt am 14.10.2019 auftechnisch).
  • Reilly P Macleod I, Macfarlane R, Windley J, Emery RJ. Dead men and radiologists don't lie: a review of cadaveric and radiological studies of rotator cuff tear prevalence. Ann R Coll Surg Engl 2006; 88(2): 116-121.
  • Alt EU Milz S, Reiser MF, Alt C, Schmitz C. Formation of new tendon tissue after treating symptomatic, partial rotator cuff tears with freshly isolated, uncultured, unmodified, autologous adipose derived regenerative cells (UA-ADRCs) at point of care: A first-in-human case report. Manuscript submitted for publication.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Veterinary Medicine (AREA)
  • Anesthesiology (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Vascular Medicine (AREA)
  • Public Health (AREA)
  • Surgery (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Sustainable Development (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Abstract

L'invention concerne des procédés et un appareil pour la récupération de cellules à partir d'un échantillon de tissu utilisant un récipient unique pour le traitement et la séparation de tissus et de cellules. L'unité de traitement comprend un compartiment de traitement présentant une paroi latérale de traitement ; au moins un compartiment de fluide de lavage présentant une paroi latérale de lavage, le compartiment de fluide de lavage étant situé radialement à l'extérieur du compartiment de traitement et séparé par la paroi latérale de traitement, une première ouverture étant formée au sommet de la paroi latérale de traitement pour permettre une communication fluidique entre le compartiment de traitement et le compartiment de fluide de lavage ; et au moins un compartiment de concentration, le récipient de concentration étant situé radialement à l'extérieur du compartiment de fluide de lavage et séparé par la paroi latérale de lavage, une seconde ouverture étant formée au sommet de la paroi latérale de lavage pour permettre une communication fluidique entre le compartiment de fluide de lavage et le compartiment de concentration.
EP21735386.1A 2020-04-30 2021-04-30 Dispositif et procédé de liposuccion et de traitement de tissus intégrés automatisés Pending EP4143292A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063018475P 2020-04-30 2020-04-30
PCT/IB2021/000295 WO2021220057A2 (fr) 2020-04-30 2021-04-30 Dispositif et procédé de liposuccion et de traitement de tissus intégrés automatisés

Publications (1)

Publication Number Publication Date
EP4143292A2 true EP4143292A2 (fr) 2023-03-08

Family

ID=76641724

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21735386.1A Pending EP4143292A2 (fr) 2020-04-30 2021-04-30 Dispositif et procédé de liposuccion et de traitement de tissus intégrés automatisés

Country Status (2)

Country Link
EP (1) EP4143292A2 (fr)
WO (1) WO2021220057A2 (fr)

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2469370C (fr) * 2001-12-07 2014-07-08 Macropore Biosurgery, Inc. Unite de traitement de cellule d'origine adipeuse
JP5036026B2 (ja) * 2006-03-09 2012-09-26 旭化成株式会社 血液成分分離装置及びその使用方法
US11174458B2 (en) * 2007-04-23 2021-11-16 Koligo Therapeutics, Inc. Cell separation apparatus and methods of use
WO2009073724A1 (fr) 2007-12-04 2009-06-11 Ingeneron, Inc. Appareil et procédé pour l'isolation de cellules
US20130006225A1 (en) * 2009-08-05 2013-01-03 Rocin Laboratories, Inc. Twin-type cannula assemblies for hand-held power-assisted tissue aspiration instruments
CN103402646B (zh) 2010-12-16 2015-10-21 英吉诺隆公司 用于增强从组织样品回收细胞和富含细胞的基质的方法和装置
ES2570730T3 (es) * 2012-08-29 2016-05-20 Euromi S A Aparato para la extracción y reinyección de tejido adiposo
KR101466923B1 (ko) * 2013-01-17 2014-12-04 주식회사 휴림바이오셀 재생성 세포 추출장치, 재생성 세포 추출 시스템, 이를 이용한 재생성 세포 추출방법
US10125345B2 (en) * 2014-01-31 2018-11-13 Dsm Ip Assets, B.V. Adipose tissue centrifuge and method of use

Also Published As

Publication number Publication date
WO2021220057A3 (fr) 2021-12-30
WO2021220057A2 (fr) 2021-11-04

Similar Documents

Publication Publication Date Title
KR101127305B1 (ko) 조직으로부터 재생성 세포를 분리하고 농축시키기 위한시스템 및 방법
KR101400544B1 (ko) 심혈관 증상의 치료에 지방조직-유래의 세포를 사용하는 방법
US7514075B2 (en) Systems and methods for separating and concentrating adipose derived stem cells from tissue
US9463203B2 (en) Methods of using regenerative cells in the treatment of cartilage defects
JP5960689B2 (ja) 脂肪吸引により抽出された小葉脂肪から移植用の組織、特に脂肪組織、を準備するための装置及び方法
KR101310578B1 (ko) 심혈관 증상의 치료에 지방조직-유래 세포를 사용하는 방법
US8883499B2 (en) Systems and methods for isolating and using clinically safe adipose derived regenerative cells
KR101951055B1 (ko) 조직 검체로부터 세포 및 세포가 풍부한 기질의 강화된 회수를 위한 방법 및 기구
JP4937119B2 (ja) 筋骨格疾患の治療に再生細胞を使用する方法
US20130034524A1 (en) Non-Enzymatic Method for Harvesting Adipose-Derived Stromal Cells and Adipose-Derived Stem Cells from Fat and Lipo-Aspirate
US20150030571A1 (en) Preparation and method for producing a preparation comprising mesenchymal stem cells
MXPA06003088A (es) Metodos para usar celulas regenerativas en el tratamiento de enfermedades vasculares perifericas y trastornos relacionados.
KR20120020143A (ko) 조직 및 세포 부유화 이식편의 최적화 시스템, 방법 및 조성물
CN113583951A (zh) 通过超声空化从脂肪组织分离干细胞及使用方法
KR20130041992A (ko) 자기혈청 첨가 골수세포 배양 시스템, 자기혈청 첨가 골수세포 배양방법 및 자기혈청 첨가 배양 골수세포를 유효성분으로 하는 의약조성물의 제조방법
KR101044371B1 (ko) 줄기세포 분리를 위한 혼합장치
EP4143292A2 (fr) Dispositif et procédé de liposuccion et de traitement de tissus intégrés automatisés
WO2012122603A1 (fr) Méthode et dispositif de traitement cellulaire
EP2268293A2 (fr) Tolérance à une transplantation liée à l'emploi d'une nouvelle composition de cellules souches
KR20070017974A (ko) 말초 혈관 질환 및 관련 장애의 치료에서의 재생 세포의이용 방법

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221124

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)