US20200032197A1 - Aseptic tissue processing method, kit and device - Google Patents

Aseptic tissue processing method, kit and device Download PDF

Info

Publication number
US20200032197A1
US20200032197A1 US16/477,366 US201816477366A US2020032197A1 US 20200032197 A1 US20200032197 A1 US 20200032197A1 US 201816477366 A US201816477366 A US 201816477366A US 2020032197 A1 US2020032197 A1 US 2020032197A1
Authority
US
United States
Prior art keywords
module
disaggregation
tissue
stabilisation
kit
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/477,366
Inventor
Ryan Dominic Guest
Nicola Kaye Price
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.)
Instil Bio UK Ltd
Original Assignee
Immetacyte Ltd
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 Immetacyte Ltd filed Critical Immetacyte Ltd
Assigned to Immetacyte Limited reassignment Immetacyte Limited ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GUEST, Ryan Dominic, PRICE, Nicola Kaye
Publication of US20200032197A1 publication Critical patent/US20200032197A1/en
Assigned to INSTIL BIO (UK) LIMITED reassignment INSTIL BIO (UK) LIMITED CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: Immetacyte Limited
Abandoned legal-status Critical Current

Links

Images

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
    • C12M47/00Means for after-treatment of the produced biomass or of the fermentation or metabolic products, e.g. storage of biomass
    • C12M47/04Cell isolation or sorting
    • 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
    • C12M41/00Means for regulation, monitoring, measurement or control, e.g. flow regulation
    • C12M41/48Automatic or computerized control
    • 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
    • C12M45/02Means for pre-treatment of biological substances by mechanical forces; Stirring; Trituration; Comminuting
    • 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
    • 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
    • C12M45/22Means for packing or storing viable microorganisms

Definitions

  • the present invention concerns a kit and a semi-automatic device using that kit for aseptic disaggregation of solid tissue derived eukaryotic cells into either single cells or small cell number aggregates.
  • the invention further relates to a semi-automatic aseptic tissue processing method comprising: a process for aseptic disaggregation of solid tissue derived eukaryotic cells into either single cells or small cell number aggregates and their further processing.
  • any sorting technology can be used. This includes for example affinity chromatography or any other antibody-dependent separation technique known in the art. Any ligand-dependent separation technique known in the art may be used in conjunction with both positive and negative separation techniques that rely on the physical properties of the cells.
  • An especially potent sorting technology is magnetic cell sorting. Methods to separate cells magnetically are commercially available e.g. from Thermo fisher, Miltenyi Biotech, Stem cell Technologies, Cellpro, Seattle, Advanced Magnetics, Boston or Quad Technologies Boston. For example, monoclonal antibodies can be directly coupled to magnetic polystyrene particles like Dynal M 450 or similar magnetic particles and used e.g. for cell separation.
  • nanobeads or MicroBeads can be either directly conjugated to monoclonal antibodies or used in combination with anti-immunoglobulin, avidin or antihapten-specific MicroBeads or coated with other mammalian molecules with selective binding properties.
  • Magnetic particle selection technologies such as those described above, allows cells to be positively or negatively separated by incubating them with magnetic nanoparticles coated with antibodies or other moieties directed against a particular surface marker. This causes the cells expressing this marker to attach to the magnetic nanoparticles. Afterwards the cell solution is placed within a solid or flexible container in a strong magnetic field. In this step, the cells attach to the nanoparticles (expressing the marker) and stay on the column, while other cells (not expressing the marker) flow through. With this method, the cells can be separated positively or negatively with respect to the particular marker(s).
  • the cells expressing the marker(s) of interest, which attached to the magnetic column are washed out to a separate vessel, after removing the column from the magnetic field.
  • the antibody or selective moiety used is directed against surface markers(s) which are known to be present on cells that are not of interest. After application of the cells/magnetic nanoparticles solution onto the column the cells expressing these antigens bind to the column and the fraction that goes through is collected, as it contains the cells of interest. As these cells are non-labelled by the selective antibodies or moiety(s) coupled to nanoparticels, they are “untouched”.
  • the processing requires strict regulated environmental conditions during handling the cell cultures, for example tissue processing as apart of or prior to disaggregation; enzymatic digestion and transfer into storing devices or incubation conditions for disaggregation/cellularisation and viable tissue yields.
  • tissue processing as apart of or prior to disaggregation; enzymatic digestion and transfer into storing devices or incubation conditions for disaggregation/cellularisation and viable tissue yields.
  • this process would require multiple pieces of laboratory and tissue processing equipment, and personal with the skills and knowledge of the scientific art with critical stages contained within either hazard containment or tissue processing facility(s) aseptic environment(s) in order to perform the same activity safely and also minimise the risk of contamination(s).
  • the present invention concerns a single use aseptic kit comprising a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate; and a stabilisation module for optionally further processing and/or storing disaggregated product material, wherein each of said modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of said modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers.
  • the kit described here allows for closed solid tissue processing eliminating the risk of contamination of the final cellularised product compared to standard non-closed tissue processing. This is especially when the process is performed within a tissue retrieval/procurement site and requires storage prior to final cell processing for its ultimate utility. In addition, safety of the operator is increased due to reduction of direct contact with biological hazardous material which may contain infectious organisms such as viruses.
  • the kit also enables either all of or a portion of the finally processed cellularised material to be stabilised for either transport or storage prior to being processed for its ultimate utility.
  • the one or more flexible containers comprise a resilient deformable material.
  • the one or more flexible containers of the disaggregation module may comprise one or more sealable openings.
  • the one or more flexible containers of the disaggregation module and/or the stabilisation module may also comprise a heat sealable weld.
  • the one or more flexible containers that are part of any module comprise internally rounded edges.
  • the one or more flexible containers of the disaggregation module may comprise disaggregation surfaces adapted to mechanically crush and shear the solid tissue therein.
  • the one or more flexible containers of the enrichment module may comprise a filter adapted to retain a retentate of cellularised disaggregated solid tissue.
  • the kit further comprises a radio frequency or other digitally recognisable identification tag so that it may be scanned and recognised during automated processing, such as with/in the automated device in embodiments of the present invention.
  • the tag provides information about the conditions and steps required to be auto processed, so simply by scanning the kit, any automated system used with the kit to process the tissue can be undertaken without further intervention or contamination.
  • the invention is particularly useful in a sample processing, particularly automated processing.
  • the invention concerns use of the single use aseptic kit described above in a semi-automated process for the aseptic disaggregation and/or enrichment and stabilisation of mammalian cells or cell aggregates.
  • a particular advantage is that solid tissue disaggregation (and optional processes including all described manipulations herein described required to achieve optimal results) can be performed in a closed system, i.e. an aseptic process with minimal risk of contaminations and with minimal user knowledge.
  • the device may have a comprising radio frequency identification tag reader to recognise the single use kit.
  • the programmable processor is capable of recognising the single use aseptic kit via its tag and subsequently able to execute the kit programme which defines the type of disaggregation, enrichment and stabilisation processes together with the respective media types required for those processes.
  • the programmable processor is adapted to communicate with and control one or more of: the disaggregation module; the enrichment module; and the stabilisation module of the device.
  • the device including its processor, may therefore have multiple functionality to assess the flow of materials through the kit making decisions of when a step is completed as part of the pre-programmed functions and the feedback the device gets from its sensors.
  • the programmable processor may control the disaggregation module to enable a physical and/or biological breakdown of the solid tissue material in that container.
  • the programmable processor may also control the disaggregation module to enable a physical and enzymatic breakdown of the solid tissue material.
  • the enzymatic breakdown of the solid tissue material is by the selection and provision of one or more media enzyme solutions selected from collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase H1, pepsin, or mixtures thereof.
  • the programmable processor may control disaggregation enabling the surfaces within the disaggregation flexible containers to mechanically crush and shear the solid tissue.
  • the disaggregation surfaces are controlled by mechanical pistons, for example.
  • the programmable processor controls the stabilisation module to cryopreserve the enriched disaggregated solid tissue in the container, for example, this may be achieved by using a programmable temperature setting, a condition which is determined by reading the tag of the kit inserted in the device.
  • one or more of the additional components of the device and/or kit are provided. Such features may be available in any combination. This may include for example: sensors in the device capable of recognising whether a disaggregation process has been completed in the disaggregation module of the kit prior to transfer of the disaggregated solid tissue to the optional enrichment module; weight sensors to determine an amount of media required in the containers of one or more of the disaggregation module; the enrichment module; and/or the stabilisation module and means to control that transfer of material between respective containers; and temperature sensors to control the temperature within the containers of the one or more of the disaggregation module; the enrichment module; and/or the stabilisation module.
  • an optional bubble sensor to control the transfer of media between the input and output ports of each container in the module
  • one or more pumps may provide means to control the transfer of media between the input and output ports
  • pressure sensors to assess the pressure within the enrichment module
  • valves to control an optional tangential flow filtration process within the enrichment module
  • one or more clamps to control the transfer of media between the input and output ports of each module.
  • the programmable processor is adapted to maintain an optimal storage temperature range in the stabilisation module until the container is removed; or executes a controlled rate of freezing.
  • inventions of the device and kit allow solid tissue derived cells or cell aggregates to be: stored for short periods (minutes to days) or stored for long periods (multiple days to years) prior to their ultimate utility depending on the type or stabilisation process used with the stabilisation module.
  • the device of the invention may further comprise a user interface.
  • That interface may comprise a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof.
  • the automated device is adapted to be transportable and thus may comprise dimensions that permit easy manoeuvrability and/or aid movement such as wheels, tyres, handles and the like.
  • the final cellular material product can then be used for but not limited to either: regenerative medicine, adoptive cell therapies, ATMPs, diagnostics or to further the basic scientific understanding of tissue, cell function or organism function.
  • the cells produced using the kit and/or device of the invention are useful for providing functional living cells and maybe cultured further for that use.
  • Cell culture is a process by which cells are grown outside the original host using controlled environmental and supportive conditions which vary by cell type and organism. These are often sterile artificial vessels which allow gas and temperature to be maintained and either manual or automated changes in essential nutrients, metabolites, growth factors and gases which enable regulation of the cells requirements to survive and in most cases thrive.
  • Cell culture requirements differ broadly by the type of cell(s) and its required purpose.
  • Cell culture conditions can be optimised for cell expansion, cell differentiation or manufacturing of different phenotypes of the cell or its products. The most commonly varied factor in culture systems is the cell culture medium, for which a vast number of recipes is known (see for example “Cell Culture Techniques” Humana Press, 1st. Edition, 2011)
  • disaggregated or cellularised material produced by the device and kit can be useful as the starting material to isolate specific cell populations which are grown out using stimulation or non-required cells are inhibited or apoptosis/cell death is induced resulting in a semi/purified population.
  • Such cells can be further sorted by one or more of the following processes: Fluorescence-activated cell sorting using antibody/protein labelling or natural fluorescence; Magnetic separation of cells, e.g. the magnetic activated cell sorting (MACS technology, Miltenyi Biotec GmbH, Germany).
  • MCS technology Magnetic activated cell sorting
  • This technology requires a marker that allows direct separation of the cells of interest by an antibody coupled to a magnetic microbead (Miltenyi et al., Cytometry 1990; 11:231-238).
  • a process of negative isolation can be employed.
  • non-target cells are magnetically labelled and depleted, thereby leaving the unlabelled cells of interest;
  • Label free cell separation and sorting using physical separation methods where either the target is not known, is a mixed population and physical cell (or clumps of cells) characteristics can be used to separate the cellular material from the current media to: remove impurities or reagents that are no longer required such as enzymes, cell debris, connective tissue, fat & mineral deposits; or exchange fluids which may be better for stabilising the cells for distribution and/or storage. It is envisaged that embodiments of the invention may include such functionality within the parameters of the processor or the automated device and operating system.
  • the purity, of the disaggregated and cellularised solid tissue product can be further increased if one or more cell surface marker(s) are used to select for or deplete a subpopulation of cells either as an independent step within the process or after processing using the methods described.
  • the present invention also relates to a method for enhanced semi-automated disaggregation cellularisation and storage of tissue derived cells.
  • steps of enrichment, formulation and cryopreservation are also provided.
  • a semi-automatic aseptic tissue processing method comprising: automatically determining aseptic disaggregation tissue processing steps and one or more further tissue processing steps and their associated conditions from a digital tag identifier on an aseptic processing kit, optionally in accordance with the kit described herein; placing a tissue sample into a flexible plastic container of the aseptic processing kit; and processing the tissue sample by automatically executing the one or more tissue processing steps by communicating with and controlling the disaggregation module; the optional enrichment module; and the stabilisation module.
  • the one or more automatically executed processes may be selected from one or more of:
  • the media maybe transferred into the disaggregation chamber, or in one embodiment also enters and collects enzymes prior to disaggregation using one or more embodiments of the invention, e.g. a mechanism such as weight sensors which will assess the required amount of media to add either determined by: direct operator input or weight of solid tissue.
  • a mechanism such as weight sensors which will assess the required amount of media to add either determined by: direct operator input or weight of solid tissue. Incubating with the media at an optimal temperature of between 30 & 37° C. but could be as low as 0° C. upto 40° C. for at least 1 minute to several hours but more preferable 15 to 45 minutes.
  • the disaggregation is designed to compress the tissues using a variable speed and time depending upon the time taken to disaggregate and feedback via sensors within the disaggregation module.
  • Steps 1) or 2) may be repeated until the tissue stops changing or has been disaggregated into a liquid cell suspension (whichever comes 1st monitored by a sensor in the disaggregation module).
  • Re-suspending the disaggregated cell product in fresh or additional media This could be a cell enrichment media in order to undergo an independent targeted enrichment procedure or direct cell culture or cold storage media (such as HypoThermosol® from BioLife Solutions).
  • cryoprotectant a freezing solution for storage of the disaggregated solid tissue derived product for days to years (such as CryoStor® Freezing solution from BioLife Solutions) and transferring to one or more flexible stabilising module having a cryopreservation container(s)
  • the disaggregated module and the storage module may comprise one and the same flexible container, for receiving the sample and storing the sample and a further flexible container for housing the media for disaggregation.
  • the same flexible containers are part of different modules of the kit.
  • “depletion” as used herein refers to a process of a negative selection that separates the desired cells from the undesired cells which are labelled by one marker-binding fragment coupled to a solid phase.
  • disaggregation or disaggregate refers to the transformation of solid tissue into a single cells or small cell number aggregates where a single cell as a spheroid has a diameter in the range of 5 ⁇ m, 6 ⁇ m, 7 ⁇ m, 8 ⁇ m, 9 ⁇ m, 10 ⁇ m, 20 ⁇ m, 30 ⁇ m, 40 ⁇ m, 50 ⁇ m, 60 ⁇ m, 70 ⁇ m, 80 ⁇ m, 90 ⁇ m, 100 ⁇ m or more where this is more usually between 7 to 20 ⁇ m.
  • cellularised or cellularisation refers to the process of disaggregation where by the solid tissue a multicellular material generally made up of multiple cell lineages/types is broken down into small numbers of cells including but not limited to one cell but could be multiple cells of various lineages or cell types in very small numbers i.e. clump of cells or cell aggregates.
  • engineered refers to either addition of nucleic material or factors which change the tissue derived cell function from their original function to have a new or improved function for its ultimate utility.
  • filtrate refers to the material that passes through a filter, mesh or membrane.
  • “flexible container” as used herein refers to a flexible packaging system in multiple formats with one or more different types of film. Each film type is selected to provide specific characteristics to preserve the physical, chemical, and functional characteristics of the sterile fluids, solid tissue derived cellular material and the container integrity depending upon the step of the process.
  • cryoprotectant is a solution that contains cryoprotective additives. These are generally permeable, non-toxic compounds which modify the physical stresses cells are exposed to during freezing in order to minimise freeze damage (i.e. due to ice formation). Most commonly a % Vol/Vol of one or more of the following: Dimethylsulphoxide (DMSO); Ethylene glycol; Glycerol; 2-Methyl-2,4-pentanediol (MPD); Propylene glycol; Sucrose; & Treha lose.
  • DMSO Dimethylsulphoxide
  • Ethylene glycol Ethylene glycol
  • Glycerol 2-Methyl-2,4-pentanediol
  • MPD 2-Methyl-2,4-pentanediol
  • Propylene glycol Sucrose; & Treha lose.
  • “media” means various solutions known in the art of cell culturing, cell handling and stabilisation used to reduce cell death, including but not limited to one or more of the following media Organ Preservation Solutions, selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove's medium, X-VIVOTM, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTETM solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's Solution.
  • the media can be standard cell media like the above mentioned media or special media for e.g. primary human cell culture (e.g.
  • the media may have supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, antibiotics, attachments factors, growth factors and cytokines, hormones, metabolic inhibitors or solubilising agents.
  • supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, antibiotics, attachments factors, growth factors and cytokines, hormones, metabolic inhibitors or solubilising agents.
  • Various media are commercially available e. g. from ThermoFisher Scientific or Sigma-Aldrich.
  • non-labelled or untouched refers to the cells which are not bound by one marker-binding fragment coupled to a solid phase.
  • the non-labelled, untouched cell fraction contains the desired target cells.
  • non-target cells refers to cells which are specifically bound by one marker-binding fragment which is coupled to a solid phase that is used to remove an unwanted cell type.
  • “positively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are the required population of cells.
  • purity refers to the percentage of the target population or populations desired from the original solid tissue.
  • regenerative medicine(s) “adoptive cell therapy(ies)” or “advanced therapy medicinal product(s)” are used interchangeably herein to refer to cellular material that is used for therapeutic purposes of one or more mammals either by: the action of a part of or all of the cellular material; the supportive actions of a part of or all of the cellular material with the aim to improve the wellbeing of the mammal after application.
  • the therapeutic cells can either be used directly or may require further processing, expansion and/or engineering to provide these actions.
  • sample refers to a sample containing cells in any ratio. Preferentially, these cells are viable. But, these cells can also be fixed or frozen cells which may be used for subsequent nucleic acids or protein extraction.
  • the samples may be from animals, especially mammals such as mouse, rats or humans. Any compressible solid tissue that contains cells can be used.
  • the invention is illustrated mainly through the isolation of hematopoietic and cancer cells from solid tumour tissue. However, the invention relates to a method for isolation of a breadth of cells from any mammalian solid tissue.
  • marker refers to a cell antigen that is specifically expressed by a certain cell type. Preferentially, the marker is a cell surface marker, so that enrichment, isolation and/or detection of living cells can be performed.
  • solid phase refers to the coupling of the marker-binding fragment, e.g. an antibody, bound to another substrate(s), e.g. particles, fluorophores, haptens like biotin, polymers, or larger surfaces such as culture dishes and microtiterplates.
  • the coupling results in direct immobilization of the antigen-binding fragment, e.g. if the antigen-binding fragment is coupled to a larger surface of a culture dish.
  • this coupling results in indirect immobilisation, e.g. an antigen-binding fragment coupled directly or indirectly (via e.g. biotin) to a magnetic bead is immobilised if said bead is retained in a magnetic field.
  • the coupling of the antigen-binding fragment to other molecules results not in a direct or indirect immobilization but allows for enrichment, separation, isolation, and detection of cells according to the present invention, e.g. if the marker-binding fragment is coupled to a chemical or physical moiety which then allows discrimination of labelled cells and non-labelled cells, e.g. via flow cytometry methods, like FACSsorting, or fluorescence microscopy.
  • solid tissue refers to a piece or pieces of animal derived mammalian solid tissue which by its three dimensions i.e. length, breadth and thickness as a geometrical body is larger than the size of multiple individual cell based units and often contains connective materials such as collagen or a similar matrix that make up structure of the tissue whereby said solid tissue cannot flow through tubes or be collected by a syringe or similar small conduit or receptacle and is i.e. with dimensions in the range of 500 ⁇ m, 1 mm, 2 mm, 3 mm, 4 mm, 5 mm, 1 cm, 2 cm, 3 cm, 4 cm, 5 cm, 10 cm, 20 cm, 30 cm or more
  • particle refers to a solid phase such as colloidal particles, microspheres, nanoparticles, or beads. Methods for generation of such particles are well known in the field of the art.
  • the particles may be magnetic particles or have other selective properties.
  • the particles may be in a solution or suspension or they may be in a lyophilised state prior to use in the present invention. The lyophilized particle is then reconstituted in convenient buffer before contacting the sample to be processed regarding the present invention.
  • Magnetic in “magnetic particle” as used herein refers to all subtypes of magnetic particles which can be prepared with methods well known to the skilled person in the art, especially ferromagnetic particles, superparamagnetic particles and paramagnetic particles.
  • “Ferromagnetic” materials are strongly susceptible to magnetic fields and are capable of retaining magnetic properties when the field is removed.
  • Paramagnetic materials have only a weak magnetic susceptibility and when the field is removed quickly lose their weak magnetism.
  • Superparamagnetic” materials are highly magnetically susceptible, i.e. they become strongly magnetic when placed in a magnetic field, but, like paramagnetic materials, rapidly lose their magnetism.
  • marker-binding fragment refers to any moiety that binds preferentially to the desired target molecule of the cell, i.e. the antigen.
  • the term moiety comprises, e.g., an antibody or antibody fragment.
  • antibody refers to polyclonal or monoclonal antibodies which can be generated by methods well known to the person skilled in the art.
  • the antibody may be of any species, e.g. murine, rat, sheep, human.
  • non-human antigen binding fragments are to be used, these can be humanized by any method known in the art.
  • the antibodies may also be modified antibodies (e.g. oligomers, reduced, oxidized and labelled antibodies).
  • antibody comprises both intact molecules and antibody fragments, such as Fab, Fab′, F(ab′)2, Fv and single-chain antibodies.
  • marker-binding fragment includes any moiety other than antibodies or antibody fragments that binds preferentially to the desired target molecule of the cell. Suitable moieties include, without limitation, oligonucleotides known as aptamers that bind to desired target molecules (Hermann and Pantel, 2000: Science 289: 820-825), carbohydrates, lectins or any other antigen binding protein (e.g. receptor-ligand interaction).
  • retentate refers to the material that does not pass through a filter, mesh or membrane.
  • “ultimate utility” as used herein refers to manufacture of or direct use in regenerative medicines, adoptive cell therapies, ATMPs, diagnostic in vitro studies or scientific research.
  • FIG. 3 a provides yet a further example of the invention in which:
  • FIG. 3 b there is provided:
  • FIG. 4 shows a further example of the device and kit of the invention in which:
  • a semi-automatic aseptic tissue processing method comprising: automatically determining aseptic disaggregation tissue processing steps and one or more further tissue processing steps and their associated conditions from a digital tag identifier on an aseptic processing kit, optionally in accordance with the kit described herein; placing a tissue sample into a flexible plastic container of the aseptic processing kit; and processing the tissue sample by automatically executing the one or more tissue processing steps by communicating with and controlling the disaggregation module; the optional enrichment module; and the stabilisation module.
  • the process may comprise taking an open ended bag (1st flexible container that is part of disaggregation module) that will receive the biopsy/tissue sample which is already connected via one or more conduits to (conduit) or can be connected via a manual operator controlled aseptic connection to
  • the digestion media can be added via the conduit or aseptic connections (conduit/ports claim 1 ) and the tissue material processed.
  • the cells can optionally be filtered prior to step 4 (optional enrichment module for filtration comprises the 1st flexible container containing sample and filtered to a 3rd container for receiving the enriched filtrate)
  • the single or small number aggregate cellular suspension in the original flexible container or which may be optionally subdivided into multiple storage stabilisation containers thereafter are maintained in a stable state on the device and/or will undergo cryopreservation prior to removal for, transport, storage and or used in their ultimately utility.
  • the stabilisation module also comprises 1st or 3rd container as used in storage/freezing/storage
  • FIGS. 6 and 7 describe further examples in which the disposable kit of the invention can be used with an automatic device for semi-automatic aseptic processing of tissue samples.
  • Process step 1 The user may login to device and scan the tag on the aseptic kit using the device to transfer the automatic processing steps to be used.
  • the device processor recognises the tag and is provided with information needed to carry out the specific processing instructions related to that particular kit.
  • Process step 2 The digestion media containing flexible bag (part of disaggregation module) and cryo/stabilisation solution containing flexible bag (part of the stabilisation module) are each hung or secured to the device.
  • Process step 3 The biopsy or tissue sample for processing may be placed into a flexible container (part of both modules) of the aseptic kit via an open end.
  • Process step 5 The user may then interact with the user interface of the processor to confirm the tissue sample is present and enter any further tissue material specific information, if required.
  • Process step 6 Digestion media and cryo/stabilisation solution flexible containers are connected with the flexible container housing the sample, after which it maybe placed into the device for automatic processing.
  • Process step 7 The device executes the cycles according to the kit information undertaking disaggregation of the sample and stabilisation/cryo preservation of resulting cells.
  • Process step 8 When stabilised/frozen disconnect and discard used media and cryo/stabilisation containers of kit. Tissue processed into single or multi-cell solution in flexible container is disconnected before transferring into storage or transport container prior to its ultimate utilisation.
  • FIG. 7 describes how flexible containers comprising the media used in the process may be shared between the modules of the aseptic processing kit and method.
  • Process step 1 The user may login to device and scan the tag on the aseptic kit using the device to transfer the automatic processing steps to be used.
  • Process step 2 A flexible bag (part of disaggregation/stabilisation module) comprising both the media and cryo/stabilisation solution is hung or otherwise secured to the device.
  • Process step 4 The flexible container comprising the sample may then be sealed using a heat weld to close the open end.
  • Process step 5 The user may then interact with the user interface of the processor to confirm the tissue sample is present and enter any tissue material specific information, if required.
  • Process step 6 Digestion media and cryo/stabilisation solution flexible container is connected with the flexible container housing the sample, after which it maybe placed into the device for automatic processing.
  • Process step 7 The device cycles to enable disaggregation of the sample and stabilisation of resulting cells, optionally via cryopreservation.
  • Process step 8 When freezing/stabilising is complete the user disconnects and discard used flexible containers of kit. Tissue processed into single or multi-cell solution in the remaining flexible container is disconnected before transferring into storage or transport container prior to its ultimate utilisation.
  • the media formulation for enzymatic digestion must be supplemented with enzymes that aid in protein breakdown causing the cell to cell boundaries to breakdown as described above.
  • liquid formulations known in the art of cell culturing or cell handling can be used as the liquid formulation used for cell disaggregation and enzymatic digestion of solid tissues, including but not limited to one or more of the following media Organ Preservation Solutions, selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove's medium, X-VIVOTM, AIM-VTM, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTETM solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's SolutionDMEM, HBSS, DPBS, RPMI, AIM-VTM, Iscove's medium, X-VIVOTM, each can be optionally supplemented with additional cell supporting factors e.g.
  • the media can be standard cell media like the above mentioned media or special media for e.g. primary human cell culture (e.g. for endothelia cells, hepatocytes or keratinocytes) or stem cells (e.g. dendritic cell maturation, hematopoietic expansion, keratonocytes, mesenchymal stem cells or T cells).
  • the media may have supplements or reagents well known in the art, e.g.
  • albumins and transport proteins amino acids and vitamins, metal-ion(s), antibiotics, attachments factors, de-attachment factors, surfactants, growth factors and cytokines, hormones or solubilising agents.
  • Various media are commercially available e. g. from ThermoFisher, Lonza or Sigma-Aldrich or similar media manufacturers and suppliers.
  • the solid tissue to be digested can be washed after disaggregation with a liquid formulation containing chelating agents EGTA and EDTA to remove adhesion factors and inhibitory proteins prior to washing and removal of EDTA and EGTA prior to enzymatic digestion.
  • liquid formulation required for enzymatic digestion is more optimal with minimal chelating agents EGTA and EDTA which can severely inhibit enzyme activity by removing calcium ions required for enzyme stability and activity.
  • EGTA and EDTA minimal chelating agents
  • ⁇ -mercaptoethanol, cysteine and 8-hydroxyquinoline-5-sulfonate are other known inhibitory substances.
  • final cell container for cryopreservation is a flexible container manufactured from resilient deformable material.
  • the final container is either transferred directly to a freezer ⁇ 20 to ⁇ 190° C. or more optimally located in the controlled rate freezing apparatus either associated with the device or supplied separately (manufactured by for example Planer Products or Asymptote Ltd) in which the temperature of the freezing chamber and the flexible storage container(s) employed to contain the enriched disaggregated solid tissue container is controlled either by: injecting a cold gas (normally nitrogen for example Planer products); or by removing heat away from the controlled cooling surface(s). Both methods result in the ability to accurately control with an error of less than 1° C. or more preferable 0.1° C.
  • the freezing process at the required rate for the specific cell(s) to be frozen based on the freezing solution and the desired viability of the product.
  • This cryopreservation process must take into account the ice nucleation temperature which is ideally as close as possible to the melting temperature of the freezing solution.
  • aqueous solutions water is removed from the system as ice, and the concentration of the residual unfrozen solution increases. As the temperature is lowered, more ice forms, decreasing the residual non-frozen fraction which further increases in concentration.
  • aqueous solutions there exists a large temperature range in which ice co-exists with a concentrated aqueous solution. Eventually through temperature reduction the solution reaches the glass transition state at which point the freezing solution and cells move from a viscous solution to a solid-like state below this temperature the cells can undergo no further biological changes and hence are stabilised, for years potentially decades, until required.
  • the disaggregated cell products achieved by the method of the present invention can be cultured and/or analysed (characterised) according to all methods known to the person skilled in the art.
  • the cells obtainable by the methods disclosed herein may be used for subsequent steps such as research, diagnostics, tissue-banks, biobanks, pharmacological or clinical applications known to the person skilled in the art.
  • Cells can then be taken into culture using a Medium optimized for this application, e.g. T cell Mixed Media (Cellular Therapeutics) usually containing but not limited to growth factors such as IL-2, IL-7, IL-15, IL-21 or stimulatory conditions such as plates or polystyrene beads coated with antibodies.
  • a Medium optimized for this application e.g. T cell Mixed Media (Cellular Therapeutics) usually containing but not limited to growth factors such as IL-2, IL-7, IL-15, IL-21 or stimulatory conditions such as plates or polystyrene beads coated with antibodies.
  • T cell Mixed Media Cellular Therapeutics
  • isolated cells were seeded into culture containers and maintained using procedures standardly used by a person skilled in the art such as a humidified atmosphere (1-20% usually 5% CO2, 80 to 99% usually
  • Such cell cultures can be used to study e.g. cell function, tumour cell killing, cell signalling, biomarkers, cell pathways, nucleic acids, and other cell or tissue related factors that may be used to identify donor, tissue, cell or nucleic acid status.
  • the enriched cells could be used before and/or after cell culturing as a pharmaceutical composition in the therapy, e.g. cellular therapy, or prevention of diseases.
  • the pharmaceutical composition can be used for the treatment and/or prevention of diseases in mammals, especially humans, possibly including administration of a pharmaceutically effective amount of the pharmaceutical composition to the mammal.
  • the disease may be any disease, which can be treated and/or prevented through the presence of solid tissue derived cells and/or through increasing the concentration of the relevant cells in/at the relevant place, i.e. the tumours or sites of disease.
  • the treated and/or preventively treated disease may be any disorder, e.g. cancer or a degenerative disorder.
  • the treatment may be the transplantation of enriched, engineered or expanded cells or any combination of these and either administered to the relevant part of the body or supplied systemically.
  • compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated (or prevented).
  • the quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
  • Peripheral blood mononuclear cells were physically disaggregated for 0, 1, 5 & 10 minutes continuously before a being cultured in vitro for 0, 24 & 96 hours to assess cell recovery.
  • the results demonstrate the physical process has negligible impact over 1 or 5 minutes and at 10 minutes the impact was transient where and initial reduction in viable cells at 0 hours was equivalent to non-disaggregated cells at 24 & 96 hours ( FIG. 5 ).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Sustainable Development (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Computer Hardware Design (AREA)
  • Mechanical Engineering (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention concerns a single use aseptic kit comprising: a disaggregation module for receipt and processing of material comprising solid mammalian tissue; and a stabilisation module for storing disaggregated product material, wherein each of the modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of the modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers. The invention further relates to an automated device for semi-automated aseptic disaggregation and/or enrichment and/or stabilisation of cells or cell aggregates from mammalian solid tissue comprising a programmable processor and the single use aseptic kit. The invention further relates to a semi-automatic aseptic tissue processing method.

Description

  • The present invention concerns a kit and a semi-automatic device using that kit for aseptic disaggregation of solid tissue derived eukaryotic cells into either single cells or small cell number aggregates. The invention further relates to a semi-automatic aseptic tissue processing method comprising: a process for aseptic disaggregation of solid tissue derived eukaryotic cells into either single cells or small cell number aggregates and their further processing.
  • BACKGROUND
  • The conditions during solid tissue disaggregation and time taken to harvest the cells have a substantial impact on the viability and recovery of the final cellularised material. Typically a solid tissue derived cell suspension, that is obtained, comprises a wide variety of different cell types and the disaggregation media and tissue debris or fluids. Often, selective targeting and or isolation of an individual or multiple cell types is prerequisite for the starting material prior to manufacture of regenerative medicines, adoptive cell therapies, ATMPs, diagnostic in vitro studies or scientific research. Generally these selection or enrichment techniques rely on one of the following properties: size, shape, density, adherence or strong protein: protein interactions (i.e. antibody: antigen) or providing a growth supporting environment by controlling the culture conditions or more complex cell marker interactions associated with semi-permanent or permanent coupling to magnetic or non-magnetic solid or semi-solid phase substrates can be used.
  • For enrichment, isolation or selection in principle any sorting technology can be used. This includes for example affinity chromatography or any other antibody-dependent separation technique known in the art. Any ligand-dependent separation technique known in the art may be used in conjunction with both positive and negative separation techniques that rely on the physical properties of the cells. An especially potent sorting technology is magnetic cell sorting. Methods to separate cells magnetically are commercially available e.g. from Thermo fisher, Miltenyi Biotech, Stem cell Technologies, Cellpro, Seattle, Advanced Magnetics, Boston or Quad Technologies Boston. For example, monoclonal antibodies can be directly coupled to magnetic polystyrene particles like Dynal M 450 or similar magnetic particles and used e.g. for cell separation. The Dynabeads technology is not column based, instead these magnetic beads with attached cells enjoy liquid phase kinetics in a sample tube, and the cells are isolated by placing the tube on a magnetic rack. However, in a preferred embodiment for enriching, sorting and/or detecting neuronal cells from a sample containing neuronal cells according the present invention monoclonal antibodies are used in conjunction with colloidal superparamagnetic microparticles having an organic coating by e.g. polysaccharides (Magnetic-activated cell sorting (MACS) technology (Miltenyi Biotec, Bergisch Gladbach, Germany)). These particles (nanobeads or MicroBeads) can be either directly conjugated to monoclonal antibodies or used in combination with anti-immunoglobulin, avidin or antihapten-specific MicroBeads or coated with other mammalian molecules with selective binding properties.
  • Magnetic particle selection technologies such as those described above, allows cells to be positively or negatively separated by incubating them with magnetic nanoparticles coated with antibodies or other moieties directed against a particular surface marker. This causes the cells expressing this marker to attach to the magnetic nanoparticles. Afterwards the cell solution is placed within a solid or flexible container in a strong magnetic field. In this step, the cells attach to the nanoparticles (expressing the marker) and stay on the column, while other cells (not expressing the marker) flow through. With this method, the cells can be separated positively or negatively with respect to the particular marker(s).
  • In case of a positive selection the cells expressing the marker(s) of interest, which attached to the magnetic column, are washed out to a separate vessel, after removing the column from the magnetic field.
  • In case of a negative selection the antibody or selective moiety used is directed against surface markers(s) which are known to be present on cells that are not of interest. After application of the cells/magnetic nanoparticles solution onto the column the cells expressing these antigens bind to the column and the fraction that goes through is collected, as it contains the cells of interest. As these cells are non-labelled by the selective antibodies or moiety(s) coupled to nanoparticels, they are “untouched”.
  • The known manual or semi-automated solid tissue processing steps are labour-intensive and require a knowledge of the art.
  • In addition where the material is used for therapeutic purposes, the processing requires strict regulated environmental conditions during handling the cell cultures, for example tissue processing as apart of or prior to disaggregation; enzymatic digestion and transfer into storing devices or incubation conditions for disaggregation/cellularisation and viable tissue yields. Typically this process would require multiple pieces of laboratory and tissue processing equipment, and personal with the skills and knowledge of the scientific art with critical stages contained within either hazard containment or tissue processing facility(s) aseptic environment(s) in order to perform the same activity safely and also minimise the risk of contamination(s).
  • The invention therefore arises from a need to provide improved solid tissue processing, including an apparatus/device that undertakes said processing that achieves the unmet need described above.
  • SUMMARY OF INVENTION
  • The present invention concerns a single use aseptic kit comprising a disaggregation module for receipt and processing of material comprising solid mammalian tissue; an optional enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate; and a stabilisation module for optionally further processing and/or storing disaggregated product material, wherein each of said modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between; and wherein each of said modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers.
  • In prior art the tissue may undergo physical and or enzymatic disaggregation/cellularisation in a single container. In the present invention sets of containers which are interconnected and have specific separate functions maintain an aseptically closed system to process, optionally enrich but stabilise the disaggregated and cellularised solid tissue product. Essentially the invention provides a rapid pre-sterilised environment to minimise the time required and risk of contamination or operator exposure during the processing of the solid tissue.
  • The kit described here allows for closed solid tissue processing eliminating the risk of contamination of the final cellularised product compared to standard non-closed tissue processing. This is especially when the process is performed within a tissue retrieval/procurement site and requires storage prior to final cell processing for its ultimate utility. In addition, safety of the operator is increased due to reduction of direct contact with biological hazardous material which may contain infectious organisms such as viruses.
  • The kit also enables either all of or a portion of the finally processed cellularised material to be stabilised for either transport or storage prior to being processed for its ultimate utility.
  • The invention will enable the solid tissue to be processed at the time of tissue retrieval, or later if required, without impact upon the retrieval procedure or the viability of the cellularised product.
  • In some embodiments employing optional enrichment via a form of physical purification to reduce impurities such as no longer required reagents; cell debris; non-disaggregated tissue and fats. A single cell or small cell number aggregates can be enriched for stabilisation after disaggregation by excluding particles and fluids of less than 5 μm or incompletely disaggregated material of or around 200 μm across or larger but this will vary upon the tissue and the efficiency of disaggregation and various embodiments in the form of tissue specific kits may be employed depending upon the tissue or ultimate utility of the disaggregated solid tissue.
  • In some embodiments the one or more flexible containers comprise a resilient deformable material. The one or more flexible containers of the disaggregation module may comprise one or more sealable openings. The one or more flexible containers of the disaggregation module and/or the stabilisation module may also comprise a heat sealable weld.
  • In further embodiments the one or more flexible containers that are part of any module comprise internally rounded edges.
  • The one or more flexible containers of the disaggregation module may comprise disaggregation surfaces adapted to mechanically crush and shear the solid tissue therein.
  • Further, the one or more flexible containers of the enrichment module may comprise a filter adapted to retain a retentate of cellularised disaggregated solid tissue.
  • In embodiments, one or more flexible containers of the stabilisation module comprise media formulation for storage of viable cells in solution or in a cryopreserved state. In some embodiments the
  • In further embodiments the kit further comprises a radio frequency or other digitally recognisable identification tag so that it may be scanned and recognised during automated processing, such as with/in the automated device in embodiments of the present invention. Crucially the tag provides information about the conditions and steps required to be auto processed, so simply by scanning the kit, any automated system used with the kit to process the tissue can be undertaken without further intervention or contamination. Once the tissue sample has been placed in the disaggregation module, it can for example be sealed, manually, or automatically, before processing begins.
  • In this regard, in preferred embodiments that include a device, the kit associated tag is detected by the device's processor and the device then runs a specific program according to a type of disaggregation and/or enrichment and/or stabilisation process; one or more types of media used in those processes; including an optional freezing solution suitable for controlled rate freezing. Put another way, the kit is therefore be readable by an automated device used to execute a specific fully automatic method for processing the specific tissue when inserted to such a device.
  • The invention is particularly useful in a sample processing, particularly automated processing. Thus, in a further aspect the invention concerns use of the single use aseptic kit described above in a semi-automated process for the aseptic disaggregation and/or enrichment and stabilisation of mammalian cells or cell aggregates.
  • A particular advantage is that solid tissue disaggregation (and optional processes including all described manipulations herein described required to achieve optimal results) can be performed in a closed system, i.e. an aseptic process with minimal risk of contaminations and with minimal user knowledge.
  • The invention further relates to an automated device for semi-automated aseptic disaggregation and/or enrichment and stabilisation of cells or cell aggregates from mammalian solid tissue comprising a programmable processor and the single use aseptic kit as described in any of the before mentioned examples above.
  • In embodiments, as previously described, the device may have a comprising radio frequency identification tag reader to recognise the single use kit. The programmable processor is capable of recognising the single use aseptic kit via its tag and subsequently able to execute the kit programme which defines the type of disaggregation, enrichment and stabilisation processes together with the respective media types required for those processes.
  • In this regard, the programmable processor is adapted to communicate with and control one or more of: the disaggregation module; the enrichment module; and the stabilisation module of the device. The device, including its processor, may therefore have multiple functionality to assess the flow of materials through the kit making decisions of when a step is completed as part of the pre-programmed functions and the feedback the device gets from its sensors.
  • For example, the programmable processor may control the disaggregation module to enable a physical and/or biological breakdown of the solid tissue material in that container. The programmable processor may also control the disaggregation module to enable a physical and enzymatic breakdown of the solid tissue material.
  • In some embodiments the enzymatic breakdown of the solid tissue material is by the selection and provision of one or more media enzyme solutions selected from collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase H1, pepsin, or mixtures thereof.
  • In addition or alternatively the programmable processor may control disaggregation enabling the surfaces within the disaggregation flexible containers to mechanically crush and shear the solid tissue. In embodiments, the disaggregation surfaces are controlled by mechanical pistons, for example.
  • In some embodiments, the programmable processor controls the stabilisation module to cryopreserve the enriched disaggregated solid tissue in the container, for example, this may be achieved by using a programmable temperature setting, a condition which is determined by reading the tag of the kit inserted in the device.
  • In some embodiments, to undertake different functions of the process, one or more of the additional components of the device and/or kit are provided. Such features may be available in any combination. This may include for example: sensors in the device capable of recognising whether a disaggregation process has been completed in the disaggregation module of the kit prior to transfer of the disaggregated solid tissue to the optional enrichment module; weight sensors to determine an amount of media required in the containers of one or more of the disaggregation module; the enrichment module; and/or the stabilisation module and means to control that transfer of material between respective containers; and temperature sensors to control the temperature within the containers of the one or more of the disaggregation module; the enrichment module; and/or the stabilisation module.
  • Other possible features include an optional bubble sensor to control the transfer of media between the input and output ports of each container in the module; one or more pumps may provide means to control the transfer of media between the input and output ports; and/or pressure sensors to assess the pressure within the enrichment module; valves to control an optional tangential flow filtration process within the enrichment module; and/or one or more clamps to control the transfer of media between the input and output ports of each module.
  • For example, the programmable processor is adapted to maintain an optimal storage temperature range in the stabilisation module until the container is removed; or executes a controlled rate of freezing.
  • These embodiments of the device and kit allow solid tissue derived cells or cell aggregates to be: stored for short periods (minutes to days) or stored for long periods (multiple days to years) prior to their ultimate utility depending on the type or stabilisation process used with the stabilisation module.
  • For ease of use, the device of the invention may further comprise a user interface. That interface may comprise a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof.
  • In many cases it is desirable that the automated device is adapted to be transportable and thus may comprise dimensions that permit easy manoeuvrability and/or aid movement such as wheels, tyres, handles and the like.
  • The final cellular material product can then be used for but not limited to either: regenerative medicine, adoptive cell therapies, ATMPs, diagnostics or to further the basic scientific understanding of tissue, cell function or organism function.
  • The combination of an aseptic kit, automated processing device and associated media formulations, which can disaggregate solid tissues to provide functional living cells or the product of the cells for subsequent therapeutic, diagnostic or scientific use, is therefore highly desirable.
  • In some embodiments the cells produced using the kit and/or device of the invention are useful for providing functional living cells and maybe cultured further for that use. Cell culture is a process by which cells are grown outside the original host using controlled environmental and supportive conditions which vary by cell type and organism. These are often sterile artificial vessels which allow gas and temperature to be maintained and either manual or automated changes in essential nutrients, metabolites, growth factors and gases which enable regulation of the cells requirements to survive and in most cases thrive. Cell culture requirements differ broadly by the type of cell(s) and its required purpose. Cell culture conditions can be optimised for cell expansion, cell differentiation or manufacturing of different phenotypes of the cell or its products. The most commonly varied factor in culture systems is the cell culture medium, for which a vast number of recipes is known (see for example “Cell Culture Techniques” Humana Press, 1st. Edition, 2011)
  • In some embodiments disaggregated or cellularised material produced by the device and kit can be useful as the starting material to isolate specific cell populations which are grown out using stimulation or non-required cells are inhibited or apoptosis/cell death is induced resulting in a semi/purified population.
  • Such cells can be further sorted by one or more of the following processes: Fluorescence-activated cell sorting using antibody/protein labelling or natural fluorescence; Magnetic separation of cells, e.g. the magnetic activated cell sorting (MACS technology, Miltenyi Biotec GmbH, Germany). This technology requires a marker that allows direct separation of the cells of interest by an antibody coupled to a magnetic microbead (Miltenyi et al., Cytometry 1990; 11:231-238). Alternatively, where it is not possible or not desirable to actively select the target population a process of negative isolation can be employed. In this approach, non-target cells are magnetically labelled and depleted, thereby leaving the unlabelled cells of interest; Label free cell separation and sorting using physical separation methods where either the target is not known, is a mixed population and physical cell (or clumps of cells) characteristics can be used to separate the cellular material from the current media to: remove impurities or reagents that are no longer required such as enzymes, cell debris, connective tissue, fat & mineral deposits; or exchange fluids which may be better for stabilising the cells for distribution and/or storage. It is envisaged that embodiments of the invention may include such functionality within the parameters of the processor or the automated device and operating system.
  • For example, the purity, of the disaggregated and cellularised solid tissue product, can be further increased if one or more cell surface marker(s) are used to select for or deplete a subpopulation of cells either as an independent step within the process or after processing using the methods described.
  • The present invention also relates to a method for enhanced semi-automated disaggregation cellularisation and storage of tissue derived cells. Optionally, steps of enrichment, formulation and cryopreservation are also provided.
  • In a further aspect of the invention, there is provided a semi-automatic aseptic tissue processing method comprising: automatically determining aseptic disaggregation tissue processing steps and one or more further tissue processing steps and their associated conditions from a digital tag identifier on an aseptic processing kit, optionally in accordance with the kit described herein; placing a tissue sample into a flexible plastic container of the aseptic processing kit; and processing the tissue sample by automatically executing the one or more tissue processing steps by communicating with and controlling the disaggregation module; the optional enrichment module; and the stabilisation module.
  • The one or more automatically executed processes may be selected from one or more of:
  • 1) transferring media, preferably enzyme media, into the disaggregation chamber (for example, in accordance with the sealable disaggregation flexible container of the kit of the invention). The media maybe transferred into the disaggregation chamber, or in one embodiment also enters and collects enzymes prior to disaggregation using one or more embodiments of the invention, e.g. a mechanism such as weight sensors which will assess the required amount of media to add either determined by: direct operator input or weight of solid tissue. Incubating with the media at an optimal temperature of between 30 & 37° C. but could be as low as 0° C. upto 40° C. for at least 1 minute to several hours but more preferable 15 to 45 minutes.
  • 2) undertaking physical disaggregation for a minimum of a few seconds up to several hours with an optimal time of between 1 and 10 minutes required to break up the solid tissue until there is no visual change (Table 1). The disaggregation is designed to compress the tissues using a variable speed and time depending upon the time taken to disaggregate and feedback via sensors within the disaggregation module.
  • Steps 1) or 2) may be repeated until the tissue stops changing or has been disaggregated into a liquid cell suspension (whichever comes 1st monitored by a sensor in the disaggregation module).
  • 3) removing disaggregated tissues, associated material and impurities by passing the disaggregated tissue and media through one or more filters enabling optional enrichment of the cell suspension. Direct pass through one or more mechanical filters with holes at least >0.1 μm to 1000 μm but most preferably between 50 and 250 μm and more preferably 100 μm to 200 μm. Alternatively, other separation methods maybe used such as:
      • I density based separation using centrifugation and/or sedimentation with or without a cell aligned density retention solution (e.g. Ficoll-paque GE Healthcare).
      • II Hydrodynamic filtration where fluid flow and flow obstructing materials enhance the resolution and fractionation of the cells and impurities based on size and shape
      • III. Field flow fractionation where an applied field (e.g. flow, electric, gravitational, centrifugal) acts in a perpendicular or reverse direction to the selection flow (e.g. Tangential flow filtration, Hollow fibre flow filtration, Asymmetric flow filtration, Centrifugal flow filtration). In which case: cells or impurities which are most responsive to the force are driven to the wall where flow is lowest and therefore a long retention time; while cells or impurities which are least responsive to the force remain laminar to the flow and elute quickly (tangential flow filtration)
      • IV Acoustophoresis where one or more an acoustic frequency(ies) tuned to or harmonized with populations of cells or impurities is used to drive the required cells or impurities in a tangential path to the input stream.
  • 4. Re-suspending the disaggregated cell product in fresh or additional media. This could be a cell enrichment media in order to undergo an independent targeted enrichment procedure or direct cell culture or cold storage media (such as HypoThermosol® from BioLife Solutions).
  • 5. Transferring to a stabilising module containers for storage for hours to days or
  • 6. Re-suspending in, or addition of a, cryoprotectant—a freezing solution for storage of the disaggregated solid tissue derived product for days to years (such as CryoStor® Freezing solution from BioLife Solutions) and transferring to one or more flexible stabilising module having a cryopreservation container(s)
  • 7. Performing a controlled rate freezing process
  • 8. Separating the aseptic processing kit from the device for independent storage or distribution.
  • During such steps it is apparent that the disaggregated module and the storage module may comprise one and the same flexible container, for receiving the sample and storing the sample and a further flexible container for housing the media for disaggregation. In some embodiments the same flexible containers are part of different modules of the kit.
  • DESCRIPTION OF THE INVENTION
  • The processing of tissue to cells according to the kit, semi-automated device and methods of present disclosure are described further in the accompanying examples and figures numbered 1 to 7.
  • Moreover, by utilising the kit, device and processes described herein, in conjunction with ordinary skills in the art, further embodiments of the present disclosure can be readily identified. Those skilled in the art will readily understand known variations.
  • Definitions of the Disclosure
  • Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
  • “depletion” as used herein refers to a process of a negative selection that separates the desired cells from the undesired cells which are labelled by one marker-binding fragment coupled to a solid phase.
  • “disaggregation or disaggregate” as used herein refers to the transformation of solid tissue into a single cells or small cell number aggregates where a single cell as a spheroid has a diameter in the range of 5 μm, 6 μm, 7 μm, 8 μm, 9 μm, 10 μm, 20 μm, 30 μm, 40 μm, 50 μm, 60 μm, 70 μm, 80 μm, 90 μm, 100 μm or more where this is more usually between 7 to 20 μm.
  • “cellularised or cellularisation” as used herein refers to the process of disaggregation where by the solid tissue a multicellular material generally made up of multiple cell lineages/types is broken down into small numbers of cells including but not limited to one cell but could be multiple cells of various lineages or cell types in very small numbers i.e. clump of cells or cell aggregates.
  • “engineered” as used herein refers to either addition of nucleic material or factors which change the tissue derived cell function from their original function to have a new or improved function for its ultimate utility.
  • “filtrate” as used herein refers to the material that passes through a filter, mesh or membrane.
  • “flexible container” as used herein refers to a flexible packaging system in multiple formats with one or more different types of film. Each film type is selected to provide specific characteristics to preserve the physical, chemical, and functional characteristics of the sterile fluids, solid tissue derived cellular material and the container integrity depending upon the step of the process.
  • “freezing solution” or “cryopreservation solution” also referred in the field to as the cryoprotectant is a solution that contains cryoprotective additives. These are generally permeable, non-toxic compounds which modify the physical stresses cells are exposed to during freezing in order to minimise freeze damage (i.e. due to ice formation). Most commonly a % Vol/Vol of one or more of the following: Dimethylsulphoxide (DMSO); Ethylene glycol; Glycerol; 2-Methyl-2,4-pentanediol (MPD); Propylene glycol; Sucrose; & Treha lose.
  • “media” means various solutions known in the art of cell culturing, cell handling and stabilisation used to reduce cell death, including but not limited to one or more of the following media Organ Preservation Solutions, selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove's medium, X-VIVO™, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTE™ solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's Solution. The media can be standard cell media like the above mentioned media or special media for e.g. primary human cell culture (e.g. for endothelia cells, hepatocytes or keratinocytes) or stem cells (e.g. dendritic cell maturation, hematopoietic expansion, keratonocytes, mesenchymal stem cells or T cell expansion). The media may have supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, antibiotics, attachments factors, growth factors and cytokines, hormones, metabolic inhibitors or solubilising agents. Various media are commercially available e. g. from ThermoFisher Scientific or Sigma-Aldrich.
  • “non-labelled” or “untouched” as used herein refers to the cells which are not bound by one marker-binding fragment coupled to a solid phase. The non-labelled, untouched cell fraction contains the desired target cells.
  • “non-target cells” as used herein refers to cells which are specifically bound by one marker-binding fragment which is coupled to a solid phase that is used to remove an unwanted cell type.
  • “positively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are the required population of cells.
  • “negatively separated” as used herein refers to the active separation of cells which are bound by one marker-binding fragment coupled to a solid phase and these cells are not the required population of cells.
  • “purity” as used herein refers to the percentage of the target population or populations desired from the original solid tissue.
  • “regenerative medicine(s)”, “adoptive cell therapy(ies)” or “advanced therapy medicinal product(s)” are used interchangeably herein to refer to cellular material that is used for therapeutic purposes of one or more mammals either by: the action of a part of or all of the cellular material; the supportive actions of a part of or all of the cellular material with the aim to improve the wellbeing of the mammal after application. The therapeutic cells can either be used directly or may require further processing, expansion and/or engineering to provide these actions.
  • “sample” as used herein refers to a sample containing cells in any ratio. Preferentially, these cells are viable. But, these cells can also be fixed or frozen cells which may be used for subsequent nucleic acids or protein extraction. The samples may be from animals, especially mammals such as mouse, rats or humans. Any compressible solid tissue that contains cells can be used. The invention is illustrated mainly through the isolation of hematopoietic and cancer cells from solid tumour tissue. However, the invention relates to a method for isolation of a breadth of cells from any mammalian solid tissue.
  • “marker” as used herein refers to a cell antigen that is specifically expressed by a certain cell type. Preferentially, the marker is a cell surface marker, so that enrichment, isolation and/or detection of living cells can be performed.
  • “solid phase” as used herein refers to the coupling of the marker-binding fragment, e.g. an antibody, bound to another substrate(s), e.g. particles, fluorophores, haptens like biotin, polymers, or larger surfaces such as culture dishes and microtiterplates. In some cases the coupling results in direct immobilization of the antigen-binding fragment, e.g. if the antigen-binding fragment is coupled to a larger surface of a culture dish. In other cases this coupling results in indirect immobilisation, e.g. an antigen-binding fragment coupled directly or indirectly (via e.g. biotin) to a magnetic bead is immobilised if said bead is retained in a magnetic field. In further cases the coupling of the antigen-binding fragment to other molecules results not in a direct or indirect immobilization but allows for enrichment, separation, isolation, and detection of cells according to the present invention, e.g. if the marker-binding fragment is coupled to a chemical or physical moiety which then allows discrimination of labelled cells and non-labelled cells, e.g. via flow cytometry methods, like FACSsorting, or fluorescence microscopy.
  • “solid tissue” as used herein refers to a piece or pieces of animal derived mammalian solid tissue which by its three dimensions i.e. length, breadth and thickness as a geometrical body is larger than the size of multiple individual cell based units and often contains connective materials such as collagen or a similar matrix that make up structure of the tissue whereby said solid tissue cannot flow through tubes or be collected by a syringe or similar small conduit or receptacle and is i.e. with dimensions in the range of 500 μm, 1 mm, 2 mm, 3 mm, 4 mm, 5 mm, 1 cm, 2 cm, 3 cm, 4 cm, 5 cm, 10 cm, 20 cm, 30 cm or more
  • “particle” as used herein refers to a solid phase such as colloidal particles, microspheres, nanoparticles, or beads. Methods for generation of such particles are well known in the field of the art. The particles may be magnetic particles or have other selective properties. The particles may be in a solution or suspension or they may be in a lyophilised state prior to use in the present invention. The lyophilized particle is then reconstituted in convenient buffer before contacting the sample to be processed regarding the present invention.
  • “magnetic” in “magnetic particle” as used herein refers to all subtypes of magnetic particles which can be prepared with methods well known to the skilled person in the art, especially ferromagnetic particles, superparamagnetic particles and paramagnetic particles. “Ferromagnetic” materials are strongly susceptible to magnetic fields and are capable of retaining magnetic properties when the field is removed. “Paramagnetic” materials have only a weak magnetic susceptibility and when the field is removed quickly lose their weak magnetism. “Superparamagnetic” materials are highly magnetically susceptible, i.e. they become strongly magnetic when placed in a magnetic field, but, like paramagnetic materials, rapidly lose their magnetism.
  • “marker-binding fragment” as used herein refers to any moiety that binds preferentially to the desired target molecule of the cell, i.e. the antigen. The term moiety comprises, e.g., an antibody or antibody fragment. The term “antibody” as used herein refers to polyclonal or monoclonal antibodies which can be generated by methods well known to the person skilled in the art. The antibody may be of any species, e.g. murine, rat, sheep, human. For therapeutic purposes, if non-human antigen binding fragments are to be used, these can be humanized by any method known in the art. The antibodies may also be modified antibodies (e.g. oligomers, reduced, oxidized and labelled antibodies). The term “antibody” comprises both intact molecules and antibody fragments, such as Fab, Fab′, F(ab′)2, Fv and single-chain antibodies. Additionally, the term “marker-binding fragment” includes any moiety other than antibodies or antibody fragments that binds preferentially to the desired target molecule of the cell. Suitable moieties include, without limitation, oligonucleotides known as aptamers that bind to desired target molecules (Hermann and Pantel, 2000: Science 289: 820-825), carbohydrates, lectins or any other antigen binding protein (e.g. receptor-ligand interaction).
  • “retentate” as used herein refers to the material that does not pass through a filter, mesh or membrane.
  • “ultimate utility” as used herein refers to manufacture of or direct use in regenerative medicines, adoptive cell therapies, ATMPs, diagnostic in vitro studies or scientific research.
  • With reference to FIG. 1 there is disclosed:
    • 1 a Flexible container for: disaggregation; and digestion in the embodiment involving enzymatic digestion.
    • 1 b Open end for transfer of solid tissue materials into container 1 a
    • 1 c hanging holes to support container 1 a
    • 1 d target heat weld location to seal container 1 a using heat welder 13 m
    • 1 e rounded edges on internal container 1 a surfaces to reduce losses which may occur as part of transfer to examples illustrated in FIG. 2 (a, b or c) or FIG. 3 (a or b)
    • 1 f tubing 1 f enables media 3 a to be transferred into container 1 a via sterile filter 2 a
    • 1 g in example tubing 1 g enables digestion enzymes 3 b to be transferred into container 1 a via sterile filter 2 b
    • 1 h after disaggregation, especially involving enzymatic digestion a phase of incubation, the mixture is transferred out via tubing 1 h via filter unit 4 a containing filter 4 b prior to entering
    • 2 a spike and sterile filter for media 3 a
    • 2 b spike and sterile filter for enzymes 3 b in one example, where enzymes are required
    • 3 a media for disaggregation and in one example enzymatic digestion
    • 3 b enzymes for disaggregation in one example
    • 4 a flexible filter unit
    • 4 b non-disaggregated tissue filter
    • 5 a tubing clamp to allow media (3 a) to enter the flexible container 1 a via filter 2 a
    • 5 b in one example where enzymes are used a tubing clamp will allow enzymes (3 b) to enter the flexible container 1 a via filter 2 b
    • 5 c tubing clamp to allow contents of flexible container 1 a to pass via filter 4 a into one or more examples identified in FIG. 2 (a-c) Or FIG. 3 (a or b)
    • FIG. 2a provides a further example of the invention in which:
    • 2 c spike and sterile filter for media
    • 3 a in one example short term storage media
    • 3 c freezing solution a media required for cryopreservation in one of the examples illustrated in FIG. 2a or FIG. 3b
    • 4 c in one example an additional flexible filter module containing filters 4 d & 4 e
    • 4 d in one example FIG. 2a a flexible filter unit may be required for additional size segregation of cell/tissue clumps
    • 4 e in one example FIG. 2a a flexible filter unit is required to retain cells but allow the media/cell fragments to be washed out
    • 5 d in one example FIG. 2a tubing clamp is in place to stop material from container 1 a that has passed though 4 a & 4 c from returning back to container 1 a
    • 5 e in one example FIG. 2a tubing clamp is in place to allow waste material from container 1 a that has passed through 4 a, 4 c and 4 e to enter container 6 a but stop media (3 a or 3 c) entering via filter 2 c from entering container 6 a
    • 5 f both tubing clamps stop material from container 1 a that has passed though filters 4 a, 4 c and 4 e from entering the tubing to the media container (3 a or 3 c) or transferring to one of the examples FIG. 3 (a or b) before the waste has passed into container 6 a via 5 e. Once the waste has been depleted then tubing clamps 5 e and 5 d close and both tubing clamps 5 f allowing media (3 a or 3 c) to transfer cells within filter 4 e into one of the examples identified in FIG. 3 (a orb)
    • 6 a a waste container
    • 6 b hanging holes to support container 6 a
    • FIG. 2b provides yet a further example in which:
    • 5 g a tubing clamp in place to allow contents of container 1 a to enter the flexible container 7 a via filter 4 a
    • 5 h a tubing clamp in place to allow contents of container 7 a to pass through filter 8 a retaining and enriching for cells while allowing waste and debris to pass through filter 8 b into container 6 a with the pressure controlled by valve 8 c before the enriched cells return to container 7 a via an open clamp 5 i
    • 5 i a tubing clamp is in place to allow contents of container 7 a via open tubing clamp 5 h to pass through filter 8 a retaining and enriching for cells while allowing waste and debris to pass through filter 8 b into container 6 a with the pressure controlled by valve 8 c before the enriched cells return to container 7 a
    • 5 j after cell enrichment has occurred then tubing clamp 5 h closes and 5 j opens allowing contents of 7 a to pass on to one of the examples FIG. 3 (a or b)
    • 6 a a waste container
    • 6 b hanging holes to support container 6 a
    • 7 a a flexible container to receive the contents of: 1 a via filter 4 a; and filter 8 a
    • 7 b hanging holes to support container 7 a
    • 7 c rounded edges on internal container 7 a to reduce losses which may occur as part of transfer to examples illustrated in FIG. 3 (a or b)
    • 7 d tubing to allow container 7 a to receive the contents of: 1 a via filter 4 a; and filter 8 a
    • 7 e tubing to allow contents of container 7 a to pass through filter 8 a retaining and enriching for cells while allowing waste and debris to pass through filter 8 b into container 6 a with the pressure controlled by valve 8 c before the enriched cells return to container 7 a via an open clamp 5 i
    • 7 f tubing to allow contents of container 7 a via open tubing clamp 5 h to pass through filter 8 a retaining and enriching for cells while allowing waste and debris to pass through filter 8 b into container 6 a with the pressure controlled by valve 8 c before the enriched cells return to container 7 a
    • 8 a contents of container 7 a can be filtered to remove waste media and debris via filter
    • 8 b while enriching for cells under the control of valve 8 c before returning to container 7 a
    • 8 b & 8 c see 8 a
  • In one example, as shown in FIG. 2c it is described that
    • 5 g a tubing clamp in place to allow contents of container 1 a to enter the flexible container 7 a via filter 4 a
    • 5 h a tubing clamp in place to allow contents of container 7 a to pass through filter 9 a retaining and enriching for cells while allowing waste and debris to pass through filter 9 b into container 6 a with the pressure controlled by valve 9 c before the enriched cells return to container 7 a via an open clamp 5 i
    • 5 i a tubing clamp is in place to allow contents of container 7 a via open tubing clamp 5 h to pass through filter 9 a retaining and enriching for cells while allowing waste and debris to pass through filter 9 b into container 6 a with the pressure controlled by valve 9 c before the enriched cells return to container 7 a
    • 5 j after cell enrichment has occurred then tubing clamp 5 h closes and 5 j opens allowing contents of 7 a to pass on to one of the examples FIG. 3 (a or b)
    • 6 a a waste container
    • 6 b hanging holes to support container 6 a
    • 7 a a flexible container to receive the contents of: 1 a via filter 4 a; and filter 9 a
    • 7 b hanging holes to support container 7 a
    • 7 c rounded edges on internal container 7 a to reduce losses which may occur as part of transfer to examples illustrated in FIG. 3 (a or b)
    • 7 d tubing to allow container 7 a to receive the contents of: 1 a via filter 4 a; and filter 9 a
    • 7 e tubing to allow contents of container 7 a to pass through filter 9 a retaining and enriching for cells while allowing waste and debris to pass through filter 9 b into container 6 a with the pressure controlled by valve 9 c before the enriched cells return to container 7 a via an open clamp 5 i
    • 7 f tubing to allow contents of container 7 a via open tubing clamp 5 h to pass through filter 9 a retaining and enriching for cells while allowing waste and debris to pass through filter 9 b into container 6 a with the pressure controlled by valve 9 c before the enriched cells return to container 7 a
    • 9 a contents of container 7 a can be filtered to remove waste media and debris via filter 9 b while enriching for cells under the control of valve 9 c before returning to container 7 a
    • 9 b & 9 c see 9 a
  • FIG. 3a provides yet a further example of the invention in which:
    • 5 k a tubing clamp is in place to allow the contents of: 1 a (in example FIG. 1 via filter 4 a or in example FIG. 2a via filter 4 c); or 7 a (in example FIG. 2b via filter 8 a or in example FIG. 2c via filter 9 a) to be transferred into container 10 a
    • 10 a a flexible container to receive the contents of: 1 a via filter 4 a (in example FIG. 1) where examples described in FIG. 2 (a, b or c) are not required; 1 a via filters 4 a & 4 c (in example FIG. 2a ); 7 a via filter 8 a (in example FIG. 2b ); or 7 a via filter 9 a (in example FIG. 2c )
    • 10 b hanging holes to support container 10 a
    • 10 c rounded edges on internal container 10 a to reduce losses which may occur as part of transfer out via 10 e or f
    • 10 d tubing to enable container 10 a to receive the contents of: 1 a via filter 4 a (in example FIG. 1) where examples described in FIG. 2 (a, b or c) are not required; 1 a via filters 4 a & 4 c (in example FIG. 2a ); 7 a via filter 8 a (in example FIG. 2b ); or 7 a via filter 9 a (in example FIG. 2c )
    • 10 e tubing to enable contents of container 10 a to be withdrawn via connector 10 h
    • 10 f tubing with a flexible membrane to enable a sterile spike to be introduced via cover
    • 10 g to enable contents of container 10 a to be withdrawn 10 g aseptic cover for tubing containing membrane 10 f
    • 10 h connector to enable contents of 10 a to be withdrawn via tubing 10 e
  • In a further example, as shown in FIG. 3b there is provided:
    • 2 c spike and sterile filter for media (3 c)
    • 3 c media required for cryopreservation
    • 5 l tubing clamp to allow the contents of: 1 a (in example FIG. 1 via filter 4 a or in example FIG. 2a via filter 4 c); or 7 a (in example FIG. 2b via filter 8 a or in example FIG. 2c via filter 9 a) to be transferred into container 11 a
    • 5 m tubing clamp to allow media (3 c) to enter the flexible container 11 a via filter and spike 2 c
    • 5 n tubing clamp to allow contents of container 11 a to enter one of the 12 a containers depending on the open or closed status of tubing clamps 5 o to 5 t
    • 5 o-5 t tubing clamps to allow contents of container 11 a to enter one of the 12 a containers depending on the open or closed status of tubing clamps 5 o to 5 t
    • 11 a a flexible container to receive the contents of: 1 a via filter 4 a (in example FIG. 1) where examples described in FIG. 2 (a, b or c) are not required; 1 a via filters 4 a & 4 c (in example FIG. 2a ); 7 a via filter 8 a (in example FIG. 2b ); or 7 a via filter 9 a (in example FIG. 2c )
    • 11 b hanging holes to support container 11 a
    • 11 c rounded edges on internal container 11 a to reduce losses which may occur as part of transfer out via 11 f
    • 11 d tubing to enable container 10 a to receive the contents of: 1 a via filter 4 a (in example FIG. 1) where examples described in FIG. 2 (a, b or c) are not required; 1 a via filters 4 a & 4 c (in example FIG. 2a ); 7 a via filter 8 a (in example FIG. 2b ); or 7 a via filter 9 a (in example FIG. 2c )
    • 11 e tubing to allow cryopreservation media 3 c to be transferred into container 11 b
    • 11 f tubing to enable the contents of 11 a to be transferred to container(s) 12 a
    • 12 a flexible containers to cryopreserve and store the final disaggregated cells product.
    • 12 b a fixture allowing aseptic transfer of the cells out of the container (12 a)
    • 12 c a space as part of 12 a suitable for the volume to be stored
    • 12 d a target location for welding the tubing and secondary flexible container as part of 12 a using welder 13 n
  • FIG. 4 shows a further example of the device and kit of the invention in which:
    • 13 a Pegs for hanging media 3 a, 3 b, 3 c
    • 13 b pegs connected to weight sensors for hanging containers 1 a and depending on the examples used these could include one or more of: 7 a, 10 a & 11 a. Where the weight sensors are used to define decision stages to control the automated processing of the materials
    • 13 c Heat welder to seal container 1 a at target site 1 d after tissue has been introduced
    • 13 d disaggregation module with an opening that can be closed and locked to enable disaggregation and in the example that uses digest enzymes is capable of controlling temperatures between 0° C. and 40° C. to a tolerance of 1° C. to enable digestion. The module also has a built in sensor to assess the level of solid tissue disaggregation by determining the variation in light distribution against time to identify change and thereby identifying completion of the disaggregation process which will occur over a period of seconds to hours.
    • 13 e final formulation module with an enclosure to allow temperature control of either container 10 a or 11 a depending on the example used which is capable of controlling temperatures between 0° C. and ambient environmental temperature to a tolerance of 1° C.
    • 13 f disaggregation surfaces which come directly into contact with container 1 a and pushes against the back of the module 13 d enclosure which can be closed and locked during disaggregation and digestion where enzymes are utilised.
    • 13 g tubing clamp
    • 13 j tubing clamp
    • 13 h peristaltic tubing pumps
    • 13 i tubing locators
    • 13 k tubing valve required for examples FIGS. 2b & 2 c
    • 13 l Pegs for hanging containers depending on the examples used these could include one or more of: 6 a & 12 a
    • 13 m tubing welder and cutter required for example FIG. 3b for tubing to container(s) 12 a
    • 13 n tubing welder required for example FIG. 3b for tubing to container(s) 12 a at target location 12 d
    • 13 o controlled rate cooling module capable of cooling or maintaining any temperature between 8° C. and at least −80° C.
    Example Method
  • The method of the invention is exemplified according to the following process. It is clearly stated that other than the essential features of the method, the various optional steps listed herein can be independently combined to achieve the relevant technical advantages associated with the type of sampling and result to be achieved.
  • A semi-automatic aseptic tissue processing method comprising: automatically determining aseptic disaggregation tissue processing steps and one or more further tissue processing steps and their associated conditions from a digital tag identifier on an aseptic processing kit, optionally in accordance with the kit described herein; placing a tissue sample into a flexible plastic container of the aseptic processing kit; and processing the tissue sample by automatically executing the one or more tissue processing steps by communicating with and controlling the disaggregation module; the optional enrichment module; and the stabilisation module.
  • Essentially the process may comprise taking an open ended bag (1st flexible container that is part of disaggregation module) that will receive the biopsy/tissue sample which is already connected via one or more conduits to (conduit) or can be connected via a manual operator controlled aseptic connection to
  • I. a single container with digestion media (2nd flexible container that is part of the disaggregation module) and with or without a stabilisation solution (same 2nd flexible container is part of the stabilisation module also)
  • II. one container with a digestion solution (2nd flexible container that is part of the disaggregation module) & another container with a stabilisation solution (4th flexible container is part of the stabilisation module)
  • on addition of the biopsy and sealing of the open ended bag the digestion media can be added via the conduit or aseptic connections (conduit/ports claim 1) and the tissue material processed.
  • On completion of the digestion by which point the tissue is now a single or small number aggregate cellular suspension the cells can optionally be filtered prior to step 4 (optional enrichment module for filtration comprises the 1st flexible container containing sample and filtered to a 3rd container for receiving the enriched filtrate)
  • Where the stabilisation media is not present in the same flexible container i.e. option 2.II.
  • this will require the container with stabilisation solution to be added by opening the attached conduit or manual operator controlled aseptically connection to be competed and said connection to be opened enabling in both cases the stabilisation solution to be added before the process continues.
  • The single or small number aggregate cellular suspension in the original flexible container or which may be optionally subdivided into multiple storage stabilisation containers thereafter are maintained in a stable state on the device and/or will undergo cryopreservation prior to removal for, transport, storage and or used in their ultimately utility. (The stabilisation module also comprises 1st or 3rd container as used in storage/freezing/storage)
  • In one further non-limiting example of the process:
    • a) Collection of tissue sample by a separate procedure such as biopsy's or surgery to collect the required tissue material (not part of the invention) is placed into the initial flexible plastic container (see FIG. 1-container 1 a).
    • b) Media (see example FIG. 1-media 3 a) is transferred into the disaggregation chamber, or in one example also enters and collects enzymes (see FIG. 1-enzymes 3 b), prior to disaggregation using one or more of the following examples of the invention a mechanism such as weight sensors (see FIGS. 1-13 b as part of module 13 d) will assess the required amount of media to add either determined by: direct operator input or weight of solid tissue.
    • c) The single use flexible disaggregation container, solid tissue, media and in one example enzymes are combined during a physical disaggregation process for a minimum of a few seconds up to several hours with an optimal time of between 1 and 10 minutes required to break up the solid tissue until there is no visual change (FIG. 5 including Table 1). The disaggregation device is designed to compress the tissues using a variable speed and time depending upon the time taken to disaggregate and feedback via sensors within the disaggregation module (see example FIG. 1-13 d).
    • d) In one embodiment where enzymes are present this will require incubation periods at an optimal temperature of between 30 & 37° C. but could be as low as 0° C. up to 40° C. for at least 1 minute to several hours but more preferable 15 to 45 minutes.
    • e) Step c and in the embodiment where enzymes step d) can be repeated until the tissue stops changing or the see example has been disaggregated into a liquid cell suspension whichever comes 1st monitored by a sensor in the disaggregation module disaggregation module (FIG. 1-13 d).
    • f) In one embodiment incompletely disaggregated tissues, associated material and impurities are removed enabling enrichment of the cell suspension by passing the disaggregated tissue and media using one or more of the following embodiments:
    • i. Direct pass through one or more mechanical filters with holes at least >0.1 μm to 1000 μm but most preferably between 50 and 250 μm and more preferably 100 μm to 200 μm (illustrated in FIG. 2a )
    • ii. Density based separation using centrifugation and/or sedimentation with or without a cell aligned density retention solution (e.g. Ficoll-paque GE Healthcare).
    • iii. Hydrodynamic filtration where fluid flow and flow obstructing materials enhance the resolution and fractionation of the cells and impurities based on size and shape
    • iv. Field flow fractionation where an applied field (e.g. flow, electric, gravitational, centrifugal) acts in a perpendicular or reverse direction to the selection flow (e.g. Tangential flow filtration, Hollow fibre flow filtration, Asymmetric flow filtration, Centrifugal flow filtration). In which case: cells or impurities which are most responsive to the force are driven to the wall where flow is lowest and therefore a long retention time; while cells or impurities which are least responsive to the force remain laminar to the flow and elute quickly (tangential flow filtration illustrated in FIG. 2b & c)
    • v. Acoustophoresis where one or more an acoustic frequency(ies) tuned to or harmonized with populations of cells or impurities is used to drive the required cells or impurities in a tangential path to the input stream.
    • g) In one embodiment the disaggregated enriched tissue product will be resuspended in a fresh media (FIG. 2a using media 3 a) such as:
    • i. a cell enrichment media in order to undergo an independent targeted enrichment procedure as described previously
    • ii. direct cell culture or cold storage media (such as HypoThermosol® from BioLife Solutions.
    • h) in the embodiment employed in g) the resuspended disaggregated solid tissue derived product will be transferred to one of the embodiment final product containers (illustrated in FIG. 3a ) for storage for hours to days prior to being used for its ultimate utility.
    • i) otherwise after step f) the embodiment (illustrated in FIG. 3b ) will apply where the disaggregated solid tissue derived product will undergo re-suspension in a cryoprotectant (FIG. 3b -media 3 c) a freezing solution for storage of the disaggregated solid tissue derived product for days to years such as CryoStor® Freezing solution from BioLife Solution.
    • j) At this stage the disaggregated solid tissue derived product re-suspended in freezing solution using the device (FIG. 4-module 13 e) will be transferred to 1 or more flexible cryopreservation container(s) (illustrated in FIG. 3a -container 12 a) and in one embodiment of the device it will perform a controlled rate freezing process using the device (FIG. 4-module 13 o).
    • k) After which the bags can be separated from the device and aseptic processing kit for independent storage or distribution.
  • FIGS. 6 and 7 describe further examples in which the disposable kit of the invention can be used with an automatic device for semi-automatic aseptic processing of tissue samples.
  • FIG. 6 describes the following semi-automatic aseptic tissue processing method using multiple flexible containers for different starting solutions that are part of the modules of the process used for disaggregation and stabilisation.
  • Process step 1—The user may login to device and scan the tag on the aseptic kit using the device to transfer the automatic processing steps to be used. The device processor recognises the tag and is provided with information needed to carry out the specific processing instructions related to that particular kit.
  • Process step 2—The digestion media containing flexible bag (part of disaggregation module) and cryo/stabilisation solution containing flexible bag (part of the stabilisation module) are each hung or secured to the device.
  • Process step 3—The biopsy or tissue sample for processing may be placed into a flexible container (part of both modules) of the aseptic kit via an open end.
  • Process step 4—The flexible container comprising the sample may then be sealed using a heat weld to close the open end (used to add the sample during initial processing).
  • Process step 5—The user may then interact with the user interface of the processor to confirm the tissue sample is present and enter any further tissue material specific information, if required.
  • Process step 6—Digestion media and cryo/stabilisation solution flexible containers are connected with the flexible container housing the sample, after which it maybe placed into the device for automatic processing.
  • Process step 7—The device executes the cycles according to the kit information undertaking disaggregation of the sample and stabilisation/cryo preservation of resulting cells.
  • Process step 8—When stabilised/frozen disconnect and discard used media and cryo/stabilisation containers of kit. Tissue processed into single or multi-cell solution in flexible container is disconnected before transferring into storage or transport container prior to its ultimate utilisation.
  • FIG. 7 describes how flexible containers comprising the media used in the process may be shared between the modules of the aseptic processing kit and method.
  • Process step 1—The user may login to device and scan the tag on the aseptic kit using the device to transfer the automatic processing steps to be used.
  • Process step 2—A flexible bag (part of disaggregation/stabilisation module) comprising both the media and cryo/stabilisation solution is hung or otherwise secured to the device.
  • Process step 3—The biopsy or tissue sample for processing may be placed into a further flexible container (part of both modules) of the aseptic kit via an open end.
  • Process step 4—The flexible container comprising the sample may then be sealed using a heat weld to close the open end.
  • Process step 5—The user may then interact with the user interface of the processor to confirm the tissue sample is present and enter any tissue material specific information, if required.
  • Process step 6—Digestion media and cryo/stabilisation solution flexible container is connected with the flexible container housing the sample, after which it maybe placed into the device for automatic processing.
  • Process step 7—The device cycles to enable disaggregation of the sample and stabilisation of resulting cells, optionally via cryopreservation.
  • Process step 8—When freezing/stabilising is complete the user disconnects and discard used flexible containers of kit. Tissue processed into single or multi-cell solution in the remaining flexible container is disconnected before transferring into storage or transport container prior to its ultimate utilisation.
  • Enzymatic Digestion
  • By way of example, in another embodiment of the method of the invention, where the disaggregation process is being supplemented with enzymatic digestion the media formulation for enzymatic digestion must be supplemented with enzymes that aid in protein breakdown causing the cell to cell boundaries to breakdown as described above.
  • Media Formulation for Enzymatic Digestion
  • Various liquid formulations known in the art of cell culturing or cell handling can be used as the liquid formulation used for cell disaggregation and enzymatic digestion of solid tissues, including but not limited to one or more of the following media Organ Preservation Solutions, selective lysis solutions, PBS, DMEM, HBSS, DPBS, RPMI, Iscove's medium, X-VIVO™, AIM-V™, Lactated Ringer's solution, Ringer's acetate, saline, PLASMALYTE™ solution, crystalloid solutions and IV fluids, colloid solutions and IV fluids, five percent dextrose in water (D5W), Hartmann's SolutionDMEM, HBSS, DPBS, RPMI, AIM-V™, Iscove's medium, X-VIVO™, each can be optionally supplemented with additional cell supporting factors e.g. with foetal calf serum, human serum or serum substitutes or other nutrients or Cytokines to aid in cell recovery and survival or specific cell depletion. The media can be standard cell media like the above mentioned media or special media for e.g. primary human cell culture (e.g. for endothelia cells, hepatocytes or keratinocytes) or stem cells (e.g. dendritic cell maturation, hematopoietic expansion, keratonocytes, mesenchymal stem cells or T cells). The media may have supplements or reagents well known in the art, e.g. albumins and transport proteins, amino acids and vitamins, metal-ion(s), antibiotics, attachments factors, de-attachment factors, surfactants, growth factors and cytokines, hormones or solubilising agents. Various media are commercially available e. g. from ThermoFisher, Lonza or Sigma-Aldrich or similar media manufacturers and suppliers.
  • The liquid formulation required for enzymatic digestion must have sufficient calcium ions present in the of at least 0.1 mM up to 50 mM with an optimal range of 2 to 7 mM ideally 5 mM.
  • The solid tissue to be digested can be washed after disaggregation with a liquid formulation containing chelating agents EGTA and EDTA to remove adhesion factors and inhibitory proteins prior to washing and removal of EDTA and EGTA prior to enzymatic digestion.
  • The liquid formulation required for enzymatic digestion is more optimal with minimal chelating agents EGTA and EDTA which can severely inhibit enzyme activity by removing calcium ions required for enzyme stability and activity. In addition β-mercaptoethanol, cysteine and 8-hydroxyquinoline-5-sulfonate are other known inhibitory substances.
  • Cryopreservation
  • As described in preferred embodiments final cell container for cryopreservation is a flexible container manufactured from resilient deformable material. In this embodiment of the device the final container is either transferred directly to a freezer −20 to −190° C. or more optimally located in the controlled rate freezing apparatus either associated with the device or supplied separately (manufactured by for example Planer Products or Asymptote Ltd) in which the temperature of the freezing chamber and the flexible storage container(s) employed to contain the enriched disaggregated solid tissue container is controlled either by: injecting a cold gas (normally nitrogen for example Planer products); or by removing heat away from the controlled cooling surface(s). Both methods result in the ability to accurately control with an error of less than 1° C. or more preferable 0.1° C. the freezing process at the required rate for the specific cell(s) to be frozen based on the freezing solution and the desired viability of the product. This cryopreservation process must take into account the ice nucleation temperature which is ideally as close as possible to the melting temperature of the freezing solution. Followed by crystal growth in an aqueous solution, water is removed from the system as ice, and the concentration of the residual unfrozen solution increases. As the temperature is lowered, more ice forms, decreasing the residual non-frozen fraction which further increases in concentration. In aqueous solutions, there exists a large temperature range in which ice co-exists with a concentrated aqueous solution. Eventually through temperature reduction the solution reaches the glass transition state at which point the freezing solution and cells move from a viscous solution to a solid-like state below this temperature the cells can undergo no further biological changes and hence are stabilised, for years potentially decades, until required.
  • Further Applications of the Invention
  • The disaggregated cell products achieved by the method of the present invention can be cultured and/or analysed (characterised) according to all methods known to the person skilled in the art.
  • The cells obtainable by the methods disclosed herein may be used for subsequent steps such as research, diagnostics, tissue-banks, biobanks, pharmacological or clinical applications known to the person skilled in the art. Cells can then be taken into culture using a Medium optimized for this application, e.g. T cell Mixed Media (Cellular Therapeutics) usually containing but not limited to growth factors such as IL-2, IL-7, IL-15, IL-21 or stimulatory conditions such as plates or polystyrene beads coated with antibodies. In the present invention isolated cells were seeded into culture containers and maintained using procedures standardly used by a person skilled in the art such as a humidified atmosphere (1-20% usually 5% CO2, 80 to 99% usually 95% air) at temperatures between 1 to 40 usually 37° C. for several weeks and supplements may be added supplemented with 10% FBS and 3000 IU/mL IL-2.
  • Such cell cultures can be used to study e.g. cell function, tumour cell killing, cell signalling, biomarkers, cell pathways, nucleic acids, and other cell or tissue related factors that may be used to identify donor, tissue, cell or nucleic acid status.
  • The enriched cells could be used before and/or after cell culturing as a pharmaceutical composition in the therapy, e.g. cellular therapy, or prevention of diseases. The pharmaceutical composition can be used for the treatment and/or prevention of diseases in mammals, especially humans, possibly including administration of a pharmaceutically effective amount of the pharmaceutical composition to the mammal.
  • The disease may be any disease, which can be treated and/or prevented through the presence of solid tissue derived cells and/or through increasing the concentration of the relevant cells in/at the relevant place, i.e. the tumours or sites of disease. The treated and/or preventively treated disease may be any disorder, e.g. cancer or a degenerative disorder. The treatment may be the transplantation of enriched, engineered or expanded cells or any combination of these and either administered to the relevant part of the body or supplied systemically.
  • Pharmaceutical compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
  • Further specific examples:
  • Example 1
  • Impact of the Length of Disaggregation
  • Peripheral blood mononuclear cells were physically disaggregated for 0, 1, 5 & 10 minutes continuously before a being cultured in vitro for 0, 24 & 96 hours to assess cell recovery. The results demonstrate the physical process has negligible impact over 1 or 5 minutes and at 10 minutes the impact was transient where and initial reduction in viable cells at 0 hours was equivalent to non-disaggregated cells at 24 & 96 hours (FIG. 5).
  • Example 2
  • Solid Tissue Sample Size, Volume of Digestion Media, Disaggregation and Incubation Times
  • Conditions of: Solid tissue size, volume of digestion media, disaggregation time and incubation conditions have been tested and demonstrate full disaggregation of solid tissue (Table 1) except where the volume of digestion media cushioned the solid tissue during the disaggregation process resulting in 30-50% of the solid tissue remaining intact.
  • EQUIVALENTS
  • The details of one or more embodiments of the disclosure are set forth in the accompanying description above. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, the preferred methods and materials are now described. Other features, objects, and advantages of the disclosure will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.
  • The foregoing description has been presented only for the purposes of illustration and is not intended to limit the disclosure to the precise form disclosed, but by the claims appended hereto.
  • NON-PATENT LITERATURE CITED
    • Miller R G and Phillips R A. Separation of cells by velocity sedimentation. J Cell Physiol 1969; 73: 191-201
    • Buckner D, Graw R G, Eisel R J, et al. Leukapheresis by continuous flow centrifugation (CFC) in patients with chronic myelocytic leukemia (CML). Blood 1969; 33: 353-369
    • Liu W, Hou Y, Chen H, et al. Sample preparation method for isolation of single-cell types from mouse liver for proteomic studies. Proteomics 2011; 11: 3556-3564
    • Nagase K, Kimura A, Shimizu T, et al. Dynamically cell separating thermo-functional biointerfaces with densely packed polymer brushes. J Mater Chem 2012; 22: 19514-19522
    • Rembaum A, Yen R C K, Kempner D H, et al. Cell labelling and magnetic separation by means of immunoreagents based on polyacrolein microspheres. J Immunol Methods 1982; 52: 341-351.
    • Cahoy J D, Emery B, Kaushal A, et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 2008; 28:264-278
    • Miltenyi S, Muller W, Weichel W, et al. High gradient magnetic cell separation with MACS. Cytometry 1990; 11:231-238.
    • Topalian S L, Muul L M, Solomon D, et al. Expansion of human tumor infiltrating lymphocytes for use in immunotherapy trials. J Immunol Methods. 1987; 102(1):127-41.
    • Bonner W A, Sweet R G, Hulett H R, et al. Fluorescence activated cell sorting. Rev Sci lnstrum 1972; 43: 404-409
    • Gossett D R, Weaver W M, Mach A J. Et al., Label-free cell separation and sorting in microfluidic systems, Anal Bioanal Chem, 2010, 397, 3249-3267
    • Barbara Cunha B, Peixoto C, Silva M M, et al., Filtration methodologies for the clarification and concentration of human mesenchymal stem cells, J. of Membrane Sci., 2015, 478, 117-129
    • Klein A B, Witonsky S G, Ahmed S A, et al. Impact of different cell isolation techniques on lymphocyte viability and function. J Immunoassay Immunochem 2006; 27: 61-76
    • Steinberg M S. ‘ECM’: its nature, origin and function in cell aggregation. Exp Cell Res 1963; 30: 257-279.
    • Hefeneider S H, McCoy S L, Morton J I, et al. DNA binding to mouse cells is mediated by cell-surface molecules: the role of these DNA-binding molecules as target antigens in murine lupus. Lupus 1992; 1: 167-173.
    • Pisetsky D S and Fairhurst A-M. The origin of extracellular DNA during the clearance of dead and dying cells—review. Autoimmunity 2007; 40: 281-284
    • Renner W A, Jordan M, Eppenberger H M, et al. Cell-cell adhesion and aggregation: influence on the growth behaviour of CHO cells. Biotechnol Bioeng 1993; 41: 188-193
    • Shedlock D J, Aviles J, Talbott K T et al., Induction of Broad Cytotoxic T Cells by Protective DNA Vaccination Against Marburg and Ebola. Molecular Therapy, 2013; 21, 1432-1444
    • Baust J G, & Baust J M, Advances in Biopreservation, 2006, Chapt. 8, 157-196
    • Seglen, P. O., Preparation of Isolated Rat Liver Cells, Methods in Cell Biology, 1976; 13, 29
    • Quistorff, B., Dich, J., & Grunnet, N. Preparation of isolated rat liver hepatocytes. Methods in molecular biology, Chapt 14, 1990; 151-160.
    • Seifter, S., Gallop, P. M., Klein, L., et al. Studies on Collagen, Part II. Properties of Purified Collagenase and Its Inhibition. J. Biol. Chem. 1959; 234:285

Claims (34)

1: A single use aseptic kit comprising:
a disaggregation module for receipt and processing of material comprising solid mammalian tissue; and
a stabilisation module for further processing and/or storing disaggregated product material,
wherein each of said modules comprises one or more flexible containers connected by one or more conduits adapted to enable flow of the tissue material there between, and
wherein each of said modules comprises one or more ports to permit aseptic input of media and/or reagents into the one or more flexible containers,
wherein the kit further comprises a digital, electronic, or electromagnetic tag identifier relating to a specific program that defines a type of disaggregation, enrichment, and/or stabilisation process and use of one or more types of media in the process(es).
2: The single use aseptic kit of claim 1, wherein the one or more flexible containers comprise a resilient deformable material.
3: The single use aseptic kit of claim 1, wherein the one or more flexible containers of the disaggregation module comprises one or more sealable openings.
4: The single use aseptic kit of claim 3, wherein the flexible container of the disaggregation module comprises a heat sealable weld.
5: The single use aseptic kit of claim 1, wherein the one or more flexible containers comprises internally rounded edges.
6: The single use aseptic kit of claim 1, wherein the one or more flexible containers of the disaggregation module comprises disaggregation surfaces adapted to mechanically crush and shear the solid tissue therein.
7: The single use aseptic kit of claim 1, further comprising an enrichment module comprising one or more flexible containers, wherein the enrichment module is configured for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate and comprising, wherein the enrichment module is connected to the stabilisation and desegregation modules by one or more conduits adapted to enable flow of the tissue material, and wherein the one or more flexible containers of the enrichment module each comprise a filter that is configured to retain a retentate of cellularised disaggregated solid tissue.
8: The single use aseptic kit of claim 1, wherein the one or more flexible containers of the stabilisation module comprises media formulation for storage of viable cells in solution or in a cryopreserved state.
9. (canceled)
10. (canceled)
11: The single use aseptic kit of claim 7, wherein the same flexible container forms part of one or more of the disaggregation modules, stabilisation modules, and/or enrichment modules.
12: The single use aseptic kit of claim 11, wherein the disaggregation module comprises a first flexible container for receipt of the tissue to be processed.
13: The single use aseptic kit of claim 12, wherein the disaggregation module comprises a second flexible container comprising the media for disaggregation.
14: The single use aseptic kit of claim 13, wherein the enrichment module comprises a first flexible container and a third flexible container configured to receive enriched filtrate.
15: The single use aseptic kit of claim 14, wherein both the disaggregation module and the stabilisation module comprise a second flexible container and wherein the second container comprises digestion media and stabilisation media.
16: The single use aseptic kit of claim 15, wherein the stabilisation module comprises a fourth flexible container comprising stabilisation media.
17: The single use aseptic kit of claim 16, wherein the stabilisation module also comprises the first flexible container and/or the third flexible container for storing and/or undergoing cryopreservation.
18: Use of the single use aseptic kit according to claim 1 in a semi-automated process for the aseptic disaggregation, stabilisation, and enrichment of mammalian cells or cell aggregates.
19: An automated device for semi-automated aseptic disaggregation, enrichment, and/or stabilisation of cells or cell aggregates from mammalian solid tissue comprising:
a programmable processor; and
a radio-frequency identification tag reader,
wherein the programmable processor is configured to recognise a single-use aseptic kit in accordance with claim 1 using the tag reader and is adapted to subsequently execute a kit-specific program, wherein the program defines the type of disaggregation, enrichment, and/or stabilisation processes and the media type(s) required for use in the process(es).
20-21. (canceled)
22: The automated device of claim 19, wherein the programmable processor is adapted to communicate with and control one or more of:
the disaggregation module;
the enrichment module; and
the stabilisation module.
23: The automated device of claim 22, wherein the programmable processor controls the disaggregation module to enable a physical and/or biological breakdown of the solid tissue material.
24: The automated device of claim 23, wherein the programmable processor controls the disaggregation module to enable a physical and enzymatic breakdown of the solid tissue material.
25: The automated device of claim 24, wherein the enzymatic breakdown of the solid tissue material is by one or more media enzyme solutions selected from collagenase, trypsin, lipase, hyaluronidase, deoxyribonuclease, Liberase H1, pepsin, or mixtures thereof.
26: The automated device of claim 19, wherein the programmable processor controls disaggregation surfaces within the disaggregation flexible containers that mechanically crush and shear the solid tissue and wherein the disaggregation surfaces are mechanical pistons.
27: The automated device of claim 19, wherein the programmable processor controls the stabilisation module to cryopreserve the enriched disaggregated solid tissue in the container using a programmable temperature.
28: The automated device of claim 19 wherein the device further comprises one or more of the additional components in any combination:
sensors capable of recognising whether a disaggregation process has been completed in the disaggregation module prior to transfer of the disaggregated solid tissue to the enrichment module;
weight sensors configured to determine an amount of media required in the containers of one or more of the disaggregation module; the enrichment module; and/or the stabilisation module and to control the transfer of material between respective containers;
sensors to sense temperature within the containers of the one or more of the disaggregation module; the enrichment module; and/or
the stabilisation module;
at least one bubble sensor to control the transfer of media between the input and output ports of each container in the module;
at least one peristaltic pump to control the transfer of media between the input and output ports;
pressure sensors to assess the pressure within the enrichment module;
one or more valves to control a tangential flow filtration process within the enrichment module; and/or
one or more clamps to control the transfer of media between the input and output ports of each module.
29: The automated device of claim 19, wherein the programmable processor is adapted to maintain an optimal storage temperature range in the stabilisation module until the container is removed; or executes a controlled freezing step.
30: The automated device of claim 19, further comprising a user interface.
31: The automated device of claim 23, wherein the interface comprises a display screen to display instructions that guide a user to input parameters, confirm pre-programmed steps, warn of errors, or combinations thereof.
32: The automated device of claim 19, wherein the automated device is adapted to be transportable.
33: A semi-automatic aseptic tissue processing method comprising:
automatically determining aseptic disaggregation tissue processing steps and their associated conditions from a digital, electronic, or electromagnetic tag identifier associated with the aseptic processing kit of claim 1;
placing a tissue sample into a flexible plastic container of the disaggregation module of the aseptic processing kit; and
processing the tissue sample by automatically executing the one or more tissue processing steps by communicating with and controlling the disaggregation module; the optional enrichment module; and the stabilisation module.
34: The single use aseptic kit of claim 1, further comprising an enrichment module for filtration of disaggregated solid tissue material and segregation of non-disaggregated tissue and filtrate.
35: The single use aseptic kit of claim 1, wherein the specific program defines use of a freezing solution for controlled rate freezing in the stabilisation module.
US16/477,366 2017-01-13 2018-01-12 Aseptic tissue processing method, kit and device Abandoned US20200032197A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB1700621.4A GB201700621D0 (en) 2017-01-13 2017-01-13 Method,device and kit for the aseptic isolation,enrichment and stabilsation of cells from mammalian solid tissue
GB1700621.4 2017-01-13
PCT/GB2018/050088 WO2018130845A1 (en) 2017-01-13 2018-01-12 Aseptic tissue processing method, kit and device

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2018/050088 A-371-Of-International WO2018130845A1 (en) 2017-01-13 2018-01-12 Aseptic tissue processing method, kit and device

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17/826,062 Continuation US11618878B2 (en) 2017-01-13 2022-05-26 Aseptic tissue processing method, kit and device
US17/826,053 Continuation US11618877B2 (en) 2017-01-13 2022-05-26 Aseptic tissue processing method, kit and device

Publications (1)

Publication Number Publication Date
US20200032197A1 true US20200032197A1 (en) 2020-01-30

Family

ID=58463237

Family Applications (4)

Application Number Title Priority Date Filing Date
US16/477,366 Abandoned US20200032197A1 (en) 2017-01-13 2018-01-12 Aseptic tissue processing method, kit and device
US17/826,062 Active US11618878B2 (en) 2017-01-13 2022-05-26 Aseptic tissue processing method, kit and device
US17/826,053 Active US11618877B2 (en) 2017-01-13 2022-05-26 Aseptic tissue processing method, kit and device
US18/295,103 Pending US20230235269A1 (en) 2017-01-13 2023-04-03 Aseptic tissue processing method, kit and device

Family Applications After (3)

Application Number Title Priority Date Filing Date
US17/826,062 Active US11618878B2 (en) 2017-01-13 2022-05-26 Aseptic tissue processing method, kit and device
US17/826,053 Active US11618877B2 (en) 2017-01-13 2022-05-26 Aseptic tissue processing method, kit and device
US18/295,103 Pending US20230235269A1 (en) 2017-01-13 2023-04-03 Aseptic tissue processing method, kit and device

Country Status (4)

Country Link
US (4) US20200032197A1 (en)
EP (1) EP3568459A1 (en)
GB (1) GB201700621D0 (en)
WO (1) WO2018130845A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021214159A1 (en) * 2020-04-22 2021-10-28 Cellunite Gmbh System and method for automated cell processing
WO2022117665A1 (en) * 2020-12-01 2022-06-09 Cutiss Ag Device and method for preparing a cell suspension
WO2023001910A1 (en) 2021-07-23 2023-01-26 Cellunite Gmbh System and method for cell processing
US11618877B2 (en) 2017-01-13 2023-04-04 Instil Bio (Uk) Limited Aseptic tissue processing method, kit and device
US11767510B2 (en) 2019-12-20 2023-09-26 Instil Bio (Uk) Limited Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
GB2586567B (en) * 2019-03-01 2023-10-18 Asymptote Ltd Closed tissue disaggregation and cryopreservation
CN113508284A (en) * 2019-03-01 2021-10-15 阿西姆普托特有限公司 Closed tissue dissociation and cryopreservation
GB202017038D0 (en) 2020-10-27 2020-12-09 Global Life Sciences Solutions Operations UK Ltd Apparatus for tissue disaggregation
CA3205464A1 (en) 2020-12-18 2022-06-23 Instil Bio (Uk) Limited Processing of tumor infiltrating lymphocytes
CA3205462A1 (en) 2020-12-18 2022-06-23 Instil Bio (Uk) Limited Processing of tumor infiltrating lymphocytes
EP4262828A1 (en) 2020-12-18 2023-10-25 Instil Bio (Uk) Limited Tumor infiltrating lymphocytes and anti-cd47 therapeutics
CN112831399B (en) * 2021-02-24 2024-01-05 通用技术集团健康管理科技有限公司 Nucleic acid detection kit, kit bin and detection method for full-automatic chemiluminescence immunoassay analyzer in intelligent hospital

Family Cites Families (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69018246T2 (en) * 1990-02-09 1995-11-02 Univ Jefferson Device for collecting and processing fatty tissue for the production of endothelial cell product.
US5814295A (en) * 1992-04-10 1998-09-29 The Ohio State University Research Foundation Determination of lymph nodes enriched in tumor reactive cells their proliferation and their use in adoptive cellular therapy
SG47030A1 (en) 1994-01-03 1998-03-20 Genentech Inc Thrombopoietin
US5498599A (en) 1994-01-21 1996-03-12 Amgen Inc. Methods for stimulating platelet production
ES2301164T3 (en) 1994-12-02 2008-06-16 Schering Corporation SLAM, ANTIGEN COESTIMULATOR OF THE SURFACE OF CELLS T.
WO1999023199A1 (en) * 1997-10-31 1999-05-14 Baxter International Inc. Closed system for processing primary tissue
WO2002090489A1 (en) 2001-05-09 2002-11-14 Bayer Pharmaceuticals Corporation A cryopreservation bag assembly for mammalian cell lines
US7745140B2 (en) 2002-01-03 2010-06-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool
DK1545204T3 (en) 2002-09-06 2016-11-14 The Government Of The Us Secretary Dept Of Health And Human Services Immunotherapy with in vitro selected antigen-specific lymphocytes following non-myeloablative lymphodepletive chemotherapy
JP2007104975A (en) * 2005-10-14 2007-04-26 Toyo Seikan Kaisha Ltd Culture container and culture method
WO2008027739A2 (en) 2006-08-28 2008-03-06 Nuvelo, Inc. Antibodies to ntb-a
ECSP077628A (en) 2007-05-03 2008-12-30 Smithkline Beechman Corp NEW PHARMACEUTICAL COMPOSITION
WO2009032057A2 (en) 2007-08-29 2009-03-12 Adam Lubin Method for the selective therapy of disease
JP5917392B2 (en) 2009-05-01 2016-05-11 ビミニ テクノロジーズ リミテッド ライアビリティ カンパニー Systems, methods and compositions for optimizing tissue and cell-enriched grafts
US8383099B2 (en) 2009-08-28 2013-02-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Adoptive cell therapy with young T cells
US20130115617A1 (en) 2009-12-08 2013-05-09 John R. Wilson Methods of cell culture for adoptive cell therapy
CN102762719A (en) 2009-12-08 2012-10-31 威尔森沃尔夫制造公司 Improved methods of cell culture for adoptive cell therapy
US8956860B2 (en) 2009-12-08 2015-02-17 Juan F. Vera Methods of cell culture for adoptive cell therapy
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2012129201A1 (en) 2011-03-22 2012-09-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of growing tumor infiltrating lymphocytes in gas-permeable containers
PL2694549T3 (en) 2011-04-08 2019-01-31 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-epidermal growth factor receptor variant iii chimeric antigen receptors and use of same for the treatment of cancer
ES2725564T3 (en) * 2011-11-08 2019-09-24 Auxocell Laboratories Inc Systems and methods of cellular processing
MX2014010183A (en) 2012-02-22 2015-03-20 Univ Pennsylvania Compositions and methods for generating a persisting population of t cells useful for the treatment of cancer.
CA2872504A1 (en) 2012-05-18 2013-11-21 Wilson Wolf Manufacturing Corporation A static animal cell culture and cell recovery
IL288241B2 (en) 2012-06-11 2023-10-01 Wilson Wolf Mfg Corporation Improved methods of cell culture for adoptive cell therapy
US20140047572A1 (en) 2012-08-13 2014-02-13 University Of Rochester Thrombopoietin mimetics for the treatment of radiation or chemical induced bone marrow injury
US10117896B2 (en) 2012-10-05 2018-11-06 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
EP2931751B1 (en) 2012-12-12 2020-02-05 Arch Oncology, Inc. Therapeutic cd47 antibodies
CN105163745B (en) 2013-03-01 2020-06-12 美国卫生和人力服务部 Method for generating enriched populations of tumor-reactive T cells from peripheral blood
AU2013205148B2 (en) 2013-03-14 2014-10-30 AVITA Medical Americas, LLC Systems and methods for tissue processing and preparation of cell suspension therefrom
EP4137564A1 (en) 2013-07-15 2023-02-22 The United States of America, as represented by the Secretary, Department of Health and Human Services Methods of preparing anti-human papillomavirus antigen t cells
CA2926856A1 (en) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anti-pd-l1 monoclonal antibodies and fragments thereof
EP3102609A4 (en) 2014-02-04 2017-08-23 The United States of America, as represented by The Secretary, Department of Health and Human Services Methods for producing autologous t cells useful to treat b cell malignancies and other cancers and compositions thereof
AU2015218396A1 (en) 2014-02-14 2016-08-11 Bellicum Pharmaceuticals, Inc. Methods for activating T cells using an inducible chimeric polypeptide
CN106068124A (en) * 2014-02-28 2016-11-02 Bdbc科学公司 A kind of system for tissue manipulation
WO2015184228A1 (en) 2014-05-29 2015-12-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-human papillomavirus 16 e7 t cell receptors
EP3155426B1 (en) 2014-06-13 2023-07-19 Immudex ApS General detection and isolation of specific cells by binding of labeled molecules
WO2016007841A1 (en) * 2014-07-10 2016-01-14 The Trustees Of Columbia University In The City Of New York Ex vivo browning of adipose tissue therapy for reversal of obesity and type ii diabetes
JP6918703B2 (en) 2015-04-27 2021-08-11 キャンサー・リサーチ・テクノロジー・リミテッド How to treat cancer
WO2016179006A1 (en) 2015-05-01 2016-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of isolating t cells and t cell receptors having antigenic specificity for a cancer-specific mutation from peripheral blood
MA42902A (en) 2015-07-08 2018-05-16 Univ Johns Hopkins MARINAL-INFILTRATING LYMPHOCYTES (MIL) AS A SOURCE OF T-LYMPHOCYTES FOR CHEMERIC ANTIGEN RECEPTOR (CAR) THERAPY
WO2017023803A1 (en) 2015-07-31 2017-02-09 Regents Of The University Of Minnesota Modified cells and methods of therapy
CN108350059B (en) 2015-09-15 2021-10-08 美国卫生和人力服务部 T cell receptor recognizing HLA-CW8 restriction mutant KRAS
US9650441B2 (en) 2015-09-21 2017-05-16 Erasmus University Medical Center Anti-CD47 antibodies and methods of use
WO2017070395A1 (en) 2015-10-20 2017-04-27 Kite Pharma, Inc. Methods of preparing t cells for t cell therapy
WO2017087784A1 (en) * 2015-11-18 2017-05-26 Duke University Tumor infiltrating lymphocytes for treatment of cancer
GB201522097D0 (en) 2015-12-15 2016-01-27 Cellular Therapeutics Ltd Cells
WO2017179015A1 (en) 2016-04-15 2017-10-19 Glaxosmithkline Intellectual Property Development Limited Compositions for the treatment of cancer
SG10202006117QA (en) 2016-04-21 2020-08-28 Immatics Biotechnologies Gmbh Immunotherapy against melanoma and other cancers
CN109843915B (en) 2016-05-06 2023-03-03 朱诺治疗学股份有限公司 Genetically engineered cell and preparation method thereof
MX2019000149A (en) 2016-07-07 2019-09-23 Iovance Biotherapeutics Inc Programmed death 1 ligand 1 (pd-l1) binding proteins and methods of use thereof.
EP3484927A1 (en) 2016-07-15 2019-05-22 Poseida Therapeutics, Inc. Chimeric antigen receptors (cars) specific for muc1 and methods for their use
EP3494133B1 (en) 2016-08-02 2022-07-06 The U.S.A. as represented by the Secretary, Department of Health and Human Services Anti-kras-g12d t cell receptors
CN110520530A (en) 2016-10-18 2019-11-29 明尼苏达大学董事会 Tumor infiltrating lymphocyte and treatment method
US11026974B2 (en) 2016-10-26 2021-06-08 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
CN110199016A (en) 2016-11-17 2019-09-03 艾欧凡斯生物治疗公司 Tumors remaining lymphocyte infiltration and its preparation and application
WO2018129332A1 (en) 2017-01-06 2018-07-12 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes (tils) with tumor necrosis factor receptor superfamily (tnfrsf) agonists and therapeutic combinations of tils and tnfrsf agonists
EP3565586A1 (en) 2017-01-06 2019-11-13 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof
GB201700621D0 (en) 2017-01-13 2017-03-01 Guest Ryan Dominic Method,device and kit for the aseptic isolation,enrichment and stabilsation of cells from mammalian solid tissue
JOP20190224A1 (en) 2017-03-29 2019-09-26 Iovance Biotherapeutics Inc Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11254913B1 (en) 2017-03-29 2022-02-22 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
CN110612446B (en) 2017-03-31 2023-07-04 美国卫生和人力服务部 Method for isolating novel antigen-specific T cell receptor sequences
WO2018209115A1 (en) 2017-05-10 2018-11-15 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
WO2018226714A1 (en) 2017-06-05 2018-12-13 Iovance Biotherapeutics, Inc. Methods of using tumor infiltrating lymphocytes in double-refractory melanoma
MX2020003117A (en) 2017-09-20 2020-08-20 Us Health HLA CLASS II–RESTRICTED T CELL RECEPTORS AGAINST MUTATED RAS.
EP3688142A1 (en) 2017-09-29 2020-08-05 The USA, as represented by The Secretary, Department of Health and Human Services Methods of isolating t cells having antigenic specificity for a p53 cancer-specific mutation
BR112020009663A2 (en) 2017-11-17 2020-11-10 Iovance Biotherapeutics, Inc. method for the expansion of tumor infiltrating lymphocytes (tils) in a therapeutic population of tils, method for the treatment of an individual with cancer, composition
JP2021503885A (en) 2017-11-22 2021-02-15 アイオバンス バイオセラピューティクス,インコーポレイテッド Expanded culture of peripheral blood lymphocytes (PBL) from peripheral blood
US20210369775A1 (en) 2017-12-15 2021-12-02 Iovance Biotherapeutics, Inc. Systems and methods for determining the beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof and beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof
WO2019136459A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
BR112020013848A2 (en) 2018-01-08 2020-12-01 Iovance Biotherapeutics, Inc. methods for expanding tumor-infiltrating lymphocytes and for treating an individual with cancer, tumor-infiltrating lymphocyte population, and, method for evaluating transcription factors
GB201801067D0 (en) 2018-01-23 2018-03-07 Price Nicola Kaye Biomarkers predictive of tumour infiltrating lymphocyte therapy and uses thereof
US10739728B2 (en) 2018-01-29 2020-08-11 Massachusetts Institute Of Technology Molecular clock
AU2019218093A1 (en) 2018-02-09 2020-09-10 Immatics US, Inc. Methods for manufacturing T cells
US20210052647A1 (en) 2018-02-09 2021-02-25 The United States Of America,As Represented By The Secretary,Department Of Health And Human Services Tethered interleukin-15 and interleukin-21
JP2021512962A (en) 2018-02-13 2021-05-20 アイオバンス バイオセラピューティクス,インコーポレイテッド Expansion culture of tumor-infiltrating lymphocytes (TIL) with adenosine A2A receptor antagonist and therapeutic combination of TIL and adenosine A2A receptor antagonist
IT201800003533A1 (en) 2018-03-14 2019-09-14 Industrie De Nora Spa ELECTRODE FOR ELECTROCHLORATION PROCESSES
EA202092319A1 (en) 2018-03-29 2021-03-04 Айовэнс Байотерапьютикс, Инк. METHODS FOR OBTAINING TUMOR-INFILTING LYMPHOCYTES AND USING THEM IN IMMUNOTHERAPY
SG11202010319RA (en) 2018-04-27 2020-11-27 Iovance Biotherapeutics Inc Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
US20220118011A1 (en) 2018-05-10 2022-04-21 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2019217753A1 (en) 2018-05-10 2019-11-14 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
GB201810181D0 (en) 2018-06-21 2018-08-08 Immetacyte Ltd Cells expressing chimeric recominant growth factor receptors
TW202031273A (en) 2018-08-31 2020-09-01 美商艾歐凡斯生物治療公司 Treatment of nsclc patients refractory for anti-pd-1 antibody
EP3852524B1 (en) 2018-09-20 2023-06-28 Iovance Biotherapeutics, Inc. Expansion of tils from cryopreserved tumor samples
MX2021004775A (en) 2018-11-05 2021-06-08 Iovance Biotherapeutics Inc Expansion of tils utilizing akt pathway inhibitors.
EP3877513A2 (en) 2018-11-05 2021-09-15 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
TW202039829A (en) 2018-11-05 2020-11-01 美商艾歐凡斯生物治療公司 Selection of improved tumor reactive t-cells
BR112021008549A2 (en) 2018-11-05 2022-01-04 Iovance Biotherapeutics Inc Method of treating non-small cell lung cancer with a population of tumor-infiltrating lymphocytes
AU2019393243A1 (en) 2018-12-07 2021-07-01 Gracell Biotechnologies (Shanghai) Co., Ltd. Compositions and methods for immunotherapy
US20220193131A1 (en) 2018-12-19 2022-06-23 Iovance Biotherapeutics, Inc. Methods of Expanding Tumor Infiltrating Lymphocytes Using Engineered Cytokine Receptor Pairs and Uses Thereof
EP3908678A1 (en) 2019-01-10 2021-11-17 Iovance Biotherapeutics, Inc. System and methods for monitoring adoptive cell therapy clonality and persistence
GB201900858D0 (en) 2019-01-22 2019-03-13 Price Nicola Kaye Receptors providing targeted costimulation for adoptive cell therapy
CN113508284A (en) 2019-03-01 2021-10-15 阿西姆普托特有限公司 Closed tissue dissociation and cryopreservation
US20220133795A1 (en) 2019-03-01 2022-05-05 Iovance Biotherapeutics, Inc. Expansion of Tumor Infiltrating Lymphocytes From Liquid Tumors and Therapeutic Uses Thereof
WO2020232029A1 (en) 2019-05-13 2020-11-19 Iovance Biotherapeutics, Inc. Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy
GB201910605D0 (en) 2019-07-24 2019-09-04 Immetacyte Ltd Tumour infltracting lymphocyte therapy amd uses thereo
CA3155727A1 (en) 2019-10-25 2021-04-29 Cecile Chartier-Courtaud Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
JP2023506734A (en) 2019-12-11 2023-02-20 アイオバンス バイオセラピューティクス,インコーポレイテッド Process for the production of tumor-infiltrating lymphocytes (TIL) and methods of using same
FR3105367A1 (en) 2019-12-20 2021-06-25 Valeo Systemes Thermiques VENTILATION DEVICE MODULE FOR MOTOR VEHICLE COOLING MODULE, VENTILATION DEVICE INCLUDING SUCH MODULE AND COOLING MODULE FOR MOTOR VEHICLE INCLUDING SUCH VENTILATION DEVICE
AU2020408017A1 (en) 2019-12-20 2022-07-14 Instil Bio (Uk) Limited Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof
US11403088B2 (en) 2020-03-06 2022-08-02 Hewlett Packard Enterprise Development Lp Upgrade of hosts hosting application units of a container-based application based on analysis of the historical workload pattern of the cluster
MX2022013065A (en) 2020-04-22 2022-12-08 Iovance Biotherapeutics Inc Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy.
EP4146794A1 (en) 2020-05-04 2023-03-15 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
JP2023524108A (en) 2020-05-04 2023-06-08 アイオバンス バイオセラピューティクス,インコーポレイテッド Selection of improved tumor-reactive T cells
WO2022010017A1 (en) 2020-07-10 2022-01-13 (주)에프에이 Specimen culture medium composition applicable to long-term storage and universal diagnostic method, and specimen transport medium applying same
JP2023534808A (en) 2020-07-17 2023-08-14 インスティル バイオ (ユーケイ) リミテッド Receptors that provide targeted co-stimulation for adoptive cell therapy
US20230277670A1 (en) 2020-07-17 2023-09-07 John Bridgeman Chimeric molecules providing targeted costimulation for adoptive cell therapy
US20230372397A1 (en) 2020-10-06 2023-11-23 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2022087324A1 (en) 2020-10-22 2022-04-28 Iovance Biotherapeutics, Inc. Cell culture system and methods of using the same
JP2024501452A (en) 2020-12-11 2024-01-12 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of cancer patients with tumor-infiltrating lymphocyte therapy in combination with BRAF inhibitors and/or MEK inhibitors
US20240123067A1 (en) 2020-12-17 2024-04-18 Iovance Biotherapeutics, Inc. Treatment of cancers with tumor infiltrating lymphocyte therapies
WO2022133140A1 (en) 2020-12-17 2022-06-23 Iovance Biotherapeutics, Inc. Treatment with tumor infiltrating lymphocyte therapies in combination with ctla-4 and pd-1 inhibitors
CA3205464A1 (en) 2020-12-18 2022-06-23 Instil Bio (Uk) Limited Processing of tumor infiltrating lymphocytes
CA3205462A1 (en) 2020-12-18 2022-06-23 Instil Bio (Uk) Limited Processing of tumor infiltrating lymphocytes
EP4262828A1 (en) 2020-12-18 2023-10-25 Instil Bio (Uk) Limited Tumor infiltrating lymphocytes and anti-cd47 therapeutics
US20240110152A1 (en) 2020-12-31 2024-04-04 Iovance Biotherapeutics, Inc. Devices and processes for automated production of tumor infiltrating lymphocytes
JP2024506557A (en) 2021-01-29 2024-02-14 アイオバンス バイオセラピューティクス,インコーポレイテッド Methods of producing modified tumor-infiltrating lymphocytes and their use in adoptive cell therapy
CA3207359A1 (en) 2021-02-05 2022-08-11 Cecile Chartier-Courtaud Adjuvant therapy for cancer
CN117279506A (en) 2021-03-05 2023-12-22 艾欧凡斯生物治疗公司 Tumor storage and cell culture compositions
WO2022198141A1 (en) 2021-03-19 2022-09-22 Iovance Biotherapeutics, Inc. Methods for tumor infiltrating lymphocyte (til) expansion related to cd39/cd69 selection and gene knockout in tils
AR125199A1 (en) 2021-03-23 2023-06-21 Iovance Biotherapeutics Inc CISH GENE EDITION OF TUMOR-INFILTRATING LYMPHOCYTES AND THEIR USES IN IMMUNOTHERAPY
KR20240032711A (en) 2021-03-25 2024-03-12 이오반스 바이오테라퓨틱스, 인크. Methods and compositions for use in T-cell co-culture efficacy assays and cell therapeutics

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11618877B2 (en) 2017-01-13 2023-04-04 Instil Bio (Uk) Limited Aseptic tissue processing method, kit and device
US11618878B2 (en) 2017-01-13 2023-04-04 Instil Bio (Uk) Limited Aseptic tissue processing method, kit and device
US11767510B2 (en) 2019-12-20 2023-09-26 Instil Bio (Uk) Limited Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof
WO2021214159A1 (en) * 2020-04-22 2021-10-28 Cellunite Gmbh System and method for automated cell processing
WO2022117665A1 (en) * 2020-12-01 2022-06-09 Cutiss Ag Device and method for preparing a cell suspension
WO2023001910A1 (en) 2021-07-23 2023-01-26 Cellunite Gmbh System and method for cell processing

Also Published As

Publication number Publication date
US20220364042A1 (en) 2022-11-17
WO2018130845A1 (en) 2018-07-19
US11618877B2 (en) 2023-04-04
US20230235269A1 (en) 2023-07-27
US11618878B2 (en) 2023-04-04
GB201700621D0 (en) 2017-03-01
EP3568459A1 (en) 2019-11-20
US20220290094A1 (en) 2022-09-15

Similar Documents

Publication Publication Date Title
US11618878B2 (en) Aseptic tissue processing method, kit and device
US10131876B2 (en) Method for automated generation of genetically modified T cells
US20220348874A1 (en) Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof
Petroff et al. Isolation and culture of term human trophoblast cells
JP5899246B2 (en) Stem cell packaging product and method for producing and transporting the packaged product
Roda et al. A novel stem cell tag-less sorting method
US11561219B2 (en) Methods of enumerating particles present in a cell composition
CN112126618B (en) Method for obtaining human gallbladder stem cells and long-term in vitro culture
Yuan et al. Extracellular vesicle collection from human stem cells grown in suspension bioreactors
WO2020161748A1 (en) Method for mesenchymal stem cell isolation and osteoblast differentiation
Shiojiri et al. Purification and culture of fetal mouse hepatoblasts that are precursors of mature hepatocytes and biliary epithelial cells
Laoui et al. Purification of Tumor-Associated Macrophages (TAM) and Tumor-Associated Dendritic Cells (TADC)
Plebanski Preparation of lymphocytes and identification of lymphocyte subpopulations
Sharma et al. Cell Culture: Recent Advance and Application
Tao et al. Establishing a method for the expansion of human regulatory T cells with a high-efficiency in vitro
TW202136502A (en) Devices and methods for isolating tumor infiltrating lymphocytes and uses thereof
TW202317756A (en) Methods of isolating of tumor infiltrating lymphocytes and use thereof
CN115491769A (en) Method for preparing feeder layer cell bank
TW202310857A (en) Processing of tumor infiltrating lymphocytes
Lowdell et al. Processing of cells for transplantation
Przyborski DERIVATION AND CULTURE OF HUMAN EM BRYONAL CARCINOMA STEM CELL LINES
McKenna Jr et al. Haemopoietic stem cell processing and storage
JP2011055717A (en) Cell culture method and cell culture device
CN104419724A (en) T lymphocyte model constructed by virtue of SV40LT gene inducing and cell tank thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: IMMETACYTE LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GUEST, RYAN DOMINIC;PRICE, NICOLA KAYE;REEL/FRAME:049780/0670

Effective date: 20190711

AS Assignment

Owner name: INSTIL BIO (UK) LIMITED, UNITED KINGDOM

Free format text: CHANGE OF NAME;ASSIGNOR:IMMETACYTE LIMITED;REEL/FRAME:053125/0416

Effective date: 20200702

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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