WO2013112936A1 - Implantable devices and applications and use thereof - Google Patents

Implantable devices and applications and use thereof Download PDF

Info

Publication number
WO2013112936A1
WO2013112936A1 PCT/US2013/023296 US2013023296W WO2013112936A1 WO 2013112936 A1 WO2013112936 A1 WO 2013112936A1 US 2013023296 W US2013023296 W US 2013023296W WO 2013112936 A1 WO2013112936 A1 WO 2013112936A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
implantable device
sub
compartments
compartment
Prior art date
Application number
PCT/US2013/023296
Other languages
French (fr)
Inventor
Heather Lynn HEINE
Original Assignee
Heine Heather Lynn
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 Heine Heather Lynn filed Critical Heine Heather Lynn
Publication of WO2013112936A1 publication Critical patent/WO2013112936A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/022Artificial gland structures using bioreactors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/64Animal cells

Definitions

  • implantable devices that can maintain the growth, development and differentiation of mammalian cells within an implantable device in a live mammal under a vascularized environment.
  • implantable devices for various biochemical and therapeutic applications, such live imaging, live biological and biochemical assays, drug testing, tissue/organ implantation, and etc.
  • integrated systems using such implantable devices for data acquisition e.g., images, videos and biochemical responses.
  • a system for maintaining mammalian cells in a live mammal e.g., for growth, development and/or differentiation.
  • An exemplary system comprises: i) an implantable device and ii) a plurality of cells contained within a compartment of the implantable device, where the plurality of cells are suspended in a biochemical composition comprising a cellular matrix, wherein the maintenance of the plurality of cells is supported partially by the biochemical composition.
  • the implantable device in turn comprises: a housing having a plurality of sides, wherein one side of the plurality of sides comprises a structural feature for securely attaching the implantable device to a site of implantation in the mammal; a compartment contained within the housing, once implanted in the mammal, the compartment is capable of exchanging content, via an interaction module on at least one side of the plurality of sides, with an extracellular space or a tissue microvasculature of the live mammal; and an observation module, which is located on one side of the plurality of sides through which the plurality of cells can be observed by an optical equipment.
  • one or more growth factors are also included in the biochemical composition.
  • the structural feature for securely attaching the implantable device is a groove on one side of the plurality of sides of the implantable device.
  • the structural feature for securely attaching the implantable device is a plurality of holes on at least one side of the plurality of sides of the implantable device.
  • the implantable device further comprises a plurality of sub-compartments within the compartment, and the plurality of cells is contained within at least one sub-compartment of the plurality of sub-compartments.
  • sub- compartments within the plurality of sub-compartments are connected via one or more channels, and where at least one of the connected sub-compartment contains the plurality of cells.
  • the connected sub-compartments upon exposure to one or more biochemical reagents, are configured to perform an assay on the plurality of cells, or blood or plasma products coming in contact with the device.
  • the assay detects the presence or absence of a biomarker, and wherein the biomarker is selected from the group consisting of a protein, a peptide, a gene, and a nucleic acid molecule.
  • the implantable device further comprises an injection port on one side of the plurality of sides, through which biochemical reagents can be added into or removed from the compartment.
  • the injection port and observation module are on the same side of the plurality of sides.
  • the interaction module comprises a permeable membrane between an opening on the at least one side of the plurality of sides of the implantable device.
  • the interaction module is an opening on the implantable device, which allows content within the device to exchange with the cellular environment where the implantable device is implanted.
  • the implantable device further comprises a wireless module for transmitting a signal from the implantable device to an external receiver or receiving a signal from an external controller, after the implantable device is implanted in a mammal.
  • the wireless module is for transmitting a signal from the implantable device to an external receiver and receiving a signal from an external controller.
  • an imaging system for collecting data from a live mammal.
  • An exemplary imaging system comprises 1) any one or combination of the system for maintaining growth and development of mammalian cells in a live mammal, as described herein; and 2) optical equipment for collecting images or video data of the content with the compartment or sub-compartments of the implantable device.
  • the optical equipment is selected from: a microscope, a fluorescence imaging device, a thermal imaging device, a radio imaging device, or a combination thereof.
  • the imaging system further comprises: a computer system for collecting, storing, displaying, and processing the image or video data.
  • the data collected comprise images or videos of the plurality of cells in the implantable device.
  • a method for collecting data from a live mammal using an imaging system comprises the steps of: i) maintaining the growth of a plurality of cells in a live mammal using a mammalian cell system as described herein, where the implantable device is implanted in a live mammal such that at least a portion of the observation module is not embedded within the live mammal and wherein data of the content within the implantable device are measured using optical equipment; and ii) collecting data of the content within the implantable device using optical equipment.
  • the optical equipment is selected from: a microscope, a fluorescence imaging device, a thermal imaging device, a radio imaging device, or a combination thereof.
  • the imaging system further comprises: a computer system for collecting, storing, displaying, and processing the image or video data.
  • the data collected comprise images or videos of the plurality of cells in the implantable device.
  • An exemplary system comprises a mammalian cell system as described herein that comprises a plurality of sub-compartments where each sub-compartment of the plurality of sub-compartments comprises a population of cells.
  • the cells are taken from a patient and a compound is added to one or more sub-compartments of the plurality of sub-compartments.
  • a method for drug testing using a mammalian cell system as described herein that comprises a plurality of sub-compartments comprises the steps of: placing, in each sub-compartment of the plurality of sub-compartments of the implantable device, a population of cells, wherein the cells are derived from a biopsy sample of a target disease, and wherein the implantable device is implanted in a live mammal; adding a test compound in one or more sub-compartments of the plurality of sub -compartments; and collecting data from each sub-compartment of the plurality of sub-compartments.
  • the method further comprises the steps of:
  • each sub-compartment of the plurality of sub- compartments contains the same type of cells and wherein cells in each sub-compartment have approximately the same quantity.
  • at least two different compounds are added to two different sub-compartments.
  • a method for producing a desired biomolecule in a live mammal using a mammalian cell system comprises the steps of: maintaining the growth of the plurality of cells in the live mammal using the implantable device, wherein the implantable device is implanted in a live mammal and wherein cells of the plurality of cells are capable of producing the desired biomolecule; adding one or more reagents to the implantable device, wherein the reagents are necessary for the plurality of cells to produce the desired biomolecule; and producing the desired biomolecule.
  • the cells are pancreas cells and the desired biomolecule is insulin.
  • An exemplary method comprises the steps of: maintaining the growth of the plurality of cells in the live mammal using the implantable device, wherein the implantable device is implanted in a live mammal such that at least a portion of the observation module is not embedded within the live mammal and wherein data of the content within the implantable device are measured using an optical equipment, and wherein cells of the plurality of cells are selected from the group consisting of stem cells, embryonic stem cells, adult stem cells and non-stem cells; measuring, via the observation module, data of the cells of the plurality of cells to determine whether the cells are suitable for transplant; removing the cells from the implantable device if the cells are suitable for transplant; and transplanting the cells in a mammal.
  • Figures 1A-1D illustrate exemplary embodiments; A) a round shape device, B) a triangular shape device, C) a fan shape device, and D) a convex curve shape device.
  • Figures 2A-2C illustrate different views of an exemplary embodiment with multiple sub-compartments.
  • Figure 3 illustrates a lab-on-a-chip design of implantable design with a miniaturized handling system.
  • Figure 4 illustrates a cylinder holding system for an exemplary implantable device.
  • Figure 5 illustrates an injection port system of an exemplary implantable device.
  • Figure 6 illustrates functional sub-compartments within an exemplary implantable device.
  • Figure 7 illustrates an exemplary tool for implantation.
  • Figure 8 illustrates an exemplary personalized cancer/tissue assay.
  • Figures 9A-9C illustrate an exemplary implantable device.
  • Figures 10A andlOB illustrate mice implanted with exemplary implantable device and control mice without the implant, A) Implant within two mice and non-implanted cagemate.
  • Figures 11A-11D illustrate an exemplary embodiment of live imaging system.
  • Figures 12A through 12E illustrate exemplary cell growth within implantable devices.
  • Figures 13A through 13F illustrate exemplary implantable devices.
  • Figure 13G depicts exemplary laser rings and baseplate template for creating lasercut acrylic device.
  • Figures 14A through 14B illustrate exemplary data from implantation study showing cell growth but no evidence of blood vessels.
  • Figures 15A through 15F illustrate exemplary data from implantation study showing blood vessels.
  • the term "implantable device” refers to an article with a main body or housing, which contains at least one structural element (e.g., compartment) that can be used to grow mammalian cells and conduct biochemical reactions while the device is implanted in a live mammal. It is sometimes used interchangeably with “implantable bioreactor.” Additional structural elements such as sub-compartments can be found in the compartment.
  • structural element e.g., compartment
  • biocompatible refers to a property of a material, which is characterized by it, or its physiological degradation products, being not, or at least minimally, toxic to living tissue; not, or at least minimally and reparably, otherwise injurious living tissue; and/or not, or at least minimally and controllably, causative of an immunological reaction in living tissue.
  • biocompatible material refers to materials, natural or synthetic, that can be used to construct an implantable device.
  • the materials forming the implantable device do not cause serious discomfort or adverse physiological reactions (such as adverse immuno response or rejection) of the mammal.
  • biodegradation refers to any means by which a polymer can be disposed of in a live mammal (e.g., in a patient's body), which includes bioabsorption, resorption, etc. Degradation occurs through hydrolysis, chemical reactions, or enzymatic reactions. Biodegradation can take place over an extended period of time, for example over 2-3 years.
  • biostable means that the polymer does not biodegrade or bioabsorb under physiological conditions, or biodegrade or bioabsorb very slowly over a very long period of time, for example, over 5 years or over 10 years.
  • compartment and “sub-compartment” refer to a structure in an implantable device that is separated from the external environment or other structures within the device. Examples of compartments and sub-compartments include chambers, channels, wells and etc.
  • biochemical composition refers to a mixture of small molecules and macromolecules within a compartment or sub-compartment of an implantable device, which can support the growth, development, and/or differentiation of mammalian cells.
  • exemplary components of a biochemical composition include but are not limited to growth factors, cellular matrices, and buffers.
  • biomarker refers to a substance used as an indicator of a biological state.
  • a biomarker can be a small molecule as well as a
  • a biomarker is a term often used to refer to a protein measured in blood whose concentration reflects the severity or presence of some disease state. More generally, a biomarker is anything that can be used as an indicator of a particular disease state or some other physiological state of an organism.
  • a biomarker is a molecule that is present or absent from a particular cell type. Biomarkers facilitate the characterization of a cell type, their identification, and eventually their isolation. A biomarker can also be used to identify a cell population, make a diagnosis, or measure the progress of a disease or the effects of a treatment. In some embodiments, the terms "biomarker” and “biomolecules” are used interchangeably.
  • optical equipment refers to an apparatus or device that has one or more sensors for collecting one or more optical signals.
  • the optical signals can be of any wavelength, visible or invisible.
  • implantable devices useful for maintaining cell growth, development and differentiation within a live mammal under a vascularized environment. Also provided herein are methods for designing and making the implantable devices.
  • an implantable device comprises a main body or housing containing at least one structural element (e.g., a compartment or sub-compartment) surrounded by one or more sides.
  • a structural element e.g., a compartment or sub-compartment
  • Exemplary embodiments of the implantable devices are depicted in Figures lA-1 ID.
  • the main body or housing provides the shape and support for the structural elements therein.
  • one or more functional or structural modules can be attached to the main body or housing of the implantable device.
  • the main body or housing of the implantable device is formed by a plurality of sides, including, for example, a top side and a bottom side.
  • device 100 has a top side-element 10 and a bottom side-element 20, which are joined through a cylindrical or tubular connecting side-element 15.
  • Compartment 30 here refers to the space formed by the top side-element 10, the bottom side-element 20, and the cylindrical or tubular connecting side-element 15.
  • the compartment is formed by sides of the main body or housing.
  • An interaction module 40 which allows the content within implantable device 100 to interact with the external environment when the device is implanted in a mammal, such as a rodent or a human.
  • a mammal such as a rodent or a human.
  • the top side and bottom side are round.
  • the connecting side element has a tubular or cylindrical shape, similar to that of the side of a barrel. It will be understood that the sides can be of any sizes and shapes, forming any shape that is suitable for the intended purpose of a particular implantable device.
  • the shape of a compartment is sometimes defined by the shape of a cross-section of compartment 30.
  • a cross section can be taken along a plane determined by the x-x' and y-y' axes, which is between the top side-element 10 and bottom side-element 20, as illustrated in Figure 1 A.
  • the resulting plane can be round, oval, semicircular, square, rectangle, triangle or any other shapes.
  • Various exemplary embodiments of device 100 are shown in Figures 1A-1D.
  • the compartment is defined by the main body or housing itself.
  • a separate structural device such as a pre-made insert can be used to form the compartment, thus allowing the compartment to have a shape that is different from that of the main body or housing.
  • the main body of an implantable device has a tubular or cylindrical shape, with a round cross-section (e.g., Figure 1 A).
  • compartments or sub-compartments within a round device can adopt different shapes such as oval, semi-circular, square, rectangle, triangle and etc.
  • round implantable devices are suitable for concentric cell growth inwards from all directions so the greatest volume of growth can occur at one time. Also advantageously, such implantable devices are compatible with implantation on or into skin. Once inserted into the skin, the skin around the circumference of the cylindrical or tubular device can be easily closed to reduce or eliminate irritation to implanted subject.
  • the main body of an implantable device is box-like with a square or rectangular cross-section.
  • compartments or sub- compartments within the box-like device also have square or rectangular cross-sections.
  • multiple sub-compartments of the same or similar shapes but smaller in size can be created within a square or rectangular compartment.
  • more quantifiable measurements can be taken between different individual implants.
  • more quantifiable measurements can be taken between sub-compartments within the same device. Further advantageously, the measurements of sub-compartments within the same device can be compared relatively easily to each other to assess
  • compartments having a square or rectangular cross-section are more suitable for data observation.
  • a rectangular cross-section corresponds to a longer distance in the 'field of view' than those of other shapes. As tissue grows, it can be studied along the longer distance of the rectangle under the coverslip.
  • the main body of an implantable device is a triangular-shaped block, with a triangular cross-section (e.g., Figure IB).
  • a triangular cross-section e.g., Figure IB
  • cell growth can start at one corner and move towards a broad face.
  • cell growth can start at a broad face and move towards a corner.
  • An exemplary embodiment is depicted in Figure IB.
  • such embodiments can be used to study the impact of increasing tissue density on tissue performance and growth.
  • they also can be used to examine tissue in response to varying concentrations of growth factors.
  • a cellular matrix containing a particular growth factor is placed at a corner, there can be a gradient of concentrations away from the corner.
  • such embodiments can be used to examine the reaction of tissues as they grow away from a corner that contains high concentration to an area of low concentration.
  • the main body of an implantable device is an irregular block, with a fan-shaped cross-section.
  • the fan-shaped compartment is triangle-like with a curved side.
  • the curved side is opposite to a growth corner and there are multiple parallel compartments.
  • such embodiments can be used to examine tissues growing from a single origin in response to different concentrations or types of stimuli that have been put at the end of the radiating wells.
  • the main body or housing of an implantable device and compartments or sub-compartments therein have a cross-section with convex curves (Figure ID).
  • the convex curves are positioned during implantation along the body of a mammal, such as around its back or abdomen (e.g., Figure ID).
  • such embodiments can better 'fit' the natural contours of the body when implanted.
  • an implantable device and the compartments or sub-compartments therein are custom-molded to fit the location of a desired implantation.
  • an implantable device and the compartment therein can be designed to fit a side of the liver or pancreas of a recipient.
  • the compartments and sub- compartments do not need to adopt the shape of the main body or housing of the implantable device.
  • a cylindrical or tubular implantable device can in fact house a compartment of rectangular, square or even triangular shape.
  • custom-molded implantable devices can better fit the natural contours of the body when implanted. Also advantageously, it is possible to attach the implantable device near a target organ. In such embodiments, pre-implantation data and planning are needed, for example, using CT scanning to analyze the morphology of the target organ, before the customized implantable devices are designed and produced.
  • sub-compartments within the same compartment have the same or a similar shape as well as functionalities (see, for example, Figures 2A-2C).
  • more quantifiable measurements can be taken between different individual implants.
  • more quantifiable measurements can be taken between sub-compartments within the same implant.
  • measurements of the sub-compartments within the same device can be compared relatively easily to each other for assessing characteristics such as the distance of growth of cell populations and complexity of tissue formation along the gradient of growth.
  • sub-compartments within the same compartment have the same shape but different functions (e.g., Figure 3).
  • sub- compartments within the same compartment have different shapes and different functions.
  • some sub-compartments can be used for storing biochemical reagents (e.g., growth factor, buffer, serum, or commercially available growth media such as MatrigelTM from Becton, Dickinson and Company etc.); they accordingly have relatively large volumes and are also called wells or storage depots.
  • some sub-compartments are used as a container where a biochemical reaction or synthesis takes place. Such sub- compartments are also called chambers or reaction chambers.
  • an implantable device can have one or more structural elements that are used for delivering biochemical reagents between sub-compartments. Such structures are also called channels.
  • one or more wells, chambers, storage depots and channels form a network of sub-compartments, which can be used to achieve sophisticated biochemical synthesis or to carry out a specific biochemical reaction.
  • the shape of the main body or housing of an implantable device can be modified or altered to facilitate secure attachment between the device and the site of implantation.
  • the main body or housing has one or more sharp protrusions, e.g., the sharp corners on a triangular, rectangular or the fan-like device (e.g., Figures IB-ID), which can render it difficult for implantation. It can be challenging to create smooth sutures around such sharp protrusions and there can be impeded pulling force on the surrounding tissues to maintain the curve in the device. For example, in some embodiments, when sutures run around a corner, the pressure on the sutures will cause the area near the sharp edge to dig into the skin leaving the adjacent areas relatively loose.
  • sharp protrusions on the implantable devices are modified to a smoother shape (e.g., rounded or curved) to create better skin contacts during implantation.
  • additional structural features are created on one or more sides of the implantable device to facilitate secure attachment between the device and the site of implantation.
  • a convex groove is created around the circumference of the side of the device to create a snug attachment of one or more sutures that is also running in a circumference around an opening in the skin.
  • holes are implemented on the side of an implantable device to facilitate skin closure by sutures.
  • the main body or housing of an implantable device comprises one or more sides.
  • one side comprises an observation module through which the content within the device (e.g., in a compartment or sub- compartment within the compartment) can be monitored or observed by one or more types of optical equipment.
  • the observation module comprises an observation window.
  • the observation window can be as large as one of the sides, as illustrated in Figure 1 A.
  • the observation window can be opened and/or removed from the main body or housing such that the content of the compartments or sub-compartment can be replenished or removed. Alternatively, new material can be added through the observation window to the compartment or sub-compartments.
  • the main body or housing comprises separate modules through which material can be removed or added.
  • an observation window is an integrated part of the device.
  • the observation windows covering the cell-filled compartments or sub-compartments will be more suitable for transmission of optical signals (e.g., clear glass).
  • parts of the observation window that do not cover the cell-filled compartments or sub-compartments will not be as suitable for transmission of optical signals (e.g., formed by solid or opaque materials).
  • optical equipment such as a microscope or micro video camera
  • a camera is fitted on the optical equipment directly or to the implantable device directly.
  • the camera is an integrated part of the microscope.
  • the video camera can capture what comes through the lenses as well as transmit the captured signals to a computer screen.
  • multiple lenses and / or cameras can be used.
  • the glass forming the observation window is of different thicknesses as required for different microscopy needs (e.g., No. 1-5 thickness) or thicker to allow greater stability.
  • the implantable device has one or more features that permit the device to be connected to the lens of a microscope using specific notches or shapes. This allows for immobilization of the device for microscopic analysis. Preferably, these features are located on the top side of the device.
  • the connection can be achieved by a simple notch or lock-and-key mechanism on the device that fits into a notch on the microscope stage or lens.
  • a recipient of the implantable device e.g., a rodent
  • a specific holder e.g., a cylinder
  • This holder can also allow anesthesia during an imaging process.
  • the notch or lock-and-key mechanism in order to facilitate repeated imaging at high magnification, also provides registration marks for the microscope system to guide and automate its imaging of identical or desirable locations and fields of view of a given region of the device in a given recipient on a real time basis as often as desired (in a living mammal).
  • registration marks on the cover glass can provide orientation and registration to the microscope.
  • video signals are collected of the content within a compartment or sub-compartment, using, for example, a CCD camera.
  • CCD camera a CCD camera
  • data collection can be automated by software that controls the movement of the microscope and camera, or the sample (device on a mammal).
  • an automated software program can guide a lens of a microscope (or a video camera attached thereto) from one sub-compartment to another sub-compartment in a pre-defined order.
  • the program also controls the length of time during which data are collected from each sub-compartments.
  • data are also collected from other parts of the device, for example, from the channels, chambers, wells or any other parts.
  • the device can be moved while the optical equipment stays stationary.
  • an observation platform (upon which a mammal is placed) can move in a controlled manner to permit position-specific and time-specific data collection.
  • a detachable observation window is held to the main body or housing using a retaining ring (or "Circlip").
  • a retaining ring or "Circlip”
  • an insulating O-ring is used between the retaining ring and the cover glass.
  • an observation window includes a cover slip that is protected by a fixed lid-like element to protect the glass from scratches, damage, sunlight, or compromising sterility of the implant.
  • the pressure within the compartments or sub-compartments is controlled, using, for example, the lid-like element.
  • one or more fasteners with tension screws are attached to the lid-like element such that a specific pressure can be applied upon the content within the
  • the lid-like element contains electrodes that pass beneath the coverslip and allow an electrical current or fluid to be added to the internal compartment, which can all influence cell growth and differentiation.
  • the lid-like element is fixed onto the device (e.g., using screws) in a circular fashion.
  • the lid-like element also contain an option to activate a cutting surface that, when turned in the circular screw motion, cuts out the content of the device and allows it to be severed from connection to the body and removed. Replacement matrix can be added to re-grow new tissue.
  • one side of the main body or housing of the implantable device also contains an interaction module (e.g., element 40 in Figure 1 A), as an integrated part of the implantable device.
  • the interaction module allows a compartment sub- compartment of the device to interact with existing extracellular space and tissues near where the device is implanted.
  • the interaction module comprises an open entrance port to the implantable device such that the content within the compartment or sub- compartment can directly interact with the outside cellular environment.
  • the implantable device comprises multiple open entrance ports that allow different sub-compartments to interact with the outside environment (Figures 2A-2C).
  • Figures 2A-2C more rapid tissue growth can be obtained because there is no interference from non-biological material.
  • Such embodiments also permit more complete interaction with the native tissue environment outside the device.
  • the content in the compartments can leak out of the device. For example, gels or semi-solids may move out of the device into the surrounding tissues.
  • the size of an interaction module can be varied to control the speed at which the implantable device exchanges content with the outside environment.
  • multiple interaction modules can be used to allow more even distribution of content exchange at various locations of the device.
  • an interaction module e.g., an opening covered with mesh or net
  • the interaction module can be located near an attachment groove for ease of construction of the device.
  • the interaction module can be located on the connecting side of the device.
  • the interaction module can be located on the bottom of the device.
  • the interaction module comprises one or more open entrance ports that are covered with a mesh or net made of synthetic or natural material.
  • the mesh or net can be made of nylon with pores of various sizes (e.g., between 10 ⁇ and 500 ⁇ ).
  • the mesh or net is formed with biocompatible or biodegradable material.
  • the same biocompatible or biodegradable material used to construct the main body of the implantable device can be used to form the mesh or net. Exemplary materials are disclosed below in connection with the making of the implantable devices.
  • the mesh or net covering the entrance port provides support for biochemical reagents within the device during implantation and before surrounding tissue invasion.
  • the mesh or net covering the entrance port is a physical barrier for cell entry or escape.
  • the mesh or net covering the entrance port can limit blood vessel sizes.
  • the mesh or net covering the entrance port can also limit potential constraints on oxygenation and waste removal, or cell product release from the implantable device.
  • an injection port can be found on one or more sides of the implantable device for injecting material into the compartment or compartments, or interacting with a microfluidic delivery system.
  • the microfluidic delivery system is embedded within the implantable device.
  • the microfluidic delivery system is external to the implantable device.
  • an injection port system allows removal and introduction of materials (cells, fluids, compounds such as growth factors or drugs) into the implantable device.
  • one or more injection ports are created within the lid or on the sides of the implantable device.
  • an injection port is built into the lid itself.
  • an injection port is built into the sides of the implantable device.
  • Figure 5 illustrates an exemplary injection port system from the top surface of the implantable device.
  • An implantable device as disclosed herein can house numerous internal structural elements, including compartments, sub-compartments within a compartment, channel-like structures connecting compartments or sub-compartments. Compartments and sub-compartments are used for numerous purposes, including as reaction chambers, storage depot or wells.
  • sub-compartments can be of the same size and shape, or of different sizes and shapes. In some embodiments, sub-compartments have the same shape as the compartment (or implantable device). In some embodiments, sub- compartments have shapes that are different from the compartment (or implantable device). [0078] In some embodiments, the main body of the implantable device itself forms all or parts of the boundaries of the compartment or sub-compartments. In some embodiments, the compartments or sub-compartments are created by inserts or molds that are pre-formed, using the same material as that of the main body of the implantable device. In some embodiments, the compartments or sub-compartments are created by inserts or molds that are pre-formed using different materials from that of the main body of the implantable device.
  • the number of sub-compartments is determined by the purpose and intent for which the device is used.
  • the implantable device is used to produce biomolecules for treatment or tissues for
  • devices with a single compartment are used.
  • devices with a few large size sub-compartments are used.
  • a larger volume of cell material can be obtained for experimentation, or to be used as transplantable new replacement tissues. Greater doses of cell products can be generated and released into the body; and greater volume of storage depot for drugs that can be released into the body.
  • implantable devices containing multiples sub- compartments with equal or different sizes are used.
  • multiple sub- compartments allow maintenance of multiple cell populations. Multiple growth factor conditions and extracellular matrix compositions can be analyzed simultaneously in parallel.
  • implantable devices containing multiples sub- compartments are used as a therapeutic delivery system.
  • multiple sub- compartments allow cellular products of different types to be maintained within the unit and provide different biological products to a recipient as needed (e.g., as personalized medicine).
  • the same quantity of a therapeutic reagent is stored in different sub- compartments.
  • different quantities of the same therapeutic reagent are stored in different sub-compartments.
  • multiple sub-compartments containing one or more therapeutic agents are designed to release their content in a sequential manner over a period of time.
  • the therapeutic agents are released over minutes or longer, an hour or long; three hours or longer; six hours or longer; eight hours or longer; twelve hours or longer; fifteen hours or longer; twenty hours or longer; one or more days; two or more days; five or more days; 1 week or longer; two weeks or longer; a month or longer; two months or longer; three months or longer; five months or longer; six months or longer; a year or longer; two years or longer; or five years or longer.
  • multiple devices or a device with multiple compartments or sub-compartments are used to conduct duplicative or parallel experiments.
  • duplicative experiments can be conducted with a device with multiple sub-compartments in the same mammal (e.g., a rodent), thus eliminating variations caused by the natural differences between different mammals (e.g., rodents such as mice).
  • parallel experiments are carried out in the same mammal in different compartments or sub-compartments to test different experimental conditions.
  • multiple compartments or sub-compartments within the implantable device are separated by semi-permeable membranes that can allow passage of materials of specific sizes or charges, which allows the diffusion of some molecules into other compartments or sub-compartments.
  • some compartments or sub- compartments can be used to store compounds that are used as substrates for enzymatic reactions.
  • an implantable device as disclosed herein can be of any size or capacity.
  • the implantable device can have a largest diameter or longest dimension of between 0.5 mm and 500 mm; 1 mm and 400 mm; 1 mm and 300 mm; 1 mm and 200 mm; 1 mm and 150 mm; 1 mm and 100 mm; 1 mm and 80 mm; 1 mm and 50 mm; between 1 mm and 30 mm; between 10 mm and 30 mm; between 15 mm and 30 mm; between 5 mm and 20 mm; between 5 mm and 15 mm; between 5 mm and 10 mm; between 3 mm and 8 mm; between 1 mm and 5 mm; between 2 mm and 5 mm; or between 0.5 mm and 4 mm.
  • the implantable device can have a largest diameter or dimension that is between 5 mm and 14 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is between 14 mm and 300 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is smaller than 0.5 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is larger than 500 mm.
  • an implantable device as disclosed herein has a largest depth of between 0.1 mm and 30 mm; 0.1 mm and 20 mm; 0.1 mm and 15 mm;
  • an implantable device as disclosed herein has a depth smaller than 0.1 mm. In some embodiments, an implantable device as disclosed herein has a depth larger than 30 mm.
  • an implantable device as disclosed herein has a volume capacity of 0.01 mm 3 (0.01 ⁇ ) or larger; 0.1 mm 3 (0.1 ⁇ ) or larger; 0.5 mm 3 (0.5 ⁇ ) or larger; 1 mm 3 (1 ⁇ ) or larger; 2 mm 3 (2 ⁇ ) or larger; 3 mm 3 (3 ⁇ ) or larger; 5 mm 3 (5 ⁇ ) or larger; 8 mm 3 (8 ⁇ ) or larger; 10 mm 3 (10 ⁇ ) or larger; 12 mm 3 (12 ⁇ ) or larger; 15 mm 3
  • implantable devices disclosed herein are micro devices.
  • a micro device can have a diameter or dimension of between 1 mm and 5 mm, a depth of between 0.3 mm and 1.0 mm and a volume capacity of about 0.1 ⁇ to 25 ⁇ .
  • it can be useful to implant a micro device near a smaller organ.
  • multiple micro implantable devices are implanted near or next to a desired location in a recipient.
  • multiple micro implantable devices are implanted in multiple locations.
  • multiple micro implantations can compensate each other in case of possible failure.
  • multiple micro implantations can create synergistic effect and result in better treatment or experimentation.
  • implantable devices disclosed herein are mini devices.
  • a mini device can have a diameter or dimension of between 5 mm and 14 mm, a depth of between 0.3 mm and 1.0 mm and a volume capacity of about 5 ⁇ to 150 ⁇ .
  • a mini device can be used to grow more tissue than the smaller micro units for analysis and production of biological products.
  • implantable devices disclosed herein are macro devices.
  • a macro device can have a diameter or dimension of between 14 mm and 300 mm, a depth of between 0.3 mm and 1.0 mm and a volume capacity of about 40 ⁇ to 100 ml.
  • a macro device can be used to grow more tissue than the smaller micro or mini units for analysis and production of biological products.
  • the size and capacity of an implantable device are adjusted to the site of implantation. In some embodiments, the size and capacity of an implantable device are adjusted to the purpose for which the device is used.
  • the implantable devices can be coated to avoid irritation or other undesirable side effects in the recipient.
  • exemplary coating materials include but are not limited to at least partially alkylated polyethyleneimine (PEI); at least partially alkylated poly(lysine); at least partially alkylated polyornithine; at least partially alkylated poly(amido amine), at least partially alkylated homo- and co-polymers of vinylamine; at least partially alkylated acrylate containing aminogroups, copolymers of vinylamine containing aminogroups with hydrophobic monomers, copolymers of acrylate containing aminogroups with hydrophobic monomers, and amino containing natural and modified polysaccharides and mixtures thereof.
  • PEI polyethyleneimine
  • lysine at least partially alkylated polyornithine
  • at least partially alkylated poly(amido amine) at least partially alkylated homo- and co-polymers of vinylamine
  • biocompatible coating materials can be found, for example, in US Pat. No. 6,127,448 to Domb, entitled “Biocompatible Polymeric Coating Material;” US Pat. Pub. No. 2004/0148016 by Klein and Brazil, entitled “Biocompatible Medical Device Coatings;” US Pat. Pub. No. 2009/0169714 by Burghard et al, entitled “Biocompatible Coatings for Medical Devices;” US Pat. No. 6,406,792 to Bhoppert et al, entitled “Biocompatible Coatings;” US Pat. Pub. No. 2008/0003256 by Martens et al, entitled “Biocompatible Coating of Medical Devices;” each of which is hereby incorporated by reference herein in its entirety.
  • any suitable material can be used to manufacture an implantable device as disclosed herein.
  • Exemplary materials for the main body or housing of the device include but are not limited to stainless steel, surgical steel (e.g., Grade 316), titanium, a plastic or polymeric material, a biocompatible or biodegradable material or a combination thereof.
  • Exemplary biocompatible materials also include but are not limited to polyacrylates, polymethacryates, polyureas, polyurethanes, polyolefms, polyvinylhalides, polyvinylidenehalides, polyvinylethers, polyvinylaromatics, polyvinylesters, polyacrylonitriles, alkyd resins, polysiloxanes and epoxy resins.
  • biocompatible biodegradable polymers include, without limitation, polycaprolactone, poly(L-lactide), poly(D,L-lactide), poly(D,L-lactide-co- PEG) block copolymers, poly(D,L-lactide-co-trimethylene carbonate), poly(lactide-co- glycolide), polydioxanone (PDS), polyorthoester, polyanhydride, poly(glycolic acid-co- trimethylene carbonate), polyphosphoester, polyphosphoester urethane, poly(amino acids), polycyanoacrylates, poly(trimethylene carbonate), poly(iminocarbonate), polycarbonates, polyurethanes, polyalkylene oxalates, polyphosphazenes, PHA-PEG, and combinations thereof.
  • polycaprolactone poly(L-lactide), poly(D,L-lactide), poly(D,L-lactide-co- PEG) block copolymers, poly
  • the PHA may include poly(a-hydroxyacids), poly(P-hydroxyacid) such as poly(3- hydroxybutyrate) (PHB), poly(3-hydroxybutyrate-co-valerate) (PHBV), poly(3- hydroxyproprionate) (PHP), poly(3-hydroxyhexanoate) (PHH), or poly(4-hydroxyacid) such as poly poly(4-hydroxybutyrate), poly(4-hydroxyvalerate), poly(4-hydroxyhexanoate), poly(hydroxyvalerate), poly(tyrosine carbonates), poly(tyrosine arylates), poly(ester amide), polyhydroxyalkanoates (PHA), poly(3-hydroxyalkanoates) such as poly(3- hydroxypropanoate), poly(3-hydroxybutyrate), poly(3-hydroxyvalerate), poly(3- hydroxyhexanoate), poly(3-hydroxyheptanoate) and poly(3-hydroxyoctanoate), poly(4- hydroxy
  • polyacrylonitrile polyvinyl ketones, polyvinyl aromatics, such as polystyrene, polyvinyl esters, such as polyvinyl acetate, copolymers of vinyl monomers with each other and olefins, such as ethylene-methyl methacrylate copolymers, acrylonitrile-styrene copolymers, ABS resins, and ethylene-vinyl acetate copolymers, polyamides, such as Nylon 66 and polycaprolactam, alkyd resins, polycarbonates, polyoxymethylenes, polyimides, polyethers, poly(glyceryl sebacate), poly(propylene fumarate), poly(n-butyl methacrylate), poly(sec- butyl methacrylate), poly(isobutyl methacrylate), poly(tert-butyl methacrylate), poly(n-propyl methacrylate), poly(isopropyl methacrylate), poly(ethyl me
  • poly(ethylene oxide-co-lactic acid) PEO/PLA
  • polyalkylene oxides such as poly(ethylene oxide), poly(propylene oxide), poly(ether ester), polyalkylene oxalates, phosphoryl choline containing polymer, choline, poly(aspirin), polymers and co-polymers of hydroxyl bearing monomers such as 2-hydroxyethyl methacrylate (HEMA), hydroxypropyl methacrylate (HPMA),
  • hydroxypropylmethacrylamide PEG acrylate (PEGA), PEG methacrylate, methacrylate polymers containing 2-methacryloyloxyethyl- phosphorylcholine (MPC) and n-vinyl pyrrolidone (VP), carboxylic acid bearing monomers such as methacrylic acid (MA), acrylic acid (AA), alkoxymethacrylate, alkoxyacrylate, and 3-trimethylsilylpropyl methacrylate (TMSPMA), poly(styrene-isoprene-styrene)-PEG (SIS-PEG), polystyrene-PEG,
  • polyisobutylene-PEG polycaprolactone-PEG (PCL-PEG), PLA-PEG, poly(methyl methacrylate), MED610, poly(methyl methacrylate)-PEG (PMMA-PEG),
  • polydimethylsiloxane-co-PEG PDMS-PEG
  • poly(vinylidene fiuoride)-PEG PVDF-PEG
  • PLURONICTM surfactants polypropylene oxide-co-polyethylene glycol
  • poly(tetramethylene glycol), hydroxy functional poly( vinyl pyrrolidone), biomolecules such as collagen, chitosan, alginate, fibrin, fibrinogen, cellulose, starch, dextran, dextrin, hyaluronic acid, fragments and derivatives of hyaluronic acid, heparin, fragments and derivatives of heparin, glycosamino glycan (GAG), GAG derivatives, polysaccharide, elastin, elastin protein mimetics, or combinations thereof.
  • biomolecules such as collagen, chitosan, alginate, fibrin, fibrinogen, cellulose, starch, dextran, dextrin, hyaluronic acid, fragments and derivatives of hyaluronic acid, heparin, fragments and derivatives of heparin, glycosamino glycan (GAG), GAG derivatives, polysaccharide, elastin, elastin
  • the device when an implantable device is formed with one or more biodegradable materials, the device degrades over one or more days, two or more days, five or more days, 1 week or longer, two weeks or longer, a month or longer, two months or longer, three months or longer, five months or longer, six months or longer, a year or longer, two years or longer or five years or longer.
  • the compartment or sub- compartments within an implantable device are made of the same material as the main body or housing of the implantable device. In some embodiments, compartments or sub- compartments within an implantable device are made of material that is different from that of the main body or housing.
  • sub-compartments are formed by inserting a pre-formed divider or insert into the main body or housing, similar to the design of an insert for an ice-cube tray.
  • a pre-formed divider is formed based on the dimension of the main body or housing.
  • multiple pre-formed dividers can be used with the same type of main body or housing to create compartments or sub-compartments of different shapes.
  • the dividers or inserts are added to the main body or housing before any material is added to the resulting compartments or sub-compartments.
  • walls between compartments or sub-compartments are impermeable. In some embodiments, walls between compartments or sub-compartments are semi-permeable and permit transmission.
  • a cover slip is modified to have dividers that slide into the main body or housing and create compartments or sub-compartments.
  • the biochemical mixture is added before the cover slip is attached.
  • compartments or sub-compartments are formed using backbone or substrate material at the same time when the main body or housing of the implantable device is created ( Figures 2A-2C).
  • the material for making the main body or housing of the implantable device is also used to construct other parts of the device, such as a reaction chamber, a well, a channel, or a mesh or net of the interaction module.
  • an implantable device is determined by the type of material used and the purpose of the device.
  • a metal device can be created by metal lathing, milling, and CNC molding.
  • a plastic device can be made by injection molding, extrusion molding or any other suitable method.
  • methods of three-dimensional printing on metals and other polymeric materials are used.
  • methods of laser printing or laser scribing are used.
  • an on-board biomarker monitoring system can be integrated with an implantable device described herein.
  • a fluid or compound delivery system can be integrated with an implantable device described herein.
  • an integration can be either functional or structural.
  • chambers, channels, wells, compartments and / or sub-compartments form a sophisticated network dedicated for a specific biological or biochemical assay.
  • each individual component of the network and the content therein provide an element for completing the desired assay, e.g., by hosting cells or tissues, detecting biomarkers, adjusting reaction conditions, and producing signals.
  • the network is designed for delivering specific compounds. The network-like configuration allows miniaturization of implantable devices that are specific for certain biological or biochemical assays.
  • a pre-designed network of chambers, channels, wells, compartments and / or sub-compartments can be pre-formed on an insert or chip-like device (e.g., Figure 3).
  • the insert or chip can be put into an implantable device.
  • a pre-designed network of chambers, channels, wells, compartments and/or sub-compartments can be constructed as parts of the implantable device.
  • various components of the network can be made individually and assembled in a pre-formed device to form the network that performs one or more biological assays.
  • the implantable device can be made before or at the same time as an assay insert.
  • an insert or chip dedicated for a specific biochemical assay can be placed in a defined position within an implantable device, e.g., in the center or close to the interaction module.
  • the system can be used to deliver precise quantities to the bioreactor at pre-set times, from storage depots to a reaction chamber.
  • a wireless communication module can be integrated with an implantable device described herein.
  • the on-board biomarker detection system can convert measured data to a digital signal and wirelessly transmitted from the implant to an external receiving device such as a computer.
  • a wireless communication module can also facilitate communication of data to a mobile phone, a PDA, other wireless communication device. Such embodiments can monitor the device and provide live or instant feedback. For example, insulin levels within and near an implantable device can be detected, monitored and recorded by a smart phone or PDA device.
  • a wireless signal receiver can be integrated with an implantable device described herein.
  • a small computer chip can be integrated with an implantable device described herein.
  • the small computer chip or signal receiver can be used to control sequential release of biochemical reagents from the chambers, channels, wells, compartments and/or sub-compartments on an implantable device.
  • a biomarker detection system can communicate with its own on-board computer equipment to send and receive signals.
  • the level of the biomolecules can be communicated to medical professionals or a data center, which in turn provides medical aid or triggers an alert for replacement of the implant.
  • the signals can be used to activate cells in particular ways as needed: to release cell products, or to release drugs or other compounds into the body or into cells within the device.
  • built-in sensors can detect sugar levels and trigger release of insulin from genetically engineered cells within the device, or insulin within insulin-storage depots within the device is released as a response.
  • new measurements of sugar levels are taken after blood circulates around the body, and returns to the device. The new measurement provides information on whether additional adjustment of the levels of insulin is needed.
  • attachment of a lens can be used to focus the camera or other optical equipment.
  • wireless capabilities can be used to transmit image or video data.
  • an implantable device as disclosed herein is equipped with the ability to apply light or radiation within the device. Such embodiments provide the advantage of creating excitation and emission of desired wavelengths of light, which can be useful in visualizing the content within the device.
  • radiation at various wavelengths e.g., ultraviolet
  • structural features are created on one or more sides of the implantable device to create secure attachment between the device and one or more additional modules.
  • multiple functionalities can be incorporated into an implantable device by microfabrication techniques. An exemplary embodiment of a multifunctional implantable device can be found in Figure 6. The different functional modules and features will be described with more details in connection with a multi-step biomolecules synthesis process.
  • the content within a compartment or sub- compartment of an implantable device includes at least a plurality of cells and a biochemical composition in which the cells may be maintained, grow, develop and/or differentiate.
  • the biochemical composition comprises a biocompatible buffer, a growth media or an extracellular matrix.
  • the cells or tissue used in the device can be suspended in a liquid trapped within a sub-compartment, adhered to the inner walls of the compartment or immobilized on an appropriate support structure provided within the compartment.
  • the cells can be embedded in a gel matrix (e.g., agar, alginate, chitosan, polyglycolic acid, polylactic acid, and the like).
  • a porous scaffold e.g., an alignate scaffold
  • microcapsules or microbeads can be used to encapsulate or capture cells in the cellular compartment.
  • a commercially available growth medium or matrix for mammalian cells is used.
  • MatrigelTM is the trade name for a gelatinous protein mixture secreted by Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells and marketed by BD Biosciences and by Trevigen Inc. under the name Cultrex BME. This mixture resembles the complex extracellular environment found in many tissues and is used by cell biologists as a substrate for cell culture.
  • Components of a standard growth medium or matrix for mammalian cells include but are not limited to extracellular matrix components, growth factors, various cytokines, and one or more pharmaceutical agents, as listed in Table 1.
  • Extract from the EHS tumor e.g., MatrigelTM from Extracellular Matrix components
  • Exemplary growth factors include but are not limited to adrenomedullin (AM), angiopoietin (Ang), autocrine motility factor, bone morphogenetic proteins (BMPs), brain-derived neurotrophic factor (BDNF), epidermal growth factor (EGF), erythropoietin (EPO), fibroblast growth factor (FGF) 1, 2, 3, glial cell line-derived neurotrophic factor (GDNF), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony- stimulating factor (GM-CSF), growth differentiation factor-9 (GDF9), hepatocyte growth factor (HGF), hepatoma-derived growth factor (HDGF), insulin-like growth factor (IGF), migration-stimulating factor, myostatin (GDF-8), nerve growth factor (NGF) and other neurotrophins, platelet-derived growth factor (PDGF), thrombopoietin (TPO), transforming growth factor alpha (TGF-a),
  • cytokines include but are not limited to interleukin 2, interleukin 15 preproprotein, tumor necrosis factor (ligand) superfamily, member 18, interleukin 26, interleukin 20, interleukin 22, interferon epsilon 1, interferon-gamma, colony stimulating factor 2, interleukin 19 isoform 2, tumor necrosis factor (ligand) superfamily, member 4, interleukin 24 isoform 1, interferon, beta 1, fibroblast, interleukin 5, interleukin 13, growth hormone 2 isoform 1, interferon, omega 1, interleukin 12 A, tumor necrosis factor (ligand) superfamily, member 10, interleukin 6 (interferon, beta 2), interferon, alpha 1, growth hormone 1 isoform 1 , leptin, interleukin 1 , beta proprotein, tumor necrosis factor alpha, interferon kappa, interleukin 3, interleukin 10, tumor necrosis factor (ligand) superfamily, member 15, prol
  • the biochemical composition also includes metabolites, small molecules or macromolecules.
  • Exemplary metabolites include but are not limited to alcohols (e.g., ethanol), amino acids (e.g., glutamic acid, aspartic acid), nucleotides (e.g., 5' guanylic acid), antioxidants (e.g., isoasorbic acid), organic acids (e.g., acetic acid, lactic acid), polyols (e.g., glycerol), vitamins (e.g., B2), minerals, and electrolytes.
  • alcohols e.g., ethanol
  • amino acids e.g., glutamic acid, aspartic acid
  • nucleotides e.g., 5' guanylic acid
  • antioxidants e.g., isoasorbic acid
  • organic acids e.g., acetic acid, lactic acid
  • polyols e.g., glycerol
  • vitamins e.g., B2
  • electrolytes e.g., B2
  • Exemplary small chemical molecules include any chemical compounds, including inorganic and organic compounds, for example, formaldehyde, acetylsalicylic acid, methanol, ibuprofen, and statins.
  • Exemplary macromolecules include but are not limited to monoclonal and polyclonal antibodies, nucleic acid, lipid, fatty acid, and insulin.
  • an implantable device as disclosed herein can be implanted in a mammal, for example, a mouse, a rat, a dog, a cow, a sheep, a goat, a cat, or a human.
  • standard surgical techniques using tools such as, forceps and sutures are used to surgically open and close the skin around the implant.
  • a punch mechanism can be used for easy attachment in subcutaneous locations.
  • the mechanism is similar to that of grommet pliers and grommets (see Figure 7).
  • the method includes punching the lid and upper compartments to lower compartments, or only a pinching mechanism running on the sides of the device that is secured with the punch.
  • Advantageous the latter can be used to create quick subcutaneous implantation.
  • the punch-based mechanism while efficient in securing the device, must be done carefully to avoid too much damage to the surrounding tissues and result in inflammation.
  • the method is only useful for subcutaneous implantation, not other locations (e.g., peritoneal cavity of the abdomen).
  • an implantable device is implanted within the skin in an accessible area of the body of a mammal, especially when the purpose of the implantation is for observing biochemical reactions and cell developments.
  • the device is attached to the dorsal skin region of a mammal (e.g., on the back of a rodent). Such attachments allow easy access for microscopy analysis.
  • the protrusion caused by the implant can create discomfort for a mammal or risk of dislodging or damaging the device; e.g., a device can be knocked around when a recipient mammal hits objects in the cage where the mammal lives.
  • an implantable device is implanted near the dorsal head region of a mammal (e.g., the "hat” position between the ears in a rodent). Such attachments allow easy access for microscopy analysis and avoid mechanical irritation (e.g., scratching by the recipient mammal). However, the implantation site can limit the size of the device.
  • an implantable device is implanted to the abdominal region. In such embodiments, it is possible to accommodate larger implantable devices.
  • an implantable device is implanted in locations internal to the skin (e.g., in the intraperitoneal cavity or beside the liver or pancreas).
  • the device is hidden from view and protected from accidental damage.
  • the biochemical composition and cells can benefit from the cellular and extracellular components of different locations within the body (e.g., via interactions with extracellular fluid components from the liver or pancreas).
  • Such implantation allows cellular products within the device to be delivered to specific locations as needed (e.g., antibodies produced within the unit are provided to regions near lymph nodes or the lymphatic system).
  • it is also more difficult surgically to implant in such locations and more difficult to analyze or monitor over time e.g., microscopy can be difficult if the location to be observed is not external facing).
  • an implantable device are made of biodegradable materials. It is possible to package the device with suitable biochemical material that will be gradually released and stimulate the growth of implanted cells/tissues. Over time, when the device is completely degraded, the resulting cells/tissue becomes an integral part of the target organ.
  • the implantable devices can be used for various purposes, including but not limited as a research tool, a drug testing tool, a tool for producing biomolecules (small or large), a tool for tissue or organ implant/transplant, a tool for detecting biomarkers or biomolecules, or an extracorporeal body monitor and modulator.
  • an implantable device as disclosed herein is used as a research tool.
  • an implantable device is used for evaluating the functions of individual biological factors in a live mammal. Because the device is implanted in a live mammal, it is possible to analyze the comprehensive response of the whole mammal to a particular stimulus.
  • pressure can be applied to the system before the effects are observed (e.g., it is possible to analyze the effects of pressure on bone growth and remodeling).
  • the system can also be subject to micro or zero- gravity conditions (e.g., experiments done under such conditions have shown unusual properties in stem cells).
  • an electrical current can be applied to the system.
  • quantum mechanical microscopy can be used in connection with the system.
  • an implantable device as disclosed herein is used to evaluate the functions of small or large biomolecules (e.g., proteins, peptides, nucleic acids, combinations and analogs thereof) in a live mammal.
  • biomolecules e.g., proteins, peptides, nucleic acids, combinations and analogs thereof
  • exemplary uses include but are not limited to angiogenesis study, anti-angiogenesis study (e.g., in the context of anti-cancer drugs analysis or screening), cancer and anti-cancer studies, stem cell studies, nerve regeneration study (e.g., determining the growth factors that contribute to nerve sprouting and re-growth in vivo), and liver toxicity study (e.g., testing liver drugs and evaluating their toxicity during pre-clinical development).
  • an implantable device as disclosed herein is used to evaluate the function of stem cells and tissue constructs before the cells or tissues are used in further treatment or therapy.
  • This use of the implantable device is applicable to all organ/tissue types, including but not limited to liver tissue, pancreas tissue, kidney tissue, lung tissue, skin tissue, bone marrow and lymph node tissue, thyroid tissue, pituitary tissue, brain tissue, muscle tissue (including myocardium, skeletal muscle and smooth muscle), cartilage tissue, gastrointestinal tissues, reproductive tissues including uterus, and ovaries.
  • an implantable device as disclosed herein is used as a part of a live imaging system, in which images, videos or other optical data of live cells can be collected, stored, displayed, and processed. Based on previous data on fully subcutaneous implants, in some embodiments, cells and microvasculature can be observed to be entering the device within 1 week. In some embodiments, approximately 1.5 mm of ingrowth can be observed within 4 weeks, including functioning blood vessels, and associated cells from the host. In some embodiments, the cover glass and retaining clip can be removed at 4 weeks and stains such as Hoescht 33342 nuclear stain, or other antibody stains can be added to stain live cells, or cells and growth factors can be added.
  • stains such as Hoescht 33342 nuclear stain, or other antibody stains can be added to stain live cells, or cells and growth factors can be added.
  • the implantable device can be resealed; and cell growth can be further monitored.
  • the kinetics of diverse cell populations as they enter the device can be observed from day 1 to 6 weeks after the implantation.
  • the relations and organization of multiple cell populations in one location can be examined.
  • blood vessel growth within a live mammal e.g., a rodent
  • micro CT scan micro CT scan
  • partial or the entire content within the implantable device can be removed and subject to biological and biochemical analyses.
  • the removed content can be subject to sectioning, staining and imaging by conventional confocal microscopy methods.
  • removed devices content cam be digested and quantified by flow cytometry, mass spectrometry, or other analytical methods.
  • contributions of bone marrow derived cells migrating, and proliferating within the device can be examined.
  • wildtype mice subject to lethal irradiation of C57B16 can be used. These mice are then injected with bone marrow from GFP transgenic wildtype mice.
  • signs of positive engraftment of GFP bone marrow can be observed at about 4 week after implantable devices are introduced into these mice.
  • An exemplary sign is the production of GFP-labeled peripheral blood.
  • content in the device can reveal GFP positive cells (which are inferred to have originated from the bone marrow) as well as non-GFP cells (which have come from tissues neighboring the device). This is a test for determining cells that are bone marrow derived and can have bone marrow stem cell potential.
  • an implantable device as disclosed herein is used as a drug- testing tool.
  • the device allows direct observation of cellular response. Also advantageously, it is possible to conduct drug metabolism study in the live mammal, including drug safety and toxicity studies, dosing studies, drug kinetics, efficiency and efficacy, and bioavailability studies.
  • a multi-functional implantable device can be used as a personalized tissue assay (e.g., drug screening on a patient biopsy sample with cancer drugs) ( Figure 8).
  • a biopsy sample can be collected from an individual's tumor by conventional methods. This sample can be collected and maintained either live or in a frozen state before division into identical pieces by shape and weight.
  • a mammal containing implantable devices can be prepared to allow the biopsy sample successfully engraft and grow within the devices.
  • immuno-compromised mice can be employed to enable engraftment of human tissues.
  • biopsy samples of the same size or weight from a mammal can be implanted within devices in multiple test mammals (which can be the same or different from the sample mammal.
  • the biopsy samples engraft and live within the devices and are analyzed by optical methods or biomarker analysis.
  • the mammals can subsequently be administered therapeutics.
  • the effects of the therapeutics can be assessed.
  • the therapeutic agents that are effective in destroying or reducing one diseased state of the biopsy sample can be selected for additional study. In some embodiments, duplicate tests are performed.
  • the selected therapeutics are given to the mammal from which the biopsy sample is taken.
  • biopsy samples of the same size or weight from a mammal can be implanted within devices in a same test mammal.
  • the implantable device comprises multiple sub-compartments or chambers that are subject to the same growth conditions and each contains a biopsy sample. Different therapeutic agents are then administered to different sub-compartments and the effects on the biopsy samples within each sub-compartment are analyzed and compared with each other.
  • the biopsy sample is associated with a type of cancer, including but not limited to Acute lymphoblastic leukemia; Acute myeloid leukemia; Adrenocortical carcinoma; AIDS-related cancers; AIDS-related lymphoma Anal cancer; Appendix cancer; Astrocytoma, childhood cerebellar or cerebral; Basal cell carcinoma; Bile duct cancer, extrahepatic; Bladder cancer; Bone cancer; Osteosarcoma/Malignant fibrous histiocytoma; Brainstem glioma; Brain tumor; Brain tumor; cerebellar astrocytoma; Brain tumor; cerebral astrocytoma/malignant glioma; Brain tumor, ependymoma; Brain tumor, medulloblastoma; Brain tumor, supratentorial primitive neuroectodermal tumors; Brain tumor, visual pathway and hypothalamic glioma; Breast cancer; Bronchial cancer; Astrocytoma, childhood cerebellar or cerebral; Basal cell carcinoma; Bil
  • adenomas/carcinoids Burkitt lymphoma; Carcinoid tumor, childhood; Carcinoid tumor, gastrointestinal; Carcinoma of unknown primary; Central nervous system lymphoma, primary; Cerebellar astrocytoma, childhood; Cerebral astrocytoma/Malignant glioma, childhood; Cervical cancer; Childhood cancers; Chronic lymphocytic leukemia; Chronic myelogenous leukemia; Chronic myeloproliferative disorders; Colon Cancer; Cutaneous T- cell lymphoma; Desmoplastic small round cell tumor; Endometrial cancer; Ependymoma; Esophageal cancer; Ewing's sarcoma in the Ewing family of tumors; Extracranial germ cell tumor, Childhood; Extragonadal Germ cell tumor; Extrahepatic bile duct cancer; Eye Cancer, Intraocular melanoma; Eye Cancer, Retinoblastoma; Gallbladder cancer; Gastric (S
  • Leukemia acute myeloid (also called acute myelogenous leukemia); Leukemia, chronic lymphocytic (also called chronic lymphocytic leukemia); Leukemia, chronic myelogenous (also called chronic myeloid leukemia); Leukemia, hairy cell; Lip and Oral Cavity Cancer; Liposarcoma; Liver Cancer (Primary); Lung Cancer, Non-Small Cell; Lung Cancer, Small Cell; Lymphoma; Lymphoma, AIDS-related; Lymphoma, Burkitt; Lymphoma, cutaneous T- Cell; Lymphoma, Hodgkin; Lymphomas, Non-Hodgkin (an old classification of all lymphomas except Hodgkin's); Lymphoma, Primary Central Nervous System;
  • Macroglobulinemia Waldenstrom; Malignant Fibrous Histiocytoma of Bone/Osteosarcoma; Medulloblastoma, Childhood; Melanoma; Melanoma, Intraocular (Eye); Merkel Cell Carcinoma; Mesothelioma, Adult Malignant; Mesothelioma, Childhood; Metastatic
  • Myelodysplasia Syndromes Myelodysplastic/Myeloproliferative Diseases; Myelogenous Leukemia, Chronic; Myeloid Leukemia, Adult Acute; Myeloid Leukemia, Childhood Acute; Myeloma, Multiple (Cancer of the Bone-Marrow); Myeloproliferative Disorders, Chronic; Nasal cavity and paranasal sinus cancer; Nasopharyngeal carcinoma; Neuroblastoma; Non- Hodgkin lymphoma; Non-small cell lung cancer; Oral cancer; Oropharyngeal cancer;
  • Testicular cancer Throat cancer; Thymoma, childhood; Thymoma and Thymic carcinoma; Thyroid cancer; Thyroid cancer, childhood; Transitional cell cancer of the renal pelvis and ureter; Trophoblastic tumor, gestational; Unknown primary site, carcinoma of, adult;
  • the biopsy sample is from a non-cancer related disease.
  • an implantable device as disclosed herein is used as a reservoir for growing small or large biomolecules.
  • exemplary small molecules include but are not limited to aspirin, morphine for chronic pain, methadone for heroine withdrawal symptoms; hormones, vitamins and other non-regulated supplements and nutrients (e.g., Vitamins A-E, etc.) in slow release from within cells or just from within the implantable device).
  • biomolecules that can be produced by the device include but are not limited to insulin, various growth factors, prolactin, Cortisol, estrogen, progesterone, testosterone, serotonin, aldosterone, erythropoietin, granulocyte-colony stimulating factor (e.g., GCSF and analogs), and all types of thyroid hormones.
  • antibodies can also be produced where the device functions as an artificial lymph node and, for example, produces custom designed antibodies.
  • a multi-functional implantable device can be used to construct a specific 'cellular' manufacturing system using different compartments and sub- compartments within the device (see Figures 3 and 6).
  • the implantable device includes a series of channels and sub-compartments containing various modules. These channels and sub-compartments can mimic the original cellular processes by releasing and mixing biochemical reagents as necessary to result in a defined cascade of biochemical reactions.
  • the device is filled with biomolecular media that stimulates invasion by endothelium and eventual blood vessel formation through the device.
  • genetically engineered cells are introduced (or pre-loaded) into the
  • a microprocessing computer sends the appropriate electrical and/or other signals to cells or chemical storage depots within the device to trigger the release of their contents. For example, insulin-producing cells will release insulin at locations 1-4 in Figure 6.
  • the out-flowing blood or body fluid leaving the device is altered before exiting to the rest of the body.
  • the product produced by cells in the implantable device can be removed from the device or transferred to a different location within the device via the injection port or observation module.
  • an implantable device as disclosed herein is used for growing cells and tissues for implants or transplants.
  • the cells that are introduced into the system can be modified or unmodified (e.g., genetically modified), differentiated or un-differentiated.
  • cells grown in the device are altered by genetic or other methods, to optimize the production of a desired small or large biomolecule. Exemplary issues that can later be transplanted to different locations include but are not limited to muscle tissues, nerve tissues, liver tissue, pancreas tissue, and blood vessels.
  • embryonic stem cells e.g., blastocyst-derived
  • embryonic stem cells are cultured and produced within an implantable device as disclosed herein.
  • blastocyst-derived are cultured and produced within an implantable device as disclosed herein.
  • blastocyst-derived stem cells isolated from the inner cell mass of blastocysts can be used.
  • adult stem cells or somatic stem cells which are found in various tissues (e.g., from bone marrow derived sources), can also be used. Additional adult stem cells include but are not limited to hematopoietic stem cells, mammary stem cells, intestinal stem cells, mesenchymal stem cells, endothelial stem cells, neural stem cells, olfactory adult stem cells, neural crest stem cells, and testicular cells.
  • non-stem cells are used. Potentially, all of the 200 or so mammalian cell types within the body can be used in an implantable device as disclosed herein. Exemplary cells include but are not limited to, for example, cells found within a non- embryonic adult, such as insulin secreting cells (e.g., from adults or cadavers) or hepatocytes; islets of Langerhands; cells via somatic cell nuclear transfer (SCNT cells); cells via induced pluripotent stem cells (iPSs cells) either derived by genetic or chemical means; and cells from umbilical cord blood (UCB) cells.
  • insulin secreting cells e.g., from adults or cadavers
  • SCNT cells somatic cell nuclear transfer
  • iPSs cells induced pluripotent stem cells
  • URB umbilical cord blood
  • donor cells are used, including autologous (self) cells or non-autologous cells (e.g., allogenic or xenogenic cells from unrelated donors or other species).
  • the implantable device which contains the cells to be transplanted, are made of synthetic biodegradable materials.
  • the implantable device can be implanted near a target organ at the time of transplant.
  • a device containing liver cells can be placed at the site where the diseased liver portion is removed.
  • the implantable device also contains the necessary biochemical composition for supporting the growth of liver cell therein.
  • the cells can develop into liver tissue over a period of time within the mammal receiving the transplant as the device degrades over a longer period of time.
  • the cells are jump-started in a separate device and can develop into tissue elements before being transplanted into a live mammal.
  • the implantable device being transplanted into the live mammal does not need to have an observation module.
  • tissue/organ include but are not limited to liver tissue, pancreas tissue, kidney tissue, lung tissue, skin tissue, bone marrow and lymph node tissue, thyroid tissue, pituitary tissue, brain tissue, muscle tissue (including myocardium, skeletal muscle and smooth muscle), cartilage tissue, gastrointestinal tissues, and reproductive tissues including tissues from uterus and ovaries.
  • an implantable device as disclosed herein is used for detecting known and unknown biomarkers or from within the device and outside the device within the body, e.g., vitamin levels, Cortisol levels, in addition to products relevant to the implant.
  • a biomarker can be a small molecule as well as a
  • a biomarker is a term often used to refer to a protein measured in blood whose concentration reflects the severity or presence of some disease state. More generally, a biomarker is anything that can be used as an indicator of a particular disease state or some other physiological state of an organism. Exemplary biomarkers for many diseases include but are not limited to various antigens or antibodies, serum LDL for cholesterol, blood pressure, Carcinoembryonic antigens for cancer. In cell biology, a biomarker is a molecule that is present or absent from a particular cell type. This facilitates the characterization of a cell type, their identification, and eventually their isolation.
  • Cell sorting techniques are based on cellular biomarkers (for example, fluorescent-activated cell sorting).
  • a biomarker can be used to identify a cell population, make a diagnosis, or measure the progress of disease or the effects of treatment.
  • the protein Oct-4 that is a biomarker found in embryonic stem cells
  • the Carcinoembryonic antigen (CEA) is a tumor marker used to follow up cancer treatment
  • the Prostate Specific Antigen (PSA) is used for diagnosis of prostate cancer.
  • biomarkers from all locations in contact with the device can be detected (e.g. locations that are internal and external to the device).
  • a multi-functional implantable device as shown in Figure 6 can be used for detecting a target biomarker or biomolecule. 6.6.6 Implantable Devices as an Extracorporeal Body Monitor and Modulator
  • an implantable device as disclosed herein can be attached to the body extracorporeally (e.g., outside the body, but attached through connection to the circulation etc.)
  • the device can be used to grow or maintain cells ex vivo, it may be provided extracorporeally and yet be connected to a subject's vasculature.
  • the device in a similar configuration can also be utilized to provide convenient vascular access, such as provided by a Portacath device, or similar system.
  • a bioreactor that is implanted under the dorsal skin can be employed to test this potentially powerful system.
  • the bioreactor for use in this protocol has an external facing glass "window" that remains facing external to the body.
  • the glass window made of specific microscope coverslip material
  • the bioreactor contains matrix components, including an extracellular matrix commercially available for growing blood vessels in cultures with additional growth factors included (in this case bFGF) .
  • the system can be implanted using known surgical procedures or as described herein.
  • an implanted system is allowed to grow blood vessels for 2 or more days; 5 or more days, 7 or more days; 10 or more days; 14 or more days; 21 or more days; 28 or more days; 36 or more days; 54 or more days; 60 or more days; 90 or more days; or even longer.
  • animals can be anesthetized in a custom-made plexiglass container and the bioreactor can be viewed under the microscope for approximately 30 minutes. Daily imaging can be undertaken at 1, 7, 14 and 28 days; or at other intervals.
  • the animals can be sacrificed at the end of the implantation period (e.g., 28 days post
  • bioreactors can be disassembled and examined by conventional methods.
  • the bioreactors can be implanted in Green
  • GFP Fluorescent Protein
  • a chamber comprising a short cylinder ( ⁇ 3mm) of Grade 316 surgical stainless steel (Prototype 2.4) is implanted subcutaneously.
  • the chamber is a disc approximately 3 mm high and 8 mm wide.
  • the top external-facing coverslip will be mounted on a stainless steel ring, to provide protection to the skin sutured in a 'cinch' up beneath it.
  • the lumen of the chamber is filled with a protein gel (Matrigel) which incorporates angiogenic stimulants.
  • the chamber was subcutaneously implanted on the midline at the mid back via an incision through the overlying skin such that it is implanted like a pacemaker, but with an external surface with an external window surrounded by a protective steel ring.
  • mice will be transferred to a custom made plexiglass anesthetic container and placed on a fluorescent microscope stage for imaging.
  • animals can be examined in initial tests for likely ⁇ lhr, and once this is established, 20 minutes of anesthesia for imaging should be sufficient. Animals will be returned to their cages and given treats following imaging. Animals will be viewed under the microscope a maximum of 3X per week, and less if not needed.
  • Various experimental endpoints can be reached for the implantation.
  • cells are allowed to grow to a maximum volume into the bioreactors of 3mm after which the animals will be euthanized.
  • bioreactors will be implanted for a maximum of 28 days.
  • a bioreactor device comprising a first surface being configured capable of fluidic communication with the extracellular tissues and tissue microvasculature of a subject and containing divided sub-compartments containing cells, matrix materials, and growth factors, with ports for delivering of these materials as needed, and the second surface being separated from the external environment by a window permitting observation intravitally.
  • provided herein is a method for delivering a cell population using the bioreactor device herein, with matrix materials, growth factors and pharmaceuticals to support its function stability.
  • a bioreactor device comprising a first compartment being configured capable of fluidic communication with the extracellular matrix of a subject and configured for containing cells, implanted matrix and growth factors.
  • the bioreactor makes it possible for blood vessels from the subject to grow into the device over time and provide further exchange with the body.
  • the permeable membrane permits passage of cells from existing tissues to the bioreactor.
  • the permeable membrane enables passage of fluids and molecules to and from the interior of the bioreactor.
  • each sub-compartment includes a cell injection port.
  • At least one of the first compartment, the second compartment and the membrane is made of the same material. In some embodiments with respect to any aspect disclosed herein, at least one of the first compartment, the second compartment and the membrane is made of different materials.
  • the first compartment and/or the membrane comprise at least one pharmaceutical agent.
  • the pharmaceutical agent is a therapeutic agent or a diagnostic agent.
  • the polymer fibers have spaces with a cutoff of about between 0.5 - 400 micrometers.
  • the at least one of the first compartment and the second compartment is made of non-woven polymer fibers.
  • the cell population comprises an insulin-secreting cell population.
  • the cell population comprises Islets of Langerhans, thyroid cells, hepatocytes, hematopoietic cells, lymphoid cells or other cell populations found within the body.
  • the cell population comprises embryonic stem cells of any species origin.
  • the cell population comprises cells derived by somatic cell nuclear transfer.
  • the cell population comprises cells derived as induced pluripotent stem cells, by genetic or chemical means.
  • the cell population comprises cells altered by genetic methods.
  • the cell population comprises cells derived from adult tissues and from adult stem cell populations.
  • the cell population comprises cells of other types derived by genetic or synthetic methods.
  • the matrix materials comprises matrix derived from living cell sources.
  • the matrix materials comprises matrix derived from mixed components of individual matrix elements.
  • the matrix materials comprises matrix derived from decellularized matrices of cadaver tissues.
  • the step of introducing the cell population, matrix materials, growth factors or pharmaceuticals into the sub-compartment is affected prior to the step of implanting the device to next to the micro vasculature of the subject.
  • the step of introducing the cell population, matrix materials, growth factors or pharmaceuticals into the sub-compartments is affected following the step of implanting the device next to the micro vasculature of the subject.
  • the bioreactor also provides a subcutaneously implanted device that provides a system allowing multiple punctures through the surface skin into the device. This will provide subcutaneous access to cells and tissues within the bioreactor for infusion of additional cells, growth factors, matrix components or pharmaceuticals as needed.
  • biosensor system with fluidic or other connection to each of the compartments in the bioreactor as well as connection to the interior body compartment also adjacent to the device.
  • the biosensor system includes capabilities for detection of biomarkers molecules indicative of normal or abnormal states of cells within the device, or from within the internal body that is also adjacent to the bioreactor.
  • the biosensor system includes capabilities for wireless transmission of biomarkers from within the device to electronic devices outside the body to facilitate detection and care of tissues within the device, and as a diagnostic tool for normal and diseased biological processes occurring in the body, and as a diagnostic to detect the influence of other interventions on the body be they pharmaceutical or otherwise.
  • This example provides a murine platform comprising four elements: a container, a container cap, an installation device, and a mouse microscopy stage.
  • the specific design of the device can be found in Figures 9A-9C.
  • the container is made of biocompatible material (e.g., PMMA, MED610 or Grade 316 Surgical Steel); see for example, Figures 9-11 A, 10 and 6B. It is implanted intradermally on the back of a mouse with the sides of the container in direct contact with the underside of the dermal layers of the skin.
  • the sides of the container include 50-200-micron mesh holes to facilitate tissue growth from the skin into the internal reservoir, through the mesh holes.
  • the container features a wide rim at the base below the holes and under the skin to hold it in place once implanted.
  • the container houses a reservoir of approximately ⁇ of gelatinous protein mixture (matrix).
  • the container is envisioned to feature fluidic injection ports that are compatible with standard pipet P200 and/or PI 000 tip used to introduce material into the reservoir at volumes between 20-200 ⁇ 1.
  • the injection ports are used initially to completely fill the reservoir with the matrix mixture after the container has been implanted so that the matrix is in full contact with the mesh holes on the side of the container and there are no air bubbles within the reservoir. Subsequently, injection ports may be used to introduce additional materials into the matrix.
  • the injection ports will introduce materials into the matrix at different locations on the x and z axes. The injection ports must not allow materials to leak back out of the reservoir through the ports. It is envisioned that subsequently introduced material will rinse/wash materials in the matrix out into the surrounding tissue through the mesh holes.
  • a No. 1 coverslip glass between 0.13-0.17 mm in thickness (e.g., element 10-c in Figure 9B) and having a diameter of about 8 mm, which is flush against the matrix when the reservoir is filled, with no air bubbles, and is held in place by a retaining clip (or circlip; e.g., element 10-a in Figure 9B) or equivalent and sealed with an O-ring or equivalent to prevent leaks (e.g., element 10-b in Figure 9B). Tissue growth will occur immediately beneath the covers lip. Opening the covers lip is envisioned to accommodate removal of the entire matrix through the top of the container.
  • the visible top surface of the container contains a stable registration element that can be used as a point of reference for microscopy, facilitating repeat imaging of the same microscopic location in serial imaging sessions. This could be implemented as a dot or indentation on the visible top surface of the container.
  • a mechanical interface Built into the design of the container is a mechanical interface to allow attachments on top of the container above the coverslip glass.
  • the coverslip and the registration element must be unobstructed by the mechanical interface to facilitate imaging the live tissue in some attachment applications.
  • attachments include a cap to protect the coverslip glass, a stage for microscopy, an irradiation system.
  • the container cap attaches to the top of the container via the container's mechanical interface and protects the coverglass slip. Its top outside surface includes space for a registration label.
  • the mechanism of implantation of the container into the skin envisioned is a grommet-style 'hole punch' rivet system using an external manual tool.
  • the mouse microscopy stage is envisioned to comprise a stable base upon which the mouse will stand/lay with an element that will attach the base above the mouse to the implanted intradermal container via the container's mechanical interface thereby stabilizing the container against mouse movement during microscopy.
  • the coverslip and the registration element must be unobstructed by the stage and the mechanical interface.
  • Implantable devices were prepared and soaked in 100% ethanol for 20 minutes before drying within a tissue culture hood. Frozen Matrigel reagent was thawed at 4 degrees Celsius, stored on ice, and then mixed with 175 ng/mL basic Fibroblast Growth Factor in 5% bovine serum albumin prior to injection into the device.
  • the device has a 12 mm No.1 glass covers lip placed into the top aspect of the device, and is closed with a retainer clip (e.g., Figures 9A-9C).
  • a one-centimeter incision was made in the skin at the location of implantation on the recipient mice near at the dorsal aspect of the chest. Each device was positioned in a subcutaneous pocket and closed using a circular synch 5-0 non-absorbable Proline suture. Alternately, the device was implanted surgically, and internal biochemical matrix was then added through the top opening of the device, before closure with the coverslip and retaining ring (e.g., Figures 10A and 10B).
  • mice were transferred to a custom made plexiglass anesthetic container and placed on a fluorescent microscope stage for imaging, (e.g., Figures 11 A-l ID) Animals were examined in initial tests for 0.5-1.0 hr. Once an initial test was established, 20 minutes of anesthesia was sufficient for imaging.
  • Figures 1 IB and 11C depict a system for live imaging which include an animal/mammal observation station, a microscope and a computer system for collecting, depicting and processing the image data.
  • Plastic Subcutaneous Model This model was encased in nylon lOOum mesh. The device underwent full subcutaneous implant, which allowed ex vivo analysis post-sacrifice. The device was able to remain in animal indefinitely.
  • Figures 12A through 12C illustrate the results of vessel growth using this prototype.
  • Modified Plastic Model This model was used for multiple endpoint analysis. Methodologies for removal of implant were developed for this model to allow utilization of fluorescent microscopy, implant digestion and rapid analysis by flow cytometry, and microCT analysis for significant improvement of data acquisition from the device.
  • FIG. 12D through 12E illustrate the results of cell and vessel growth using this prototype.
  • Galvanized Steel Model This was the first stainless steel model that was implanted in live animals (e.g., Figures 13A and 13B; Figures 9A-9C).
  • the device provides grooves for skin attachment, an encasing for 12mm coverglass and retaining clip. Proline, ethylon and vicryl sutures were tested for implantation. The device remained in animal for up to 4 weeks.
  • Surgical Steel Prototype This model was made with substantially harder material, which was more difficult to mill (e.g., Figures 13C and 13D, Figures 9B and 9C).
  • the device provided an enlarged bottom groove lip to retain position under the skin once implanted. It also provided an encasement notch for an 8mm coverglass and a retaining clip.
  • the device was fully biocompatible and required less matrix material for improved cost effectiveness.
  • Smaller 8mm Surgical Stainless Steel Model This model provides a smaller design from previous 12mm implants, which reduced weight and size of device (e.g., Figure 13E).
  • AutoCAD design prepared for biocompatible lighter multi-well and rendered 3D printed prototypes possible (e.g., Figures 2A-2C). This fabrication process allowed for more rapid prototyping and integration of smaller features.
  • Model with Surgical Interface Improvements This model is smaller and lighter (e.g., Figure 13F). It provided an enlarged bottom groove lip to retain position under the skin once implanted. The device also provided encasement notch for an 8mm cover- glass, a retaining clip and a screw clip with sealant. This design required less matrix material for improved cost effectiveness. When implanted, first live blood vessels were visible in transgenic mice (e.g., Figure 1 ID).
  • Lasercut Acrylic Prototype Lasercutting methods were used to create chamber in clear acrylic. Fabrication by this type of technology produced very nice clear implants.
  • Figure 13G depicts exemplary laser rings and baseplate template provided to the laser cutter and used to produce the Lasercut Acrylic Prototype.
  • hydrogels as a matrix component were added and could go inside the implants; for example, human synthetic matrixes offer the advantages of not causing damages to the host immune system. Additions could be made to the window coverglass surface.
  • the retainer ring for closing the top of the implant could be replaced with part of a coil of a spring.
  • Commercial glue products such as 'fish tank glue' were used to close the coverglass seal, which did not appear to cause any problems with immunity and rendered an airtight seal in the device chamber.
  • FIG. 15 A live vessels were clearly seen here in black at low power. The rim of the implant is also visible in the shot. The brightest white in the image is fluorescent cells that are building the new blood vessel. There is also a clear fluid bubble just below the vasculature indicating their leaky edematous state as expected.
  • Figure 15B depicts a closeup view of the branching capillary inside live chamber with GFP positive cells nearby in white.

Abstract

Provided are implantable devices for maintaining cell growth, development and differentiation within an implantable device in a live mammal under a vascularized environment. Also provided are systems and methods of using such implantable devices for various biochemical applications, such live imaging, live biological and biochemical assays, drug testing, implantation and etc. Further provided are integrated systems using such implantable devices for data acquisition, e.g., images, videos and biochemical responses.

Description

IMPLANTABLE DEVICES AND APPLICATIONS AND USE THEREOF
1. CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. Provisional Application No.
61/591,751 filed on January 27, 2012; U.S. Provisional Application No. 61/592,103 filed on January 30, 2012; and U.S. Provisional Application No. 61/618,485 filed on March 30, 2012, each of which is hereby incorporated by reference herein in its entirety.
2. FIELD OF THE INVENTION
[0002] Provided herein are implantable devices that can maintain the growth, development and differentiation of mammalian cells within an implantable device in a live mammal under a vascularized environment.
[0003] Also provided herein are systems and methods of using such implantable devices for various biochemical and therapeutic applications, such live imaging, live biological and biochemical assays, drug testing, tissue/organ implantation, and etc. Further provided herein are integrated systems using such implantable devices for data acquisition, e.g., images, videos and biochemical responses.
3. BACKGROUND
[0004] Developments have been made to maintain mammalian cell cultures by mimicking the natural cellular environments of live mammals. Growth, development and differentiation of mammalian cells, however, can require a complex and delicate system balancing numerous factors. Improvements are still necessary, especially after tissue implant/transplant and stem cell treatments have been developed.
[0005] There is thus a widely recognized need for systems and apparatuses that can allow cells to be housed within a mammal, thereby maintaining an immuno-privileged biochemical environment that is as close as possible to the desired mammalian environment.
4. SUMMARY OF THE INVENTION
[0006] In one aspect, provided herein is a system for maintaining mammalian cells in a live mammal, e.g., for growth, development and/or differentiation. An exemplary system comprises: i) an implantable device and ii) a plurality of cells contained within a compartment of the implantable device, where the plurality of cells are suspended in a biochemical composition comprising a cellular matrix, wherein the maintenance of the plurality of cells is supported partially by the biochemical composition. The implantable device in turn comprises: a housing having a plurality of sides, wherein one side of the plurality of sides comprises a structural feature for securely attaching the implantable device to a site of implantation in the mammal; a compartment contained within the housing, once implanted in the mammal, the compartment is capable of exchanging content, via an interaction module on at least one side of the plurality of sides, with an extracellular space or a tissue microvasculature of the live mammal; and an observation module, which is located on one side of the plurality of sides through which the plurality of cells can be observed by an optical equipment. In some embodiments, one or more growth factors are also included in the biochemical composition.
[0007] In some embodiments, the structural feature for securely attaching the implantable device is a groove on one side of the plurality of sides of the implantable device.
[0008] In some embodiments, the structural feature for securely attaching the implantable device is a plurality of holes on at least one side of the plurality of sides of the implantable device.
[0009] In some embodiments, the implantable device further comprises a plurality of sub-compartments within the compartment, and the plurality of cells is contained within at least one sub-compartment of the plurality of sub-compartments. In some embodiments, sub- compartments within the plurality of sub-compartments are connected via one or more channels, and where at least one of the connected sub-compartment contains the plurality of cells. In some embodiments, the connected sub-compartments, upon exposure to one or more biochemical reagents, are configured to perform an assay on the plurality of cells, or blood or plasma products coming in contact with the device. In some embodiments, the assay detects the presence or absence of a biomarker, and wherein the biomarker is selected from the group consisting of a protein, a peptide, a gene, and a nucleic acid molecule.
[0010] In some embodiments, the implantable device further comprises an injection port on one side of the plurality of sides, through which biochemical reagents can be added into or removed from the compartment. In some embodiments, the injection port and observation module are on the same side of the plurality of sides.
[0011] In some embodiments, the interaction module comprises a permeable membrane between an opening on the at least one side of the plurality of sides of the implantable device. In some embodiments, the interaction module is an opening on the implantable device, which allows content within the device to exchange with the cellular environment where the implantable device is implanted.
[0012] In some embodiments, the implantable device further comprises a wireless module for transmitting a signal from the implantable device to an external receiver or receiving a signal from an external controller, after the implantable device is implanted in a mammal. In some embodiments, the wireless module is for transmitting a signal from the implantable device to an external receiver and receiving a signal from an external controller.
[0013] In another aspect, provided herein is an imaging system for collecting data from a live mammal. An exemplary imaging system comprises 1) any one or combination of the system for maintaining growth and development of mammalian cells in a live mammal, as described herein; and 2) optical equipment for collecting images or video data of the content with the compartment or sub-compartments of the implantable device. In some
embodiments, the optical equipment is selected from: a microscope, a fluorescence imaging device, a thermal imaging device, a radio imaging device, or a combination thereof. In some embodiments, the imaging system further comprises: a computer system for collecting, storing, displaying, and processing the image or video data. In some embodiments, the data collected comprise images or videos of the plurality of cells in the implantable device.
[0014] In another aspect, provided herein is a method for collecting data from a live mammal using an imaging system. An exemplary method comprises the steps of: i) maintaining the growth of a plurality of cells in a live mammal using a mammalian cell system as described herein, where the implantable device is implanted in a live mammal such that at least a portion of the observation module is not embedded within the live mammal and wherein data of the content within the implantable device are measured using optical equipment; and ii) collecting data of the content within the implantable device using optical equipment. In some embodiments, the optical equipment is selected from: a microscope, a fluorescence imaging device, a thermal imaging device, a radio imaging device, or a combination thereof. In some embodiments, the imaging system further comprises: a computer system for collecting, storing, displaying, and processing the image or video data. In some embodiments, the data collected comprise images or videos of the plurality of cells in the implantable device.
[0015] In another aspect, provided herein is a drug testing system. An exemplary system comprises a mammalian cell system as described herein that comprises a plurality of sub-compartments where each sub-compartment of the plurality of sub-compartments comprises a population of cells. The cells are taken from a patient and a compound is added to one or more sub-compartments of the plurality of sub-compartments.
[0016] In another aspect, provided herein is a method for drug testing using a mammalian cell system as described herein that comprises a plurality of sub-compartments. An exemplary method comprises the steps of: placing, in each sub-compartment of the plurality of sub-compartments of the implantable device, a population of cells, wherein the cells are derived from a biopsy sample of a target disease, and wherein the implantable device is implanted in a live mammal; adding a test compound in one or more sub-compartments of the plurality of sub -compartments; and collecting data from each sub-compartment of the plurality of sub-compartments.
[0017] In some embodiments, the method further comprises the steps of:
comparing data from different sub-compartments to determine the presence of differences between the sub-compartments; and correlating any differences with the presence of the compound in each sub-compartment. In some embodiments, the biopsy sample is associated with a type of cancer. In some embodiments, each sub-compartment of the plurality of sub- compartments contains the same type of cells and wherein cells in each sub-compartment have approximately the same quantity. In some embodiments, at least two different compounds are added to two different sub-compartments.
[0018] In another aspect, provided herein is a method for producing a desired biomolecule in a live mammal using a mammalian cell system provided herein. An exemplary method comprises the steps of: maintaining the growth of the plurality of cells in the live mammal using the implantable device, wherein the implantable device is implanted in a live mammal and wherein cells of the plurality of cells are capable of producing the desired biomolecule; adding one or more reagents to the implantable device, wherein the reagents are necessary for the plurality of cells to produce the desired biomolecule; and producing the desired biomolecule. In some embodiments, the cells are pancreas cells and the desired biomolecule is insulin.
[0019] In another aspect, provided herein is a method for growing cells or tissues for transplant in a live mammal using a mammalian cell system provided herein. An exemplary method comprises the steps of: maintaining the growth of the plurality of cells in the live mammal using the implantable device, wherein the implantable device is implanted in a live mammal such that at least a portion of the observation module is not embedded within the live mammal and wherein data of the content within the implantable device are measured using an optical equipment, and wherein cells of the plurality of cells are selected from the group consisting of stem cells, embryonic stem cells, adult stem cells and non-stem cells; measuring, via the observation module, data of the cells of the plurality of cells to determine whether the cells are suitable for transplant; removing the cells from the implantable device if the cells are suitable for transplant; and transplanting the cells in a mammal.
5. BRIEF DESCRIPTION OF THE DRAWINGS
[0020] Those of skill in the art will understand that the drawings, described below, are for illustrative purposes only. The drawings are not intended to limit the scope of the present teachings in any way.
[0021] Figures 1A-1D illustrate exemplary embodiments; A) a round shape device, B) a triangular shape device, C) a fan shape device, and D) a convex curve shape device.
[0022] Figures 2A-2C illustrate different views of an exemplary embodiment with multiple sub-compartments.
[0023] Figure 3 illustrates a lab-on-a-chip design of implantable design with a miniaturized handling system.
[0024] Figure 4 illustrates a cylinder holding system for an exemplary implantable device.
[0025] Figure 5 illustrates an injection port system of an exemplary implantable device.
[0026] Figure 6 illustrates functional sub-compartments within an exemplary implantable device.
[0027] Figure 7 illustrates an exemplary tool for implantation.
[0028] Figure 8 illustrates an exemplary personalized cancer/tissue assay.
[0029] Figures 9A-9C illustrate an exemplary implantable device.
[0030] Figures 10A andlOB illustrate mice implanted with exemplary implantable device and control mice without the implant, A) Implant within two mice and non-implanted cagemate.
[0031] Figures 11A-11D illustrate an exemplary embodiment of live imaging system.
[0032] Figures 12A through 12E illustrate exemplary cell growth within implantable devices. [0033] Figures 13A through 13F illustrate exemplary implantable devices. Figure 13G depicts exemplary laser rings and baseplate template for creating lasercut acrylic device.
[0034] Figures 14A through 14B illustrate exemplary data from implantation study showing cell growth but no evidence of blood vessels.
[0035] Figures 15A through 15F illustrate exemplary data from implantation study showing blood vessels.
6. DETAILED DESCRIPTION OF THE INVENTION
6.1 Definitions
[0036] Unless otherwise noted, terms are to be understood according to conventional usage by those of ordinary skill in the relevant art.
[0037] As used herein, the term "implantable device" refers to an article with a main body or housing, which contains at least one structural element (e.g., compartment) that can be used to grow mammalian cells and conduct biochemical reactions while the device is implanted in a live mammal. It is sometimes used interchangeably with "implantable bioreactor." Additional structural elements such as sub-compartments can be found in the compartment.
[0038] As used herein, "biocompatible" refers to a property of a material, which is characterized by it, or its physiological degradation products, being not, or at least minimally, toxic to living tissue; not, or at least minimally and reparably, otherwise injurious living tissue; and/or not, or at least minimally and controllably, causative of an immunological reaction in living tissue.
[0039] As used herein, the term "biocompatible material" refers to materials, natural or synthetic, that can be used to construct an implantable device. When implanted in a mammal, the materials forming the implantable device do not cause serious discomfort or adverse physiological reactions (such as adverse immuno response or rejection) of the mammal.
[0040] As used herein, the term "biodegradation" refers to any means by which a polymer can be disposed of in a live mammal (e.g., in a patient's body), which includes bioabsorption, resorption, etc. Degradation occurs through hydrolysis, chemical reactions, or enzymatic reactions. Biodegradation can take place over an extended period of time, for example over 2-3 years. The term "biostable" means that the polymer does not biodegrade or bioabsorb under physiological conditions, or biodegrade or bioabsorb very slowly over a very long period of time, for example, over 5 years or over 10 years.
[0041] As used herein, the terms "compartment" and "sub-compartment" refer to a structure in an implantable device that is separated from the external environment or other structures within the device. Examples of compartments and sub-compartments include chambers, channels, wells and etc.
[0042] As used herein, the term "biochemical composition" refers to a mixture of small molecules and macromolecules within a compartment or sub-compartment of an implantable device, which can support the growth, development, and/or differentiation of mammalian cells. Exemplary components of a biochemical composition include but are not limited to growth factors, cellular matrices, and buffers.
[0043] As used herein, the term "biomarker" refers to a substance used as an indicator of a biological state. A biomarker can be a small molecule as well as a
macromolecule, or a signal associated with a particular small molecule or macromolecule. In medicine, a biomarker is a term often used to refer to a protein measured in blood whose concentration reflects the severity or presence of some disease state. More generally, a biomarker is anything that can be used as an indicator of a particular disease state or some other physiological state of an organism. In cell biology, a biomarker is a molecule that is present or absent from a particular cell type. Biomarkers facilitate the characterization of a cell type, their identification, and eventually their isolation. A biomarker can also be used to identify a cell population, make a diagnosis, or measure the progress of a disease or the effects of a treatment. In some embodiments, the terms "biomarker" and "biomolecules" are used interchangeably.
[0044] As used herein, the term "optical equipment" refers to an apparatus or device that has one or more sensors for collecting one or more optical signals. The optical signals can be of any wavelength, visible or invisible.
6.2 Implantable Device: Design and Making
[0045] Provided herein are implantable devices useful for maintaining cell growth, development and differentiation within a live mammal under a vascularized environment. Also provided herein are methods for designing and making the implantable devices.
6.2.1 Design of the Main Body or Housing of an Implantable Device [0046] In one aspect, an implantable device comprises a main body or housing containing at least one structural element (e.g., a compartment or sub-compartment) surrounded by one or more sides. Exemplary embodiments of the implantable devices are depicted in Figures lA-1 ID.
[0047] In one aspect, the main body or housing provides the shape and support for the structural elements therein. In another aspect, one or more functional or structural modules can be attached to the main body or housing of the implantable device.
[0048] In some embodiments, the main body or housing of the implantable device is formed by a plurality of sides, including, for example, a top side and a bottom side.
Referring to Figure 1A, device 100 has a top side-element 10 and a bottom side-element 20, which are joined through a cylindrical or tubular connecting side-element 15. Compartment 30 here refers to the space formed by the top side-element 10, the bottom side-element 20, and the cylindrical or tubular connecting side-element 15. In this example, the compartment is formed by sides of the main body or housing. An interaction module 40, which allows the content within implantable device 100 to interact with the external environment when the device is implanted in a mammal, such as a rodent or a human. In the exemplary
embodiment, the top side and bottom side are round. In this embodiment, the connecting side element has a tubular or cylindrical shape, similar to that of the side of a barrel. It will be understood that the sides can be of any sizes and shapes, forming any shape that is suitable for the intended purpose of a particular implantable device.
[0049] Here, the shape of a compartment is sometimes defined by the shape of a cross-section of compartment 30. As an example, a cross section can be taken along a plane determined by the x-x' and y-y' axes, which is between the top side-element 10 and bottom side-element 20, as illustrated in Figure 1 A. The resulting plane can be round, oval, semicircular, square, rectangle, triangle or any other shapes. Various exemplary embodiments of device 100 are shown in Figures 1A-1D. In these exemplary embodiments, the compartment is defined by the main body or housing itself. In some embodiments, a separate structural device such as a pre-made insert can be used to form the compartment, thus allowing the compartment to have a shape that is different from that of the main body or housing.
[0050] In some embodiments, the main body of an implantable device has a tubular or cylindrical shape, with a round cross-section (e.g., Figure 1 A). In some embodiments, compartments or sub-compartments within a round device can adopt different shapes such as oval, semi-circular, square, rectangle, triangle and etc. Advantageously, round implantable devices are suitable for concentric cell growth inwards from all directions so the greatest volume of growth can occur at one time. Also advantageously, such implantable devices are compatible with implantation on or into skin. Once inserted into the skin, the skin around the circumference of the cylindrical or tubular device can be easily closed to reduce or eliminate irritation to implanted subject.
[0051] In some embodiments, the main body of an implantable device is box-like with a square or rectangular cross-section. In some embodiments, compartments or sub- compartments within the box-like device also have square or rectangular cross-sections. Advantageously, multiple sub-compartments of the same or similar shapes but smaller in size can be created within a square or rectangular compartment. Advantageously, more quantifiable measurements can be taken between different individual implants.
Advantageously, more quantifiable measurements can be taken between sub-compartments within the same device. Further advantageously, the measurements of sub-compartments within the same device can be compared relatively easily to each other to assess
characteristics such as the distance of growth of cell populations and complexity of tissue formation along the gradient of growth. Also advantageously, compartments having a square or rectangular cross-section are more suitable for data observation. For example, when a microscope is used to collect data through an observation window, a rectangular cross-section corresponds to a longer distance in the 'field of view' than those of other shapes. As tissue grows, it can be studied along the longer distance of the rectangle under the coverslip.
[0052] In some embodiments, the main body of an implantable device is a triangular-shaped block, with a triangular cross-section (e.g., Figure IB). In embodiments where compartments and sub-compartments also adopt a triangular shape, cell growth can start at one corner and move towards a broad face. Alternatively, cell growth can start at a broad face and move towards a corner. An exemplary embodiment is depicted in Figure IB. Advantageously, such embodiments can be used to study the impact of increasing tissue density on tissue performance and growth. Advantageously, they also can be used to examine tissue in response to varying concentrations of growth factors. For example, if a cellular matrix containing a particular growth factor is placed at a corner, there can be a gradient of concentrations away from the corner. Also advantageously, such embodiments can be used to examine the reaction of tissues as they grow away from a corner that contains high concentration to an area of low concentration.
[0053] In some embodiments the main body of an implantable device is an irregular block, with a fan-shaped cross-section. Referring to Figure 1C, the fan-shaped compartment is triangle-like with a curved side. In some embodiments, the curved side is opposite to a growth corner and there are multiple parallel compartments. Advantageously, such embodiments can be used to examine tissues growing from a single origin in response to different concentrations or types of stimuli that have been put at the end of the radiating wells.
[0054] In some embodiments, the main body or housing of an implantable device and compartments or sub-compartments therein have a cross-section with convex curves (Figure ID). In some embodiments, the convex curves are positioned during implantation along the body of a mammal, such as around its back or abdomen (e.g., Figure ID).
Advantageously, such embodiments can better 'fit' the natural contours of the body when implanted.
[0055] In some embodiments, an implantable device and the compartments or sub-compartments therein are custom-molded to fit the location of a desired implantation. For example, an implantable device and the compartment therein can be designed to fit a side of the liver or pancreas of a recipient. In some embodiments, it is advantageous and spatially efficient to have compartments or sub-compartments that have a shape similar to the main body of the implantable device. In some embodiments, the compartments and sub- compartments do not need to adopt the shape of the main body or housing of the implantable device. For example, a cylindrical or tubular implantable device can in fact house a compartment of rectangular, square or even triangular shape.
[0056] Advantageously, custom-molded implantable devices can better fit the natural contours of the body when implanted. Also advantageously, it is possible to attach the implantable device near a target organ. In such embodiments, pre-implantation data and planning are needed, for example, using CT scanning to analyze the morphology of the target organ, before the customized implantable devices are designed and produced.
[0057] In some embodiments, sub-compartments within the same compartment have the same or a similar shape as well as functionalities (see, for example, Figures 2A-2C). In such embodiments, more quantifiable measurements can be taken between different individual implants. In some embodiments, more quantifiable measurements can be taken between sub-compartments within the same implant. In some embodiments, measurements of the sub-compartments within the same device can be compared relatively easily to each other for assessing characteristics such as the distance of growth of cell populations and complexity of tissue formation along the gradient of growth.
[0058] In some embodiments, sub-compartments within the same compartment have the same shape but different functions (e.g., Figure 3). In some embodiments, sub- compartments within the same compartment have different shapes and different functions. For example, some sub-compartments can be used for storing biochemical reagents (e.g., growth factor, buffer, serum, or commercially available growth media such as Matrigel™ from Becton, Dickinson and Company etc.); they accordingly have relatively large volumes and are also called wells or storage depots. In some embodiments, some sub-compartments are used as a container where a biochemical reaction or synthesis takes place. Such sub- compartments are also called chambers or reaction chambers. Additionally, an implantable device can have one or more structural elements that are used for delivering biochemical reagents between sub-compartments. Such structures are also called channels. In some embodiments, one or more wells, chambers, storage depots and channels form a network of sub-compartments, which can be used to achieve sophisticated biochemical synthesis or to carry out a specific biochemical reaction.
[0059] In some embodiments, the shape of the main body or housing of an implantable device can be modified or altered to facilitate secure attachment between the device and the site of implantation. For example, in certain embodiments, the main body or housing has one or more sharp protrusions, e.g., the sharp corners on a triangular, rectangular or the fan-like device (e.g., Figures IB-ID), which can render it difficult for implantation. It can be challenging to create smooth sutures around such sharp protrusions and there can be impeded pulling force on the surrounding tissues to maintain the curve in the device. For example, in some embodiments, when sutures run around a corner, the pressure on the sutures will cause the area near the sharp edge to dig into the skin leaving the adjacent areas relatively loose. In some embodiments, sharp protrusions on the implantable devices are modified to a smoother shape (e.g., rounded or curved) to create better skin contacts during implantation.
[0060] In some embodiments, additional structural features are created on one or more sides of the implantable device to facilitate secure attachment between the device and the site of implantation. For example, as depicted in Figures 2A-2C, a convex groove is created around the circumference of the side of the device to create a snug attachment of one or more sutures that is also running in a circumference around an opening in the skin. In some embodiments, holes are implemented on the side of an implantable device to facilitate skin closure by sutures.
6.2.2 Functional Modules [0061] In some embodiments, the main body or housing of an implantable device comprises one or more sides. In some embodiments, one side comprises an observation module through which the content within the device (e.g., in a compartment or sub- compartment within the compartment) can be monitored or observed by one or more types of optical equipment. In some embodiments, the observation module comprises an observation window. The observation window can be as large as one of the sides, as illustrated in Figure 1 A. In some embodiments, the observation window can be opened and/or removed from the main body or housing such that the content of the compartments or sub-compartment can be replenished or removed. Alternatively, new material can be added through the observation window to the compartment or sub-compartments. In some embodiments, the main body or housing comprises separate modules through which material can be removed or added.
[0062] In some embodiments, an observation window is an integrated part of the device. In some embodiments, there are multiple windows that cover different compartments of the device. For example, the observation windows covering the cell-filled compartments or sub-compartments will be more suitable for transmission of optical signals (e.g., clear glass). In contrast, parts of the observation window that do not cover the cell-filled compartments or sub-compartments will not be as suitable for transmission of optical signals (e.g., formed by solid or opaque materials).
[0063] In some embodiments, optical equipment, such as a microscope or micro video camera, is used to collect optical signals of the sample within the implantable device through the observation window. In some embodiments, a camera is fitted on the optical equipment directly or to the implantable device directly. In some embodiments, the camera is an integrated part of the microscope. In some embodiments, the video camera can capture what comes through the lenses as well as transmit the captured signals to a computer screen. In some embodiments, multiple lenses and / or cameras can be used. In some embodiments, the glass forming the observation window is of different thicknesses as required for different microscopy needs (e.g., No. 1-5 thickness) or thicker to allow greater stability.
[0064] In some embodiments, the implantable device has one or more features that permit the device to be connected to the lens of a microscope using specific notches or shapes. This allows for immobilization of the device for microscopic analysis. Preferably, these features are located on the top side of the device. In some embodiments, the connection can be achieved by a simple notch or lock-and-key mechanism on the device that fits into a notch on the microscope stage or lens. In some embodiments, a recipient of the implantable device (e.g., a rodent) can be positioned within a specific holder (e.g., a cylinder) with an opening allowing immobilization, as in Figure 4. This holder can also allow anesthesia during an imaging process. In some embodiments, in order to facilitate repeated imaging at high magnification, the notch or lock-and-key mechanism also provides registration marks for the microscope system to guide and automate its imaging of identical or desirable locations and fields of view of a given region of the device in a given recipient on a real time basis as often as desired (in a living mammal). In some embodiments, registration marks on the cover glass can provide orientation and registration to the microscope.
[0065] In some embodiments, video signals are collected of the content within a compartment or sub-compartment, using, for example, a CCD camera. In such embodiments, there are known computer hardware and software programs that can facilitate automated cataloguing of each recipient, each implanted device, each location of interest within an implanted device, thereby allowing rapid image acquisition and analysis.
[0066] In some embodiments, data collection can be automated by software that controls the movement of the microscope and camera, or the sample (device on a mammal). For example, in devices containing multiple sub-compartments that are fixed to the main body of the device, an automated software program can guide a lens of a microscope (or a video camera attached thereto) from one sub-compartment to another sub-compartment in a pre-defined order. In some embodiments, the program also controls the length of time during which data are collected from each sub-compartments. In some embodiments, data are also collected from other parts of the device, for example, from the channels, chambers, wells or any other parts. In some embodiments, the device can be moved while the optical equipment stays stationary. For example, an observation platform (upon which a mammal is placed) can move in a controlled manner to permit position-specific and time-specific data collection.
[0067] In some embodiments, a detachable observation window is held to the main body or housing using a retaining ring (or "Circlip"). In some embodiments, an insulating O-ring is used between the retaining ring and the cover glass.
[0068] In some embodiments, an observation window includes a cover slip that is protected by a fixed lid-like element to protect the glass from scratches, damage, sunlight, or compromising sterility of the implant. In some embodiments, the pressure within the compartments or sub-compartments is controlled, using, for example, the lid-like element. In some embodiments, one or more fasteners with tension screws are attached to the lid-like element such that a specific pressure can be applied upon the content within the
compartments or sub-compartments. It has been known that pressure can change growth and differentiation behaviors. In some embodiments, the lid-like element contains electrodes that pass beneath the coverslip and allow an electrical current or fluid to be added to the internal compartment, which can all influence cell growth and differentiation. In some embodiments, the lid-like element is fixed onto the device (e.g., using screws) in a circular fashion. In some embodiments, the lid-like element also contain an option to activate a cutting surface that, when turned in the circular screw motion, cuts out the content of the device and allows it to be severed from connection to the body and removed. Replacement matrix can be added to re-grow new tissue.
[0069] In some embodiments, one side of the main body or housing of the implantable device also contains an interaction module (e.g., element 40 in Figure 1 A), as an integrated part of the implantable device. The interaction module allows a compartment sub- compartment of the device to interact with existing extracellular space and tissues near where the device is implanted.
[0070] In some embodiments, the interaction module comprises an open entrance port to the implantable device such that the content within the compartment or sub- compartment can directly interact with the outside cellular environment. In some
embodiments, the implantable device comprises multiple open entrance ports that allow different sub-compartments to interact with the outside environment (Figures 2A-2C). In such embodiments, more rapid tissue growth can be obtained because there is no interference from non-biological material. Such embodiments also permit more complete interaction with the native tissue environment outside the device. In some embodiments, the content in the compartments can leak out of the device. For example, gels or semi-solids may move out of the device into the surrounding tissues.
[0071] In some embodiments, the size of an interaction module can be varied to control the speed at which the implantable device exchanges content with the outside environment. In some embodiments, multiple interaction modules can be used to allow more even distribution of content exchange at various locations of the device.
[0072] In some embodiments, an interaction module (e.g., an opening covered with mesh or net) can be located near an attachment groove for ease of construction of the device. In some embodiments, the interaction module can be located on the connecting side of the device. In some embodiments, the interaction module can be located on the bottom of the device.
[0073] In some embodiments, the interaction module comprises one or more open entrance ports that are covered with a mesh or net made of synthetic or natural material. For example, the mesh or net can be made of nylon with pores of various sizes (e.g., between 10 μηι and 500 μηι). In some embodiments, the mesh or net is formed with biocompatible or biodegradable material. In some embodiments, the same biocompatible or biodegradable material used to construct the main body of the implantable device can be used to form the mesh or net. Exemplary materials are disclosed below in connection with the making of the implantable devices.
[0074] Advantageously, the mesh or net covering the entrance port provides support for biochemical reagents within the device during implantation and before surrounding tissue invasion. In some embodiments, the mesh or net covering the entrance port is a physical barrier for cell entry or escape. In some embodiments, the mesh or net covering the entrance port can limit blood vessel sizes. In some embodiments, the mesh or net covering the entrance port can also limit potential constraints on oxygenation and waste removal, or cell product release from the implantable device.
[0075] In some embodiments, an injection port can be found on one or more sides of the implantable device for injecting material into the compartment or compartments, or interacting with a microfluidic delivery system. In some embodiments, the microfluidic delivery system is embedded within the implantable device. In some embodiments, the microfluidic delivery system is external to the implantable device. In some embodiments, an injection port system allows removal and introduction of materials (cells, fluids, compounds such as growth factors or drugs) into the implantable device. In some embodiments, one or more injection ports are created within the lid or on the sides of the implantable device. In some embodiments, an injection port is built into the lid itself. In some embodiments, an injection port is built into the sides of the implantable device. Figure 5 illustrates an exemplary injection port system from the top surface of the implantable device.
6.2.3 Internal Structure Elements within the Device
[0076] An implantable device as disclosed herein can house numerous internal structural elements, including compartments, sub-compartments within a compartment, channel-like structures connecting compartments or sub-compartments. Compartments and sub-compartments are used for numerous purposes, including as reaction chambers, storage depot or wells.
[0077] In some embodiments, sub-compartments can be of the same size and shape, or of different sizes and shapes. In some embodiments, sub-compartments have the same shape as the compartment (or implantable device). In some embodiments, sub- compartments have shapes that are different from the compartment (or implantable device). [0078] In some embodiments, the main body of the implantable device itself forms all or parts of the boundaries of the compartment or sub-compartments. In some embodiments, the compartments or sub-compartments are created by inserts or molds that are pre-formed, using the same material as that of the main body of the implantable device. In some embodiments, the compartments or sub-compartments are created by inserts or molds that are pre-formed using different materials from that of the main body of the implantable device.
[0079] In any given implantable device, the number of sub-compartments is determined by the purpose and intent for which the device is used. In some embodiments, the implantable device is used to produce biomolecules for treatment or tissues for
transplantation. In some embodiments, devices with a single compartment are used. In some embodiments, devices with a few large size sub-compartments are used. Advantageously, a larger volume of cell material can be obtained for experimentation, or to be used as transplantable new replacement tissues. Greater doses of cell products can be generated and released into the body; and greater volume of storage depot for drugs that can be released into the body.
[0080] In some embodiments, implantable devices containing multiples sub- compartments with equal or different sizes are used. Advantageously, multiple sub- compartments allow maintenance of multiple cell populations. Multiple growth factor conditions and extracellular matrix compositions can be analyzed simultaneously in parallel.
[0081] In some embodiments, implantable devices containing multiples sub- compartments are used as a therapeutic delivery system. Advantageously, multiple sub- compartments allow cellular products of different types to be maintained within the unit and provide different biological products to a recipient as needed (e.g., as personalized medicine). In some embodiments, the same quantity of a therapeutic reagent is stored in different sub- compartments. In some embodiments, different quantities of the same therapeutic reagent are stored in different sub-compartments. In some embodiments, multiple sub-compartments containing one or more therapeutic agents are designed to release their content in a sequential manner over a period of time. In some embodiments, the therapeutic agents are released over minutes or longer, an hour or long; three hours or longer; six hours or longer; eight hours or longer; twelve hours or longer; fifteen hours or longer; twenty hours or longer; one or more days; two or more days; five or more days; 1 week or longer; two weeks or longer; a month or longer; two months or longer; three months or longer; five months or longer; six months or longer; a year or longer; two years or longer; or five years or longer. [0082] In some embodiments, multiple devices or a device with multiple compartments or sub-compartments are used to conduct duplicative or parallel experiments. In some embodiments, duplicative experiments can be conducted with a device with multiple sub-compartments in the same mammal (e.g., a rodent), thus eliminating variations caused by the natural differences between different mammals (e.g., rodents such as mice). In some embodiments, parallel experiments are carried out in the same mammal in different compartments or sub-compartments to test different experimental conditions. In some embodiments, it is possible to obtain data from the same rodent (e.g., a mouse) imaged over time and substantially reduce experimental error or variation.
[0083] In some embodiments, multiple compartments or sub-compartments within the implantable device are separated by semi-permeable membranes that can allow passage of materials of specific sizes or charges, which allows the diffusion of some molecules into other compartments or sub-compartments. For example, some compartments or sub- compartments can be used to store compounds that are used as substrates for enzymatic reactions.
6.2.4 Size/Capacity
[0084] In another aspect, an implantable device as disclosed herein can be of any size or capacity. In some embodiments, the implantable device can have a largest diameter or longest dimension of between 0.5 mm and 500 mm; 1 mm and 400 mm; 1 mm and 300 mm; 1 mm and 200 mm; 1 mm and 150 mm; 1 mm and 100 mm; 1 mm and 80 mm; 1 mm and 50 mm; between 1 mm and 30 mm; between 10 mm and 30 mm; between 15 mm and 30 mm; between 5 mm and 20 mm; between 5 mm and 15 mm; between 5 mm and 10 mm; between 3 mm and 8 mm; between 1 mm and 5 mm; between 2 mm and 5 mm; or between 0.5 mm and 4 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is between 5 mm and 14 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is between 14 mm and 300 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is smaller than 0.5 mm. In some embodiments, the implantable device can have a largest diameter or dimension that is larger than 500 mm.
[0085] In some embodiments, an implantable device as disclosed herein has a largest depth of between 0.1 mm and 30 mm; 0.1 mm and 20 mm; 0.1 mm and 15 mm;
between 0.2 mm and 10 mm; between 0.3 mm and 10 mm; between 0.3 mm and 8 mm;
between 0.3 mm and 5 mm; between 0.3 mm and 2 mm; or between 0.3 mm and 1.0 mm. In some embodiments, an implantable device as disclosed herein has a depth smaller than 0.1 mm. In some embodiments, an implantable device as disclosed herein has a depth larger than 30 mm.
[0086] In some embodiments, an implantable device as disclosed herein has a volume capacity of 0.01 mm 3 (0.01 μΐ) or larger; 0.1 mm 3 (0.1 μΐ) or larger; 0.5 mm 3 (0.5 μΐ) or larger; 1 mm 3 (1 μΐ) or larger; 2 mm 3 (2 μΐ) or larger; 3 mm 3 (3 μΐ) or larger; 5 mm 3 (5 μΐ) or larger; 8 mm 3 (8 μΐ) or larger; 10 mm 3 (10 μΐ) or larger; 12 mm 3 (12 μΐ) or larger; 15 mm 3
(15 μΐ) or larger; 20 mm 3 (20 μΐ) or larger; 25 mm 3 (25 μΐ) or larger; 30 mm 3 (30 μΐ) or larger; 50 mm 3 (50 μΐ) or larger; 75 mm 3 (75 μΐ) or larger; 100 mm 3 (100 μΐ) or larger; 150 mm 3 (150 μΐ) or larger; 200 mm 3 (200 μΐ) or larger; 250 mm 3 (250 μΐ) or larger; 350 mm 3
(350 μΐ) or larger; 500 mm 3 (500 μΐ) or larger; 750 mm 3 (750 μΐ) or larger; 1,000 mm 3 (1 ml) or larger; 1,200 mm 3 (1.2 ml) or larger; 1,500 mm 3 (1.5 ml) or larger; 1,750 mm 3 (1.75 ml) or larger; 2,000 mm 3 (2 ml) or larger; 2,500 mm 3 (2.5 ml) or larger; 3,000 mm 3 (3 ml) or larger;
5,000 mm 3 (5 ml) or larger; 7,500 mm 3 (7.5 ml) or larger; 10,000 mm 3 (10 ml) or larger;
12,000 mm 3 (12 ml) or larger; 15,000 mm 3 (15 ml) or larger; 20,000 mm 3 (20 ml) or larger; 50,000 mm3 (50 ml) or larger; 75,000 mm3 (75 ml) or larger; or 100,000 mm3 (100 ml) or larger.
[0087] In some embodiments, implantable devices disclosed herein are micro devices. For example, a micro device can have a diameter or dimension of between 1 mm and 5 mm, a depth of between 0.3 mm and 1.0 mm and a volume capacity of about 0.1 μΐ to 25 μΐ. For example, it can be useful to implant a micro device near a smaller organ. In some embodiments, multiple micro implantable devices are implanted near or next to a desired location in a recipient. In some embodiments, multiple micro implantable devices are implanted in multiple locations. Advantageously, in some embodiments, multiple micro implantations can compensate each other in case of possible failure. Also advantageously, in some embodiments, multiple micro implantations can create synergistic effect and result in better treatment or experimentation.
[0088] In some embodiments, implantable devices disclosed herein are mini devices. For example, a mini device can have a diameter or dimension of between 5 mm and 14 mm, a depth of between 0.3 mm and 1.0 mm and a volume capacity of about 5 μΐ to 150 μΐ. For example, a mini device can be used to grow more tissue than the smaller micro units for analysis and production of biological products. In some embodiments, it is possible to implant multiple mini devices in a recipient. [0089] In some embodiments, implantable devices disclosed herein are macro devices. For example, a macro device can have a diameter or dimension of between 14 mm and 300 mm, a depth of between 0.3 mm and 1.0 mm and a volume capacity of about 40 μΐ to 100 ml. For example, a macro device can be used to grow more tissue than the smaller micro or mini units for analysis and production of biological products. In some embodiments, it is possible to implant multiple macro devices in a recipient.
[0090] In some embodiments, the size and capacity of an implantable device are adjusted to the site of implantation. In some embodiments, the size and capacity of an implantable device are adjusted to the purpose for which the device is used.
[0091] In some embodiments, the implantable devices can be coated to avoid irritation or other undesirable side effects in the recipient. Exemplary coating materials include but are not limited to at least partially alkylated polyethyleneimine (PEI); at least partially alkylated poly(lysine); at least partially alkylated polyornithine; at least partially alkylated poly(amido amine), at least partially alkylated homo- and co-polymers of vinylamine; at least partially alkylated acrylate containing aminogroups, copolymers of vinylamine containing aminogroups with hydrophobic monomers, copolymers of acrylate containing aminogroups with hydrophobic monomers, and amino containing natural and modified polysaccharides and mixtures thereof.
[0092] Additional examples of biocompatible coating materials can be found, for example, in US Pat. No. 6,127,448 to Domb, entitled "Biocompatible Polymeric Coating Material;" US Pat. Pub. No. 2004/0148016 by Klein and Brazil, entitled "Biocompatible Medical Device Coatings;" US Pat. Pub. No. 2009/0169714 by Burghard et al, entitled "Biocompatible Coatings for Medical Devices;" US Pat. No. 6,406,792 to Briquet et al, entitled "Biocompatible Coatings;" US Pat. Pub. No. 2008/0003256 by Martens et al, entitled "Biocompatible Coating of Medical Devices;" each of which is hereby incorporated by reference herein in its entirety.
6.2.5 Material and Method for Making an Implantable Device and Parts thereof
[0093] Any suitable material can be used to manufacture an implantable device as disclosed herein. Exemplary materials for the main body or housing of the device include but are not limited to stainless steel, surgical steel (e.g., Grade 316), titanium, a plastic or polymeric material, a biocompatible or biodegradable material or a combination thereof.
[0100] Exemplary biocompatible materials also include but are not limited to polyacrylates, polymethacryates, polyureas, polyurethanes, polyolefms, polyvinylhalides, polyvinylidenehalides, polyvinylethers, polyvinylaromatics, polyvinylesters, polyacrylonitriles, alkyd resins, polysiloxanes and epoxy resins.
[0101] Additional examples of biocompatible biodegradable polymers include, without limitation, polycaprolactone, poly(L-lactide), poly(D,L-lactide), poly(D,L-lactide-co- PEG) block copolymers, poly(D,L-lactide-co-trimethylene carbonate), poly(lactide-co- glycolide), polydioxanone (PDS), polyorthoester, polyanhydride, poly(glycolic acid-co- trimethylene carbonate), polyphosphoester, polyphosphoester urethane, poly(amino acids), polycyanoacrylates, poly(trimethylene carbonate), poly(iminocarbonate), polycarbonates, polyurethanes, polyalkylene oxalates, polyphosphazenes, PHA-PEG, and combinations thereof. The PHA may include poly(a-hydroxyacids), poly(P-hydroxyacid) such as poly(3- hydroxybutyrate) (PHB), poly(3-hydroxybutyrate-co-valerate) (PHBV), poly(3- hydroxyproprionate) (PHP), poly(3-hydroxyhexanoate) (PHH), or poly(4-hydroxyacid) such as poly poly(4-hydroxybutyrate), poly(4-hydroxyvalerate), poly(4-hydroxyhexanoate), poly(hydroxyvalerate), poly(tyrosine carbonates), poly(tyrosine arylates), poly(ester amide), polyhydroxyalkanoates (PHA), poly(3-hydroxyalkanoates) such as poly(3- hydroxypropanoate), poly(3-hydroxybutyrate), poly(3-hydroxyvalerate), poly(3- hydroxyhexanoate), poly(3-hydroxyheptanoate) and poly(3-hydroxyoctanoate), poly(4- hydroxyalkanaote) such as poly(4-hydroxybutyrate), poly(4-hydroxyvalerate), poly(4- hydroxyhexanote), poly(4-hydroxyheptanoate), poly(4-hydroxyoctanoate) and copolymers including any of the 3-hydroxyalkanoate or 4-hydroxyalkanoate monomers described herein or blends thereof, poly(D,L-lactide), poly(L-lactide), polyglycolide, poly(D,L-lactide-co- glycolide), poly(L-lactide-co-glycolide), polycaprolactone, poly(lactide-co-caprolactone), poly(glycolide-co-caprolactone), poly(dioxanone), poly(ortho esters), poly(anhydrides), poly(tyrosine carbonates) and derivatives thereof, poly(tyrosine ester) and derivatives thereof, poly(imino carbonates), poly(glycolic acid-co-trimethylene carbonate), polyphosphoester, polyphosphoester urethane, poly(amino acids), polycyanoacrylates, poly(trimethylene carbonate), poly(iminocarbonate), polyphosphazenes, silicones, polyesters, polyolefms, polyisobutylene and ethylene-alphaolefm copolymers, acrylic polymers and copolymers, vinyl halide polymers and copolymers, such as polyvinyl chloride, polyvinyl ethers, such as polyvinyl methyl ether, polyvinylidene halides, such as polyvinylidene chloride,
polyacrylonitrile, polyvinyl ketones, polyvinyl aromatics, such as polystyrene, polyvinyl esters, such as polyvinyl acetate, copolymers of vinyl monomers with each other and olefins, such as ethylene-methyl methacrylate copolymers, acrylonitrile-styrene copolymers, ABS resins, and ethylene-vinyl acetate copolymers, polyamides, such as Nylon 66 and polycaprolactam, alkyd resins, polycarbonates, polyoxymethylenes, polyimides, polyethers, poly(glyceryl sebacate), poly(propylene fumarate), poly(n-butyl methacrylate), poly(sec- butyl methacrylate), poly(isobutyl methacrylate), poly(tert-butyl methacrylate), poly(n-propyl methacrylate), poly(isopropyl methacrylate), poly(ethyl methacrylate), poly(methyl methacrylate), epoxy resins, polyurethanes, rayon, rayon-triacetate, cellulose acetate, cellulose butyrate, cellulose acetate butyrate, cellophane, cellulose nitrate, cellulose propionate, cellulose ethers, carboxymethyl cellulose, polyethers such as poly(ethylene glycol) (PEG), copoly(ether-esters) (e.g. poly(ethylene oxide-co-lactic acid) (PEO/PLA)), polyalkylene oxides such as poly(ethylene oxide), poly(propylene oxide), poly(ether ester), polyalkylene oxalates, phosphoryl choline containing polymer, choline, poly(aspirin), polymers and co-polymers of hydroxyl bearing monomers such as 2-hydroxyethyl methacrylate (HEMA), hydroxypropyl methacrylate (HPMA),
hydroxypropylmethacrylamide, PEG acrylate (PEGA), PEG methacrylate, methacrylate polymers containing 2-methacryloyloxyethyl- phosphorylcholine (MPC) and n-vinyl pyrrolidone (VP), carboxylic acid bearing monomers such as methacrylic acid (MA), acrylic acid (AA), alkoxymethacrylate, alkoxyacrylate, and 3-trimethylsilylpropyl methacrylate (TMSPMA), poly(styrene-isoprene-styrene)-PEG (SIS-PEG), polystyrene-PEG,
polyisobutylene-PEG, polycaprolactone-PEG (PCL-PEG), PLA-PEG, poly(methyl methacrylate), MED610, poly(methyl methacrylate)-PEG (PMMA-PEG),
polydimethylsiloxane-co-PEG (PDMS-PEG), poly(vinylidene fiuoride)-PEG (PVDF-PEG), PLURONIC™ surfactants (polypropylene oxide-co-polyethylene glycol),
poly(tetramethylene glycol), hydroxy functional poly( vinyl pyrrolidone), biomolecules such as collagen, chitosan, alginate, fibrin, fibrinogen, cellulose, starch, dextran, dextrin, hyaluronic acid, fragments and derivatives of hyaluronic acid, heparin, fragments and derivatives of heparin, glycosamino glycan (GAG), GAG derivatives, polysaccharide, elastin, elastin protein mimetics, or combinations thereof.
[0102] In some embodiments, when an implantable device is formed with one or more biodegradable materials, the device degrades over one or more days, two or more days, five or more days, 1 week or longer, two weeks or longer, a month or longer, two months or longer, three months or longer, five months or longer, six months or longer, a year or longer, two years or longer or five years or longer. In some embodiments, the compartment or sub- compartments within an implantable device are made of the same material as the main body or housing of the implantable device. In some embodiments, compartments or sub- compartments within an implantable device are made of material that is different from that of the main body or housing. In some embodiments, sub-compartments are formed by inserting a pre-formed divider or insert into the main body or housing, similar to the design of an insert for an ice-cube tray. A pre-formed divider is formed based on the dimension of the main body or housing. In some embodiments, multiple pre-formed dividers can be used with the same type of main body or housing to create compartments or sub-compartments of different shapes. In some embodiments, the dividers or inserts are added to the main body or housing before any material is added to the resulting compartments or sub-compartments.
[0103] In some embodiments, walls between compartments or sub-compartments are impermeable. In some embodiments, walls between compartments or sub-compartments are semi-permeable and permit transmission.
[0104] In some embodiments, a cover slip is modified to have dividers that slide into the main body or housing and create compartments or sub-compartments. In such embodiments, the biochemical mixture is added before the cover slip is attached.
[0105] In some embodiments, compartments or sub-compartments are formed using backbone or substrate material at the same time when the main body or housing of the implantable device is created (Figures 2A-2C).
[0106] In some embodiments, the material for making the main body or housing of the implantable device is also used to construct other parts of the device, such as a reaction chamber, a well, a channel, or a mesh or net of the interaction module.
[0107] It will be understood that the method for manufacturing an implantable device is determined by the type of material used and the purpose of the device. For example, a metal device can be created by metal lathing, milling, and CNC molding. A plastic device can be made by injection molding, extrusion molding or any other suitable method. In some embodiments, methods of three-dimensional printing on metals and other polymeric materials are used. In some embodiments, methods of laser printing or laser scribing are used.
6.3 Device as Part of a System through Hardware and Software Integration
[0108] In some embodiments, an on-board biomarker monitoring system can be integrated with an implantable device described herein. In some embodiments, a fluid or compound delivery system can be integrated with an implantable device described herein.
The integration can be achieved using, for example, a technology similar to the lab-on-a-chip microfluidic technology (e.g., Figure 3). [0109] As used herein, an integration can be either functional or structural. In some embodiments, chambers, channels, wells, compartments and / or sub-compartments form a sophisticated network dedicated for a specific biological or biochemical assay. In such embodiments, each individual component of the network and the content therein provide an element for completing the desired assay, e.g., by hosting cells or tissues, detecting biomarkers, adjusting reaction conditions, and producing signals. In some embodiments, the network is designed for delivering specific compounds. The network-like configuration allows miniaturization of implantable devices that are specific for certain biological or biochemical assays. In some compartments, a pre-designed network of chambers, channels, wells, compartments and / or sub-compartments can be pre-formed on an insert or chip-like device (e.g., Figure 3). The insert or chip can be put into an implantable device. In some compartments, a pre-designed network of chambers, channels, wells, compartments and/or sub-compartments can be constructed as parts of the implantable device. In some
embodiments, various components of the network can be made individually and assembled in a pre-formed device to form the network that performs one or more biological assays. The implantable device can be made before or at the same time as an assay insert.
[0110] In some embodiments, an insert or chip dedicated for a specific biochemical assay can be placed in a defined position within an implantable device, e.g., in the center or close to the interaction module. In some embodiments, it is possible to evaluate the viability and health of the tissues or cells within the device by measurements of biomarkers that reflect certain physiological parameters (such as acidity which may indicate lactic acidosis or cell necrosis). In some embodiments, the system can be used to deliver precise quantities to the bioreactor at pre-set times, from storage depots to a reaction chamber.
[0111] In some embodiments, a wireless communication module can be integrated with an implantable device described herein. In order to allow continuous monitoring of the status of the implantable devices, the on-board biomarker detection system can convert measured data to a digital signal and wirelessly transmitted from the implant to an external receiving device such as a computer. A wireless communication module can also facilitate communication of data to a mobile phone, a PDA, other wireless communication device. Such embodiments can monitor the device and provide live or instant feedback. For example, insulin levels within and near an implantable device can be detected, monitored and recorded by a smart phone or PDA device. [0112] In some embodiments, a wireless signal receiver can be integrated with an implantable device described herein. In some embodiments, a small computer chip can be integrated with an implantable device described herein. In some embodiments, the small computer chip or signal receiver can be used to control sequential release of biochemical reagents from the chambers, channels, wells, compartments and/or sub-compartments on an implantable device.
[0113] In some embodiments, a biomarker detection system can communicate with its own on-board computer equipment to send and receive signals. In some
embodiments, e.g., when an implantable device is used to produce and provide live-saving biomolecules, the level of the biomolecules can be communicated to medical professionals or a data center, which in turn provides medical aid or triggers an alert for replacement of the implant. In some embodiments, when built-in sensors are used to detect appropriate signals, the signals can be used to activate cells in particular ways as needed: to release cell products, or to release drugs or other compounds into the body or into cells within the device. For example, built-in sensors can detect sugar levels and trigger release of insulin from genetically engineered cells within the device, or insulin within insulin-storage depots within the device is released as a response. In some embodiments, new measurements of sugar levels are taken after blood circulates around the body, and returns to the device. The new measurement provides information on whether additional adjustment of the levels of insulin is needed.
[0114] In some embodiments, attachment of a lens can be used to focus the camera or other optical equipment. In some embodiments, wireless capabilities can be used to transmit image or video data. In some embodiments, an implantable device as disclosed herein is equipped with the ability to apply light or radiation within the device. Such embodiments provide the advantage of creating excitation and emission of desired wavelengths of light, which can be useful in visualizing the content within the device. In some embodiments, radiation at various wavelengths (e.g., ultraviolet) can be used to reduce or eliminate inflammation or other immune reaction to the device. This can reduce immunogenicity of the device or its content, particularly if cell populations within the device differ from those of the host.
[0115] In some embodiments, structural features (such as grooves) are created on one or more sides of the implantable device to create secure attachment between the device and one or more additional modules. [0116] In some embodiments, multiple functionalities can be incorporated into an implantable device by microfabrication techniques. An exemplary embodiment of a multifunctional implantable device can be found in Figure 6. The different functional modules and features will be described with more details in connection with a multi-step biomolecules synthesis process.
6.4 Biological and Chemical Media Compositions
[0117] In some embodiments, the content within a compartment or sub- compartment of an implantable device includes at least a plurality of cells and a biochemical composition in which the cells may be maintained, grow, develop and/or differentiate. In some embodiments, the biochemical composition comprises a biocompatible buffer, a growth media or an extracellular matrix.
[0118] In some embodiments, the cells or tissue used in the device can be suspended in a liquid trapped within a sub-compartment, adhered to the inner walls of the compartment or immobilized on an appropriate support structure provided within the compartment. For example, the cells can be embedded in a gel matrix (e.g., agar, alginate, chitosan, polyglycolic acid, polylactic acid, and the like). In some embodiments, a porous scaffold (e.g., an alignate scaffold) can be used to seed the content within a compartment or sub-compartments of an implantable device. In some embodiments, microcapsules or microbeads can be used to encapsulate or capture cells in the cellular compartment.
[0119] In some embodiments, a commercially available growth medium or matrix for mammalian cells is used. For example, Matrigel™ is the trade name for a gelatinous protein mixture secreted by Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells and marketed by BD Biosciences and by Trevigen Inc. under the name Cultrex BME. This mixture resembles the complex extracellular environment found in many tissues and is used by cell biologists as a substrate for cell culture. Components of a standard growth medium or matrix for mammalian cells include but are not limited to extracellular matrix components, growth factors, various cytokines, and one or more pharmaceutical agents, as listed in Table 1.
Table 1. Components of exemplary biochemical composition.
Extracellular Matrix components
Undefined media
Extract from the EHS tumor (e.g., Matrigel™ from Extracellular Matrix components
BD Biosciences)
Growth Factor Reduced Matrigel™
High Concentration Matrigel™
Exemplary individual components:
Laminin
Entactin 1
Collagens I- VI
Heparin sulfate proteoglycans
agar
alginate
chitosan
polyglycolic acid
polylactic acid
[0120] Exemplary growth factors include but are not limited to adrenomedullin (AM), angiopoietin (Ang), autocrine motility factor, bone morphogenetic proteins (BMPs), brain-derived neurotrophic factor (BDNF), epidermal growth factor (EGF), erythropoietin (EPO), fibroblast growth factor (FGF) 1, 2, 3, glial cell line-derived neurotrophic factor (GDNF), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony- stimulating factor (GM-CSF), growth differentiation factor-9 (GDF9), hepatocyte growth factor (HGF), hepatoma-derived growth factor (HDGF), insulin-like growth factor (IGF), migration-stimulating factor, myostatin (GDF-8), nerve growth factor (NGF) and other neurotrophins, platelet-derived growth factor (PDGF), thrombopoietin (TPO), transforming growth factor alpha (TGF-a), transforming growth factor beta(TGF-P), tumor necrosis factor- alpha (TNF-a), vascular endothelial growth factor (VEGF), placental growth factor (PIGF), fetal bovine somatotrophin (FBS), IL-1, IL-2, IL-3, IL-4, IL-5, IL-6 and IL-7.
[0121] Exemplary cytokines include but are not limited to interleukin 2, interleukin 15 preproprotein, tumor necrosis factor (ligand) superfamily, member 18, interleukin 26, interleukin 20, interleukin 22, interferon epsilon 1, interferon-gamma, colony stimulating factor 2, interleukin 19 isoform 2, tumor necrosis factor (ligand) superfamily, member 4, interleukin 24 isoform 1, interferon, beta 1, fibroblast, interleukin 5, interleukin 13, growth hormone 2 isoform 1, interferon, omega 1, interleukin 12 A, tumor necrosis factor (ligand) superfamily, member 10, interleukin 6 (interferon, beta 2), interferon, alpha 1, growth hormone 1 isoform 1 , leptin, interleukin 1 , beta proprotein, tumor necrosis factor alpha, interferon kappa, interleukin 3, interleukin 10, tumor necrosis factor (ligand) superfamily, member 15, prolactin, interleukin 28 A, interleukin 17B, ciliary neurotrophic factor, thymic stromal lymphopoietin isoform 1, interleukin 4 isoform 1, interleukin 17E isoform 1, chemokine (C-C motif) ligand 16, interleukin 9, interleukin 1, alpha proprotein, chemokine (C-C motif) ligand 15, chemokine (C motif) ligand 2, tumor necrosis factor ligand superfamily, member 14 isoform 1, chemokine (C motif) ligand 1, chemokine (C-C motif) ligand 25, CD40 ligand, chemokine (C-X-C motif) ligand 13 (B-cell chemoattractant), interleukin 29, tumor necrosis factor (ligand) superfamily, member 8, and interleukin 28B.
[0122] In some embodiments, the biochemical composition also includes metabolites, small molecules or macromolecules.
[0123] Exemplary metabolites include but are not limited to alcohols (e.g., ethanol), amino acids (e.g., glutamic acid, aspartic acid), nucleotides (e.g., 5' guanylic acid), antioxidants (e.g., isoasorbic acid), organic acids (e.g., acetic acid, lactic acid), polyols (e.g., glycerol), vitamins (e.g., B2), minerals, and electrolytes. In some embodiments, metabolites also include secondary metabolites.
[0124] Exemplary small chemical molecules include any chemical compounds, including inorganic and organic compounds, for example, formaldehyde, acetylsalicylic acid, methanol, ibuprofen, and statins. Exemplary macromolecules include but are not limited to monoclonal and polyclonal antibodies, nucleic acid, lipid, fatty acid, and insulin.
6.5 Methods and Locations of Implantation
[0125] In some embodiments, an implantable device as disclosed herein can be implanted in a mammal, for example, a mouse, a rat, a dog, a cow, a sheep, a goat, a cat, or a human.
[0126] In some embodiments, standard surgical techniques (using tools such as, forceps and sutures) are used to surgically open and close the skin around the implant.
[0127] In some embodiments, a punch mechanism can be used for easy attachment in subcutaneous locations. The mechanism is similar to that of grommet pliers and grommets (see Figure 7). In some embodiments, the method includes punching the lid and upper compartments to lower compartments, or only a pinching mechanism running on the sides of the device that is secured with the punch. Advantageous the latter can be used to create quick subcutaneous implantation. The punch-based mechanism, while efficient in securing the device, must be done carefully to avoid too much damage to the surrounding tissues and result in inflammation. The method is only useful for subcutaneous implantation, not other locations (e.g., peritoneal cavity of the abdomen). [0128] In some embodiments, an implantable device is implanted within the skin in an accessible area of the body of a mammal, especially when the purpose of the implantation is for observing biochemical reactions and cell developments. In some embodiments, the device is attached to the dorsal skin region of a mammal (e.g., on the back of a rodent). Such attachments allow easy access for microscopy analysis. The protrusion caused by the implant can create discomfort for a mammal or risk of dislodging or damaging the device; e.g., a device can be knocked around when a recipient mammal hits objects in the cage where the mammal lives.
[0129] In some embodiments, an implantable device is implanted near the dorsal head region of a mammal (e.g., the "hat" position between the ears in a rodent). Such attachments allow easy access for microscopy analysis and avoid mechanical irritation (e.g., scratching by the recipient mammal). However, the implantation site can limit the size of the device.
[0130] In some embodiments, an implantable device is implanted to the abdominal region. In such embodiments, it is possible to accommodate larger implantable devices.
[0131] In some embodiments, an implantable device is implanted in locations internal to the skin (e.g., in the intraperitoneal cavity or beside the liver or pancreas). In such embodiments, the device is hidden from view and protected from accidental damage. The biochemical composition and cells can benefit from the cellular and extracellular components of different locations within the body (e.g., via interactions with extracellular fluid components from the liver or pancreas). Such implantation allows cellular products within the device to be delivered to specific locations as needed (e.g., antibodies produced within the unit are provided to regions near lymph nodes or the lymphatic system). However, it is also more difficult surgically to implant in such locations and more difficult to analyze or monitor over time (e.g., microscopy can be difficult if the location to be observed is not external facing).
[0132] In some embodiments, an implantable device are made of biodegradable materials. It is possible to package the device with suitable biochemical material that will be gradually released and stimulate the growth of implanted cells/tissues. Over time, when the device is completely degraded, the resulting cells/tissue becomes an integral part of the target organ.
6.6 Applications of Implantable Devices [0133] The implantable devices can be used for various purposes, including but not limited as a research tool, a drug testing tool, a tool for producing biomolecules (small or large), a tool for tissue or organ implant/transplant, a tool for detecting biomarkers or biomolecules, or an extracorporeal body monitor and modulator.
6.6.1 Implantable Devices as a Research Tool
[0134] In one aspect, an implantable device as disclosed herein is used as a research tool. In some embodiments, an implantable device is used for evaluating the functions of individual biological factors in a live mammal. Because the device is implanted in a live mammal, it is possible to analyze the comprehensive response of the whole mammal to a particular stimulus. In some embodiments, pressure can be applied to the system before the effects are observed (e.g., it is possible to analyze the effects of pressure on bone growth and remodeling). In some embodiments, the system can also be subject to micro or zero- gravity conditions (e.g., experiments done under such conditions have shown unusual properties in stem cells). In some embodiments, an electrical current can be applied to the system. In some embodiments, quantum mechanical microscopy can be used in connection with the system.
[0135] In some embodiments, an implantable device as disclosed herein is used to evaluate the functions of small or large biomolecules (e.g., proteins, peptides, nucleic acids, combinations and analogs thereof) in a live mammal. Exemplary uses include but are not limited to angiogenesis study, anti-angiogenesis study (e.g., in the context of anti-cancer drugs analysis or screening), cancer and anti-cancer studies, stem cell studies, nerve regeneration study (e.g., determining the growth factors that contribute to nerve sprouting and re-growth in vivo), and liver toxicity study (e.g., testing liver drugs and evaluating their toxicity during pre-clinical development).
[0136] In some embodiments, an implantable device as disclosed herein is used to evaluate the function of stem cells and tissue constructs before the cells or tissues are used in further treatment or therapy. This use of the implantable device is applicable to all organ/tissue types, including but not limited to liver tissue, pancreas tissue, kidney tissue, lung tissue, skin tissue, bone marrow and lymph node tissue, thyroid tissue, pituitary tissue, brain tissue, muscle tissue (including myocardium, skeletal muscle and smooth muscle), cartilage tissue, gastrointestinal tissues, reproductive tissues including uterus, and ovaries.
[0137] In some embodiments, an implantable device as disclosed herein is used as a part of a live imaging system, in which images, videos or other optical data of live cells can be collected, stored, displayed, and processed. Based on previous data on fully subcutaneous implants, in some embodiments, cells and microvasculature can be observed to be entering the device within 1 week. In some embodiments, approximately 1.5 mm of ingrowth can be observed within 4 weeks, including functioning blood vessels, and associated cells from the host. In some embodiments, the cover glass and retaining clip can be removed at 4 weeks and stains such as Hoescht 33342 nuclear stain, or other antibody stains can be added to stain live cells, or cells and growth factors can be added. In some embodiments, the implantable device can be resealed; and cell growth can be further monitored. In some embodiments, the kinetics of diverse cell populations as they enter the device can be observed from day 1 to 6 weeks after the implantation. In some embodiments, the relations and organization of multiple cell populations in one location can be examined. In some embodiments, blood vessel growth within a live mammal (e.g., a rodent) can be observed and examined by micro CT scan.
[0138] In some embodiments, partial or the entire content within the implantable device can be removed and subject to biological and biochemical analyses. In some embodiments, the removed content can be subject to sectioning, staining and imaging by conventional confocal microscopy methods. In some embodiments, removed devices content cam be digested and quantified by flow cytometry, mass spectrometry, or other analytical methods.
[0139] In some embodiments, contributions of bone marrow derived cells migrating, and proliferating within the device can be examined. In some embodiments, wildtype mice subject to lethal irradiation of C57B16 can be used. These mice are then injected with bone marrow from GFP transgenic wildtype mice. In some embodiments, signs of positive engraftment of GFP bone marrow can be observed at about 4 week after implantable devices are introduced into these mice. An exemplary sign is the production of GFP-labeled peripheral blood.
[0140] In some embodiments, after about 4 weeks of implantation, content in the device can reveal GFP positive cells (which are inferred to have originated from the bone marrow) as well as non-GFP cells (which have come from tissues neighboring the device). This is a test for determining cells that are bone marrow derived and can have bone marrow stem cell potential.
6.6.2 Implantable Devices as a Drug Testing Tool [0141] In one aspect, an implantable device as disclosed herein is used as a drug- testing tool. Advantageously, the device allows direct observation of cellular response. Also advantageously, it is possible to conduct drug metabolism study in the live mammal, including drug safety and toxicity studies, dosing studies, drug kinetics, efficiency and efficacy, and bioavailability studies.
[0142] In some embodiments, a multi-functional implantable device can be used as a personalized tissue assay (e.g., drug screening on a patient biopsy sample with cancer drugs) (Figure 8). In some embodiments, a biopsy sample can be collected from an individual's tumor by conventional methods. This sample can be collected and maintained either live or in a frozen state before division into identical pieces by shape and weight. In some embodiments, a mammal containing implantable devices can be prepared to allow the biopsy sample successfully engraft and grow within the devices. In some embodiments, immuno-compromised mice can be employed to enable engraftment of human tissues. In some embodiments, biopsy samples of the same size or weight from a mammal (e.g., a rodent or a human) can be implanted within devices in multiple test mammals (which can be the same or different from the sample mammal. In some embodiments, the biopsy samples engraft and live within the devices and are analyzed by optical methods or biomarker analysis. The mammals can subsequently be administered therapeutics. The effects of the therapeutics can be assessed. The therapeutic agents that are effective in destroying or reducing one diseased state of the biopsy sample can be selected for additional study. In some embodiments, duplicate tests are performed. In some embodiments, the selected therapeutics are given to the mammal from which the biopsy sample is taken.
[0143] In some embodiments, biopsy samples of the same size or weight from a mammal (e.g., a rodent or a human) can be implanted within devices in a same test mammal. Here the implantable device comprises multiple sub-compartments or chambers that are subject to the same growth conditions and each contains a biopsy sample. Different therapeutic agents are then administered to different sub-compartments and the effects on the biopsy samples within each sub-compartment are analyzed and compared with each other.
[0144] In some embodiments, the biopsy sample is associated with a type of cancer, including but not limited to Acute lymphoblastic leukemia; Acute myeloid leukemia; Adrenocortical carcinoma; AIDS-related cancers; AIDS-related lymphoma Anal cancer; Appendix cancer; Astrocytoma, childhood cerebellar or cerebral; Basal cell carcinoma; Bile duct cancer, extrahepatic; Bladder cancer; Bone cancer; Osteosarcoma/Malignant fibrous histiocytoma; Brainstem glioma; Brain tumor; Brain tumor; cerebellar astrocytoma; Brain tumor; cerebral astrocytoma/malignant glioma; Brain tumor, ependymoma; Brain tumor, medulloblastoma; Brain tumor, supratentorial primitive neuroectodermal tumors; Brain tumor, visual pathway and hypothalamic glioma; Breast cancer; Bronchial
adenomas/carcinoids; Burkitt lymphoma; Carcinoid tumor, childhood; Carcinoid tumor, gastrointestinal; Carcinoma of unknown primary; Central nervous system lymphoma, primary; Cerebellar astrocytoma, childhood; Cerebral astrocytoma/Malignant glioma, childhood; Cervical cancer; Childhood cancers; Chronic lymphocytic leukemia; Chronic myelogenous leukemia; Chronic myeloproliferative disorders; Colon Cancer; Cutaneous T- cell lymphoma; Desmoplastic small round cell tumor; Endometrial cancer; Ependymoma; Esophageal cancer; Ewing's sarcoma in the Ewing family of tumors; Extracranial germ cell tumor, Childhood; Extragonadal Germ cell tumor; Extrahepatic bile duct cancer; Eye Cancer, Intraocular melanoma; Eye Cancer, Retinoblastoma; Gallbladder cancer; Gastric (Stomach) cancer; Gastrointestinal Carcinoid Tumor; Gastrointestinal stromal tumor (GIST); Germ cell tumor: extracranial, extragonadal, or ovarian; Gestational trophoblastic tumor; Glioma of the brain stem; Glioma, Childhood Cerebral Astrocytoma; Glioma, Childhood Visual Pathway; Hypothalamic; Gastric carcinoid; Hairy cell leukemia; Head and neck cancer; Heart cancer; Hepatocellular (liver) cancer; Hodgkin lymphoma; Hypopharyngeal cancer; Hypothalamic and visual pathway glioma, childhood; Intraocular Melanoma; Islet Cell Carcinoma
(Endocrine Pancreas); Kaposi sarcoma; Kidney cancer (renal cell cancer); Laryngeal Cancer; Leukemia; Leukemia, acute lymphoblastic (also called acute lymphocytic leukemia);
Leukemia, acute myeloid (also called acute myelogenous leukemia); Leukemia, chronic lymphocytic (also called chronic lymphocytic leukemia); Leukemia, chronic myelogenous (also called chronic myeloid leukemia); Leukemia, hairy cell; Lip and Oral Cavity Cancer; Liposarcoma; Liver Cancer (Primary); Lung Cancer, Non-Small Cell; Lung Cancer, Small Cell; Lymphoma; Lymphoma, AIDS-related; Lymphoma, Burkitt; Lymphoma, cutaneous T- Cell; Lymphoma, Hodgkin; Lymphomas, Non-Hodgkin (an old classification of all lymphomas except Hodgkin's); Lymphoma, Primary Central Nervous System;
Macroglobulinemia, Waldenstrom; Malignant Fibrous Histiocytoma of Bone/Osteosarcoma; Medulloblastoma, Childhood; Melanoma; Melanoma, Intraocular (Eye); Merkel Cell Carcinoma; Mesothelioma, Adult Malignant; Mesothelioma, Childhood; Metastatic
Squamous Neck Cancer with Occult Primary; Mouth Cancer; Multiple Endocrine Neoplasia Syndrome, Childhood; Multiple Myeloma/Plasma Cell Neoplasm; Mycosis Fungoides;
Myelodysplasia Syndromes; Myelodysplastic/Myeloproliferative Diseases; Myelogenous Leukemia, Chronic; Myeloid Leukemia, Adult Acute; Myeloid Leukemia, Childhood Acute; Myeloma, Multiple (Cancer of the Bone-Marrow); Myeloproliferative Disorders, Chronic; Nasal cavity and paranasal sinus cancer; Nasopharyngeal carcinoma; Neuroblastoma; Non- Hodgkin lymphoma; Non-small cell lung cancer; Oral cancer; Oropharyngeal cancer;
Osteosarcoma/malignant fibrous histiocytoma of bone; Ovarian cancer; Ovarian epithelial cancer (Surface epithelial-stromal tumor); Ovarian germ cell tumor; Ovarian low malignant potential tumor; Pancreatic cancer; Pancreatic cancer, islet cell; Paranasal sinus and nasal cavity cancer; Parathyroid cancer; Penile cancer; Pharyngeal cancer; Pheochromocytoma; Pineal astrocytoma; Pineal germinoma; Pineoblastoma and supratentorial primitive neuroectodermal tumors, childhood; Pituitary adenoma; Plasma cell neoplasia/Multiple myeloma; Pleuropulmonary blastoma; Primary central nervous system lymphoma; Prostate cancer; Rectal cancer; Renal cell carcinoma (kidney cancer); Renal pelvis and ureter, transitional cell cancer; Retinoblastoma; Rhabdomyosarcoma, childhood; Salivary gland cancer; Sarcoma, Ewing family of tumors; Sarcoma, Kaposi; Sarcoma, soft tissue; Sarcoma, uterine; Sezary syndrome; Skin cancer (nonmelanoma); Skin cancer (melanoma); Skin carcinoma, Merkel cell; Small cell lung cancer; Small intestine cancer; Soft tissue sarcoma; Squamous cell carcinoma - see Skin cancer (nonmelanoma); Squamous neck cancer with occult primary, metastatic; Stomach cancer; Supratentorial primitive neuroectodermal tumor, childhood; T-Cell lymphoma, cutaneous (Mycosis Fungoides and Sezary syndrome);
Testicular cancer; Throat cancer; Thymoma, childhood; Thymoma and Thymic carcinoma; Thyroid cancer; Thyroid cancer, childhood; Transitional cell cancer of the renal pelvis and ureter; Trophoblastic tumor, gestational; Unknown primary site, carcinoma of, adult;
Unknown primary site, cancer of, childhood; Ureter and renal pelvis, transitional cell cancer; Urethral cancer; Uterine cancer, endometrial; Uterine sarcoma; Vaginal cancer; Visual pathway and hypothalamic glioma, childhood; Vulvar cancer; Waldenstrom
macro globulinemia and Wilms tumor (kidney cancer), childhood.
[0145] In some embodiments, the biopsy sample is from a non-cancer related disease.
6.6.3 Implantable Devices as a Tool for Producing Molecules
[0146] In one aspect, an implantable device as disclosed herein is used as a reservoir for growing small or large biomolecules. Exemplary small molecules include but are not limited to aspirin, morphine for chronic pain, methadone for heroine withdrawal symptoms; hormones, vitamins and other non-regulated supplements and nutrients (e.g., Vitamins A-E, etc.) in slow release from within cells or just from within the implantable device). Exemplary biomolecules that can be produced by the device include but are not limited to insulin, various growth factors, prolactin, Cortisol, estrogen, progesterone, testosterone, serotonin, aldosterone, erythropoietin, granulocyte-colony stimulating factor (e.g., GCSF and analogs), and all types of thyroid hormones. In some embodiments, antibodies can also be produced where the device functions as an artificial lymph node and, for example, produces custom designed antibodies.
[0147] In some embodiments, a multi-functional implantable device can be used to construct a specific 'cellular' manufacturing system using different compartments and sub- compartments within the device (see Figures 3 and 6). In such embodiments, the implantable device includes a series of channels and sub-compartments containing various modules. These channels and sub-compartments can mimic the original cellular processes by releasing and mixing biochemical reagents as necessary to result in a defined cascade of biochemical reactions. In some embodiments, the device is filled with biomolecular media that stimulates invasion by endothelium and eventual blood vessel formation through the device. In some embodiments, genetically engineered cells are introduced (or pre-loaded) into the
compartments or sub-compartments that are in contact with the blood vessels (e.g., at locations 1-4 in Figure 6). In some embodiments, specific biomarker sensors are contained within the device (at position A and downstream position B in Figure 6), which come in contact with in-flowing blood and body fluids. Upon detection of a target substance or a characteristic thereof (e.g., the sugar level) information is transmitted from the biosensor to the microprocessing device (e.g., at position C in Figure 6). In some embodiments, a microprocessing computer sends the appropriate electrical and/or other signals to cells or chemical storage depots within the device to trigger the release of their contents. For example, insulin-producing cells will release insulin at locations 1-4 in Figure 6. In some embodiments, as demonstrated in Figure 6, the out-flowing blood or body fluid leaving the device is altered before exiting to the rest of the body.
[0148] In some embodiments, the product produced by cells in the implantable device can be removed from the device or transferred to a different location within the device via the injection port or observation module.
6.6.4 Implantable Devices as a Tool for Tissue or Organ Implant/Transplant
[0149] In one aspect, an implantable device as disclosed herein is used for growing cells and tissues for implants or transplants. In some embodiments, the cells that are introduced into the system can be modified or unmodified (e.g., genetically modified), differentiated or un-differentiated. In some embodiments, cells grown in the device are altered by genetic or other methods, to optimize the production of a desired small or large biomolecule. Exemplary issues that can later be transplanted to different locations include but are not limited to muscle tissues, nerve tissues, liver tissue, pancreas tissue, and blood vessels.
[0150] In some embodiments, embryonic stem cells (e.g., blastocyst-derived) are cultured and produced within an implantable device as disclosed herein. In some
embodiments, blastocyst-derived stem cells isolated from the inner cell mass of blastocysts can be used. In some embodiments, adult stem cells or somatic stem cells, which are found in various tissues (e.g., from bone marrow derived sources), can also be used. Additional adult stem cells include but are not limited to hematopoietic stem cells, mammary stem cells, intestinal stem cells, mesenchymal stem cells, endothelial stem cells, neural stem cells, olfactory adult stem cells, neural crest stem cells, and testicular cells.
[0151] In some embodiments, non-stem cells are used. Potentially, all of the 200 or so mammalian cell types within the body can be used in an implantable device as disclosed herein. Exemplary cells include but are not limited to, for example, cells found within a non- embryonic adult, such as insulin secreting cells (e.g., from adults or cadavers) or hepatocytes; islets of Langerhands; cells via somatic cell nuclear transfer (SCNT cells); cells via induced pluripotent stem cells (iPSs cells) either derived by genetic or chemical means; and cells from umbilical cord blood (UCB) cells.
[0152] In some embodiments, donor cells are used, including autologous (self) cells or non-autologous cells (e.g., allogenic or xenogenic cells from unrelated donors or other species).
[0153] In some embodiments, a combination of different types of cells can be used.
[0154] In some embodiments, the implantable device, which contains the cells to be transplanted, are made of synthetic biodegradable materials. In such embodiments, the implantable device can be implanted near a target organ at the time of transplant. For example, a device containing liver cells can be placed at the site where the diseased liver portion is removed. In such embodiments, the implantable device also contains the necessary biochemical composition for supporting the growth of liver cell therein. In such
embodiments, the cells can develop into liver tissue over a period of time within the mammal receiving the transplant as the device degrades over a longer period of time. [0155] In some embodiments, the cells are jump-started in a separate device and can develop into tissue elements before being transplanted into a live mammal. In such embodiments, the implantable device being transplanted into the live mammal does not need to have an observation module.
[0156] It will be understood that all types of organs/tissues can be used in a transplant or implant. Exemplary tissue/organ include but are not limited to liver tissue, pancreas tissue, kidney tissue, lung tissue, skin tissue, bone marrow and lymph node tissue, thyroid tissue, pituitary tissue, brain tissue, muscle tissue (including myocardium, skeletal muscle and smooth muscle), cartilage tissue, gastrointestinal tissues, and reproductive tissues including tissues from uterus and ovaries.
6.6.5 Implantable Devices as a Tool for Detecting Biomarkers or Biomolecules
[0157] In another aspect, an implantable device as disclosed herein is used for detecting known and unknown biomarkers or from within the device and outside the device within the body, e.g., vitamin levels, Cortisol levels, in addition to products relevant to the implant. As disclosed herein, a biomarker can be a small molecule as well as a
macromolecule, or a signal related to a small molecule or macromolecule. In medicine, a biomarker is a term often used to refer to a protein measured in blood whose concentration reflects the severity or presence of some disease state. More generally, a biomarker is anything that can be used as an indicator of a particular disease state or some other physiological state of an organism. Exemplary biomarkers for many diseases include but are not limited to various antigens or antibodies, serum LDL for cholesterol, blood pressure, Carcinoembryonic antigens for cancer. In cell biology, a biomarker is a molecule that is present or absent from a particular cell type. This facilitates the characterization of a cell type, their identification, and eventually their isolation. Cell sorting techniques are based on cellular biomarkers (for example, fluorescent-activated cell sorting). A biomarker can be used to identify a cell population, make a diagnosis, or measure the progress of disease or the effects of treatment. For example, the protein Oct-4 that is a biomarker found in embryonic stem cells; the Carcinoembryonic antigen (CEA) is a tumor marker used to follow up cancer treatment; and the Prostate Specific Antigen (PSA) is used for diagnosis of prostate cancer.
[0158] In some embodiments, biomarkers from all locations in contact with the device can be detected (e.g. locations that are internal and external to the device).
[0159] As an example, a multi-functional implantable device as shown in Figure 6 can be used for detecting a target biomarker or biomolecule. 6.6.6 Implantable Devices as an Extracorporeal Body Monitor and Modulator
[0160] In another aspect, an implantable device as disclosed herein can be attached to the body extracorporeally (e.g., outside the body, but attached through connection to the circulation etc.) The device can be used to grow or maintain cells ex vivo, it may be provided extracorporeally and yet be connected to a subject's vasculature. The device in a similar configuration can also be utilized to provide convenient vascular access, such as provided by a Portacath device, or similar system.
6.6.7 Methods of Use
[0161] In some embodiments, a bioreactor that is implanted under the dorsal skin can be employed to test this potentially powerful system. In some embodiments, the bioreactor for use in this protocol has an external facing glass "window" that remains facing external to the body. The glass window (made of specific microscope coverslip material) facilitates examination of blood vessel (and tissue) growth within the bioreactor in a living, but anesthetized animal by microscopy at regular intervals.
[0162] In some embodiments, the bioreactor contains matrix components, including an extracellular matrix commercially available for growing blood vessels in cultures with additional growth factors included (in this case bFGF) . The system can be implanted using known surgical procedures or as described herein. In some embodiments, an implanted system is allowed to grow blood vessels for 2 or more days; 5 or more days, 7 or more days; 10 or more days; 14 or more days; 21 or more days; 28 or more days; 36 or more days; 54 or more days; 60 or more days; 90 or more days; or even longer. Upon vessel development, animals can be anesthetized in a custom-made plexiglass container and the bioreactor can be viewed under the microscope for approximately 30 minutes. Daily imaging can be undertaken at 1, 7, 14 and 28 days; or at other intervals. In some embodiments, the animals can be sacrificed at the end of the implantation period (e.g., 28 days post
implantation), where the bioreactors can be disassembled and examined by conventional methods.
[0163] In some embodiments, the bioreactors can be implanted in Green
Fluorescent Protein (GFP) labeled C57BL/6 mice (GFP Strain 003115) mice. Matrigel with bFGF growth factors can be mixed within sterile bioreactors prior to their implantation into the dorsal skin of the mice. This allows fluorescent microscopic observation of the GFP labeled blood vessels that grow into the bioreactor against the dark background of the empty chamber.
[0164] In some embodiments, a chamber comprising a short cylinder (~3mm) of Grade 316 surgical stainless steel (Prototype 2.4) is implanted subcutaneously. In some embodiments, the chamber is a disc approximately 3 mm high and 8 mm wide. The top external-facing coverslip will be mounted on a stainless steel ring, to provide protection to the skin sutured in a 'cinch' up beneath it. The lumen of the chamber is filled with a protein gel (Matrigel) which incorporates angiogenic stimulants. The chamber was subcutaneously implanted on the midline at the mid back via an incision through the overlying skin such that it is implanted like a pacemaker, but with an external surface with an external window surrounded by a protective steel ring.
[0165] The implantation will be performed under general anesthesia with Isofiurane using aseptic techniques and recovery will be monitored carefully (available Surgical SOP). In some embodiments, a single 2 mg/kg dose of Metacam (aka Meloxicam) is administered prior to surgery. Previous experience has shown that mice recover very well from this surgery and no additional analgesic has been necessary. The wound will be closed with sutures which are 'cinched' around the tubing beneath the stainless steel ring. Morbidity will be carefully monitored on a daily basis and mice in any distress will be euthanized. In some embodiments, after 28 days, depending on cell growth into the chamber visible through the window, mice will be transferred to a custom made plexiglass anesthetic container and placed on a fluorescent microscope stage for imaging. In some embodiments, animals can be examined in initial tests for likely ~lhr, and once this is established, 20 minutes of anesthesia for imaging should be sufficient. Animals will be returned to their cages and given treats following imaging. Animals will be viewed under the microscope a maximum of 3X per week, and less if not needed.
[0166] Various experimental endpoints can be reached for the implantation. In some embodiments, cells are allowed to grow to a maximum volume into the bioreactors of 3mm after which the animals will be euthanized. In some embodiments, to reach this distance, bioreactors will be implanted for a maximum of 28 days.
6.6.8 Additional Embodiments
[0167] In one aspect, provided herein is a bioreactor device comprising a first surface being configured capable of fluidic communication with the extracellular tissues and tissue microvasculature of a subject and containing divided sub-compartments containing cells, matrix materials, and growth factors, with ports for delivering of these materials as needed, and the second surface being separated from the external environment by a window permitting observation intravitally.
[0168] In one aspect, provided herein is a method for delivering a cell population using the bioreactor device herein, with matrix materials, growth factors and pharmaceuticals to support its function stability.
[0169] In one aspect, provided herein is a bioreactor device comprising a first compartment being configured capable of fluidic communication with the extracellular matrix of a subject and configured for containing cells, implanted matrix and growth factors. The bioreactor makes it possible for blood vessels from the subject to grow into the device over time and provide further exchange with the body.
[0170] In another aspect, provided herein is a method of delivering a cell population into a subject in need thereof, and further division of the bioreactor into sub- compartments by a membrane, thereby delivering a cell population or multiple different cell populations and their necessary extracellular matrix and growth factors into a subject in need thereof.
[0171] In some embodiments with respect to any aspect disclosed herein, the permeable membrane permits passage of cells from existing tissues to the bioreactor.
[0172] In some embodiments with respect to any aspect disclosed herein, the permeable membrane enables passage of fluids and molecules to and from the interior of the bioreactor.
[0173] In some embodiments with respect to any aspect disclosed herein, each sub-compartment includes a cell injection port.
[0174] In some embodiments with respect to any aspect disclosed herein, at least one of the first compartment, the second compartment and the membrane is made of the same material. In some embodiments with respect to any aspect disclosed herein, at least one of the first compartment, the second compartment and the membrane is made of different materials.
[0175] In some embodiments with respect to any aspect disclosed herein, the first compartment and/or the membrane comprise at least one pharmaceutical agent.
[0176] In some embodiments with respect to any aspect disclosed herein, the pharmaceutical agent is a therapeutic agent or a diagnostic agent. [0177] In some embodiments with respect to any aspect disclosed herein, embodiments the polymer fibers have spaces with a cutoff of about between 0.5 - 400 micrometers.
[0178] In some embodiments with respect to any aspect disclosed herein, the at least one of the first compartment and the second compartment is made of non-woven polymer fibers.
[0179] In some embodiments with respect to any aspect disclosed herein, the cell population comprises an insulin-secreting cell population.
[0180] In some embodiments with respect to any aspect disclosed herein, the cell population comprises Islets of Langerhans, thyroid cells, hepatocytes, hematopoietic cells, lymphoid cells or other cell populations found within the body.
[0181] In some embodiments with respect to any aspect disclosed herein, the cell population comprises embryonic stem cells of any species origin.
[0182] In some embodiments with respect to any aspect disclosed herein, the cell population comprises cells derived by somatic cell nuclear transfer.
[0183] In some embodiments with respect to any aspect disclosed herein, the cell population comprises cells derived as induced pluripotent stem cells, by genetic or chemical means.
[0184] In some embodiments with respect to any aspect disclosed herein, the cell population comprises cells altered by genetic methods.
[0185] In some embodiments with respect to any aspect disclosed herein, the cell population comprises cells derived from adult tissues and from adult stem cell populations.
[0186] In some embodiments with respect to any aspect disclosed herein, the cell population comprises cells of other types derived by genetic or synthetic methods.
[0187] In some embodiments with respect to any aspect disclosed herein, the matrix materials comprises matrix derived from living cell sources.
[0188] In some embodiments with respect to any aspect disclosed herein, the matrix materials comprises matrix derived from mixed components of individual matrix elements.
[0189] In some embodiments with respect to any aspect disclosed herein, the matrix materials comprises matrix derived from decellularized matrices of cadaver tissues.
[0190] In some embodiments with respect to any aspect disclosed herein, the step of introducing the cell population, matrix materials, growth factors or pharmaceuticals into the sub-compartment is affected prior to the step of implanting the device to next to the micro vasculature of the subject.
[0191] In some embodiments with respect to any aspect disclosed herein, the step of introducing the cell population, matrix materials, growth factors or pharmaceuticals into the sub-compartments is affected following the step of implanting the device next to the micro vasculature of the subject.
[0192] In some embodiments with respect to any aspect disclosed herein, the bioreactor also provides a subcutaneously implanted device that provides a system allowing multiple punctures through the surface skin into the device. This will provide subcutaneous access to cells and tissues within the bioreactor for infusion of additional cells, growth factors, matrix components or pharmaceuticals as needed.
[0193] In some embodiments with respect to any aspect disclosed herein, there includes a biosensor system with fluidic or other connection to each of the compartments in the bioreactor as well as connection to the interior body compartment also adjacent to the device.
[0194] In some embodiments with respect to any aspect disclosed herein, the biosensor system includes capabilities for detection of biomarkers molecules indicative of normal or abnormal states of cells within the device, or from within the internal body that is also adjacent to the bioreactor.
[0195] In some embodiments with respect to any aspect disclosed herein, the biosensor system includes capabilities for wireless transmission of biomarkers from within the device to electronic devices outside the body to facilitate detection and care of tissues within the device, and as a diagnostic tool for normal and diseased biological processes occurring in the body, and as a diagnostic to detect the influence of other interventions on the body be they pharmaceutical or otherwise.
[0196] Having described the invention in detail, it will be apparent that modifications, variations, and equivalent embodiments are possible without departing the scope of the invention defined in the appended claims. Furthermore, it should be appreciated that all examples in the present disclosure are provided as non- limiting examples.
7. EXAMPLES
[0197] The following non-limiting examples are provided to further illustrate embodiments of the invention disclosed herein. It should be appreciated by those of skill in the art that the techniques disclosed in the examples that follow represent approaches that have been found to function well in the practice of the invention, and thus can be considered to constitute examples of modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
EXAMPLE 1
Device Design
[0198] This example provides a murine platform comprising four elements: a container, a container cap, an installation device, and a mouse microscopy stage. The specific design of the device can be found in Figures 9A-9C.
1. Container
[0199] The container is made of biocompatible material (e.g., PMMA, MED610 or Grade 316 Surgical Steel); see for example, Figures 9-11 A, 10 and 6B. It is implanted intradermally on the back of a mouse with the sides of the container in direct contact with the underside of the dermal layers of the skin. The sides of the container include 50-200-micron mesh holes to facilitate tissue growth from the skin into the internal reservoir, through the mesh holes. The container features a wide rim at the base below the holes and under the skin to hold it in place once implanted.
[0200] The container houses a reservoir of approximately ΙΟΟμΙ of gelatinous protein mixture (matrix). The container is envisioned to feature fluidic injection ports that are compatible with standard pipet P200 and/or PI 000 tip used to introduce material into the reservoir at volumes between 20-200μ1. The injection ports are used initially to completely fill the reservoir with the matrix mixture after the container has been implanted so that the matrix is in full contact with the mesh holes on the side of the container and there are no air bubbles within the reservoir. Subsequently, injection ports may be used to introduce additional materials into the matrix. The injection ports will introduce materials into the matrix at different locations on the x and z axes. The injection ports must not allow materials to leak back out of the reservoir through the ports. It is envisioned that subsequently introduced material will rinse/wash materials in the matrix out into the surrounding tissue through the mesh holes.
[0201] Attached to the container's open top is a No. 1 coverslip glass between 0.13-0.17 mm in thickness (e.g., element 10-c in Figure 9B) and having a diameter of about 8 mm, which is flush against the matrix when the reservoir is filled, with no air bubbles, and is held in place by a retaining clip (or circlip; e.g., element 10-a in Figure 9B) or equivalent and sealed with an O-ring or equivalent to prevent leaks (e.g., element 10-b in Figure 9B). Tissue growth will occur immediately beneath the covers lip. Opening the covers lip is envisioned to accommodate removal of the entire matrix through the top of the container.
[0202] The visible top surface of the container contains a stable registration element that can be used as a point of reference for microscopy, facilitating repeat imaging of the same microscopic location in serial imaging sessions. This could be implemented as a dot or indentation on the visible top surface of the container.
[0203] Built into the design of the container is a mechanical interface to allow attachments on top of the container above the coverslip glass. The coverslip and the registration element must be unobstructed by the mechanical interface to facilitate imaging the live tissue in some attachment applications. Examples of attachments include a cap to protect the coverslip glass, a stage for microscopy, an irradiation system.
2. Container Cap
[0204] The container cap attaches to the top of the container via the container's mechanical interface and protects the coverglass slip. Its top outside surface includes space for a registration label.
3. Installation Device
[0205] The mechanism of implantation of the container into the skin envisioned is a grommet-style 'hole punch' rivet system using an external manual tool.
4. Mouse Microscopy Stage
[0206] The mouse microscopy stage is envisioned to comprise a stable base upon which the mouse will stand/lay with an element that will attach the base above the mouse to the implanted intradermal container via the container's mechanical interface thereby stabilizing the container against mouse movement during microscopy. The coverslip and the registration element must be unobstructed by the stage and the mechanical interface.
EXAMPLE 2
Implantable Device in Mice
[0207] Implantable devices were prepared and soaked in 100% ethanol for 20 minutes before drying within a tissue culture hood. Frozen Matrigel reagent was thawed at 4 degrees Celsius, stored on ice, and then mixed with 175 ng/mL basic Fibroblast Growth Factor in 5% bovine serum albumin prior to injection into the device. The device has a 12 mm No.1 glass covers lip placed into the top aspect of the device, and is closed with a retainer clip (e.g., Figures 9A-9C).
[0208] A one-centimeter incision was made in the skin at the location of implantation on the recipient mice near at the dorsal aspect of the chest. Each device was positioned in a subcutaneous pocket and closed using a circular synch 5-0 non-absorbable Proline suture. Alternately, the device was implanted surgically, and internal biochemical matrix was then added through the top opening of the device, before closure with the coverslip and retaining ring (e.g., Figures 10A and 10B).
[0209] The implantation was performed under general anesthesia with Isoflurane anesthetic using aseptic techniques. Recovery was monitored carefully. A single dose of Metacam (aka Meloxicam) at 2 mg/kg was administered prior to surgery. Mice recovered very well from the surgery; and no additional analgesic was necessary. The wound was closed with sutures that were "cinched" around the tubing beneath the stainless steel ring. After implantation, blood and serous fluid entering and exiting the device were observed and monitored through direct observation from the observation window.
EXAMPLE 3
Imaging in Live Mammals
[0210] After 7 or 14 days, when the cell growth within the chamber became visible through the observation window, mice were transferred to a custom made plexiglass anesthetic container and placed on a fluorescent microscope stage for imaging, (e.g., Figures 11 A-l ID) Animals were examined in initial tests for 0.5-1.0 hr. Once an initial test was established, 20 minutes of anesthesia was sufficient for imaging.
[0211] Figures 1 IB and 11C depict a system for live imaging which include an animal/mammal observation station, a microscope and a computer system for collecting, depicting and processing the image data.
[0212] As shown in Figure 1 ID, live cells within the implanted device are visible after being stained with Hoechst 33342 nuclear stain.
EXAMPLE 4
Model and Prototypes
[0213] Plastic Subcutaneous Model: This model was encased in nylon lOOum mesh. The device underwent full subcutaneous implant, which allowed ex vivo analysis post-sacrifice. The device was able to remain in animal indefinitely. Figures 12A through 12C illustrate the results of vessel growth using this prototype.
[0214] Modified Plastic Model: This model was used for multiple endpoint analysis. Methodologies for removal of implant were developed for this model to allow utilization of fluorescent microscopy, implant digestion and rapid analysis by flow cytometry, and microCT analysis for significant improvement of data acquisition from the device.
Detailed demonstration of incorporation of multiple cell types into the device and kinetics of tissue formation over time. With this model, alternate matrix materials were used to support human cells within the device. The device was use in transgenic mice to demonstrate unique animal phenotypes. Figures 12D through 12E illustrate the results of cell and vessel growth using this prototype.
[0215] Galvanized Steel Model: This was the first stainless steel model that was implanted in live animals (e.g., Figures 13A and 13B; Figures 9A-9C). The device provides grooves for skin attachment, an encasing for 12mm coverglass and retaining clip. Proline, ethylon and vicryl sutures were tested for implantation. The device remained in animal for up to 4 weeks. Several challenges existed in this model. For example, the device was not completely biocompatible. The unit became rusted after several uses. There were some difficulties maintaining position in skin. Some coverglass was broken during experiments due to contact with cage elements, and animal interacting with the device. Some devices dislodged from animals. Occasional air bubbles also created problems.
[0216] Surgical Steel Prototype: This model was made with substantially harder material, which was more difficult to mill (e.g., Figures 13C and 13D, Figures 9B and 9C). The device provided an enlarged bottom groove lip to retain position under the skin once implanted. It also provided an encasement notch for an 8mm coverglass and a retaining clip. The device was fully biocompatible and required less matrix material for improved cost effectiveness.
[0217] Smaller 8mm Surgical Stainless Steel Model: This model provides a smaller design from previous 12mm implants, which reduced weight and size of device (e.g., Figure 13E). AutoCAD design prepared for biocompatible lighter multi-well and rendered 3D printed prototypes possible (e.g., Figures 2A-2C). This fabrication process allowed for more rapid prototyping and integration of smaller features.
[0218] Model with Surgical Interface Improvements: This model is smaller and lighter (e.g., Figure 13F). It provided an enlarged bottom groove lip to retain position under the skin once implanted. The device also provided encasement notch for an 8mm cover- glass, a retaining clip and a screw clip with sealant. This design required less matrix material for improved cost effectiveness. When implanted, first live blood vessels were visible in transgenic mice (e.g., Figure 1 ID).
[0219] Lasercut Acrylic Prototype: Lasercutting methods were used to create chamber in clear acrylic. Fabrication by this type of technology produced very nice clear implants. Figure 13G depicts exemplary laser rings and baseplate template provided to the laser cutter and used to produce the Lasercut Acrylic Prototype.
[0220] With all models, hydrogels as a matrix component were added and could go inside the implants; for example, human synthetic matrixes offer the advantages of not causing damages to the host immune system. Additions could be made to the window coverglass surface. The retainer ring for closing the top of the implant could be replaced with part of a coil of a spring. Commercial glue products such as 'fish tank glue' were used to close the coverglass seal, which did not appear to cause any problems with immunity and rendered an airtight seal in the device chamber.
EXAMPLE 5
Implant Study
[0221] The model shown in Figure 13F was tested in two groups of transgenic mice: group 1) three Mice had GFP (Green fluorescent protein expressed in all their cells), implants with normal matrix; and group 2) three Mice had GFP (Green fluorescent protein expressed in all their cells), implants with matrix, spiked with 5X concentration of bFGF (a pro-angiogenic growth factor).
[0222] At 4 weeks when anesthetizing these mice and viewing them under a standard fluorescent microscope, GFP+ fluorescent cells were observed in group 1. GFP positive cells (likely immune cells) were shown in Figures 14A and 14B, but there was no obvious evidence of vessels. The images were collected in grayscale and changed to green in post processing (not shown).
[0223] In addition to single GFP+ cells, in group 2 there was also evidence of extensive vasculature in the chambers, in a live mouse on repeated imaging sessions. The vasculature is not stained, but is clearly evident as the darker hemoglobin running through the chamber in new capillaries inside the implants:
[0224] As depicted in Figure 15 A, live vessels were clearly seen here in black at low power. The rim of the implant is also visible in the shot. The brightest white in the image is fluorescent cells that are building the new blood vessel. There is also a clear fluid bubble just below the vasculature indicating their leaky edematous state as expected. Figure 15B depicts a closeup view of the branching capillary inside live chamber with GFP positive cells nearby in white.
[0225] As depicted in Figure 15C, in other areas of the implant, evidence of blood was observe, which was only possible if capillaries were leading to these new (previously non- vascular) areas that are at least 2-3mm away from the pre-existing vasculature in the animal. More evidence of capillaries grown to the surface of the chamber was observed in another mouse from group 2 (Figures 15D and 15E). In Figure 15E, the image was processed and recolored to more clearly show the green fluorescent cells inside alongside the vessels. Figure 15F depicts another closeup view beside the rim of the device clearly showing the capillaries.
[0226] Note that in all new implants at day 1 , there was no GFP signal and no dark hemoglobin in the device at all. All tissues seen in these images were new live growing tissues observed in a living animal.
[0227] The various methods and techniques described above provide a number of ways to carry out the invention. Of course, it is to be understood that not necessarily all objectives or advantages described may be achieved in accordance with any particular embodiment described herein. Thus, for example, those skilled in the art will recognize that the methods can be performed in a manner that achieves or optimizes one advantage or group of advantages as taught herein without necessarily achieving other objectives or advantages as may be taught or suggested herein. A variety of advantageous and disadvantageous alternatives are mentioned herein. It is to be understood that some preferred embodiments specifically include one, another, or several advantageous features, while others specifically exclude one, another, or several disadvantageous features, while still others specifically mitigate a present disadvantageous feature by inclusion of one, another, or several advantageous features.

Claims

WHAT IS CLAIMED IS:
1. A system for maintaining mammalian cells in a live mammal, comprising:
i) an implantable device comprising:
a housing having a plurality of sides, wherein one side of the plurality of sides comprises a structural feature for securely attaching the implantable device to a site of implantation in the mammal;
a compartment contained within the housing, wherein once implanted in the mammal the compartment is capable of exchanging content, via an interaction module on at least one side of the plurality of sides, with an extracellular space or a tissue microvasculature of the live mammal; and
an observation module, wherein the observation module is located on one side of the plurality of sides through which the plurality of cells can be observed by an optical equipment; and
ii) a plurality of cells contained within the compartment of the implantable device, wherein the plurality of cells are suspended in a biochemical composition comprising a cellular matrix, wherein the maintenance of the plurality of cells is supported partially by the biochemical composition.
2. The system of claim 1 , wherein the structural feature for securely attaching the
implantable device comprises a groove on one side of the plurality of sides of the implantable device.
3. The system of claim 1 , wherein the structural feature for securely attaching the
implantable device comprises a plurality of holes on at least one side of the plurality of sides of the implantable device.
4. The system of claim 1 , wherein the implantable device further comprises a plurality of sub-compartments within the compartment, and wherein the plurality of cells is contained within at least one sub-compartment of the plurality of sub-compartments.
5. The system of claim 4, wherein sub-compartments within the plurality of sub- compartments are connected via one or more channels, and wherein at least one of the connected sub-compartment contains the plurality of cells.
6. The system of claim 5, wherein the connected sub-compartments, upon exposure to one or more biochemical reagents, are configured to perform an assay on the plurality of cells.
7. The system of claim 6, wherein the assay detects the presence or absence of a biomarker, and wherein the biomarker is selected from the group consisting of a protein, a peptide, a gene, and a nucleic acid molecule.
8. The system of claim 1 , wherein the implantable device further comprises an injection port on one side of the plurality of sides, through which biochemical reagents can be added into or removed from the compartment.
9. The system of claim 8, wherein the injection port and observation module are on the same side of the plurality of sides.
10. The system of claim 1, wherein the interaction module comprises a permeable membrane between an opening on the at least one side of the plurality of sides of the implantable device.
11. The system of claim 1 , wherein the interaction module is an opening on the implantable device, which allows content within the device to exchange with the cellular environment where the implantable device is implanted.
12. The system of claim 1, wherein the implantable device further comprises a wireless module for transmitting a signal from the implantable device to an external receiver or receiving a signal from an external controller, after the implantable device is implanted in a mammal.
13. The system of claim 12, wherein the wireless module is for transmitting a signal from the implantable device to an external receiver and receiving a signal from an external controller.
14. The system of claim 1, wherein the biochemical composition further comprises one or more growth factors.
15. An imaging system for collecting data from a live mammal, comprising: the system of claims 1-14; and
an optical equipment for collecting image or video data of the content with the compartment or sub-compartments of the implantable device.
16. The imaging system of claim 15, wherein the optical equipment is selected from the group consisting of a microscope, a fluorescence imaging device, a thermal imaging device, a radio imaging device, and a combination thereof.
17. The imaging system of claim 15, further comprising:
a computer system for collecting, storing, displaying, and processing the image or video data.
18. A method for collecting data from a live mammal using an imaging system, comprising: maintaining the growth of a plurality of cells in a live mammal using the system of claim 1 , wherein the implantable device is implanted in a live mammal such that at least a portion of the observation module is not embedded within the live mammal and wherein data of the content within the implantable device are measured using an optical equipment; and collecting data of the content within the implantable device using an optical equipment.
19. The method of claim 18, wherein the optical equipment is selected from the group consisting of a microscope, a fluorescence imaging device, a thermal imaging device, and a radio imaging device.
20. The method of claim 18, wherein the data collected comprise images or videos of the plurality of cells in the implantable device.
21. A drug testing system, comprising:
the system of claim 4, wherein each sub-compartment of the plurality of sub- compartments comprises a population of cells; wherein the cells are taken from a patient; wherein a compound is added to one or more sub-compartments of the plurality of sub- compartments.
22. A method for drug testing using the system of claim 4, comprising: placing, in each sub-compartment of the plurality of sub-compartments of the implantable device, a population of cells, wherein the cells are derived from a biopsy sample of a target disease, and wherein the implantable device is implanted in a live mammal;
adding a test compound in one or more sub-compartments of the plurality of sub- compartments; and
collecting data from each sub-compartment of the plurality of sub-compartments.
23. The method of claim 22, further comprising:
comparing data from different sub-compartments to determine the presence of differences between the sub-compartments; and
correlating any differences with the presence of the compound in each sub- compartment.
24. The method of claim 22, wherein the biopsy sample is associated with a type of cancer.
25. The method of claim 22, wherein each sub-compartment of the plurality of sub- compartments contains the same type of cells and wherein cells in each sub-compartment have approximately the same quantity.
26. The method of claim 22, wherein at least two different compounds are added to two different sub-compartments.
27. A method for producing a desired biomolecule in a live mammal using the system of claim 1, comprising:
maintaining the growth of the plurality of cells in the live mammal using the implantable device, wherein the implantable device is implanted in a live mammal and wherein cells of the plurality of cells are capable of producing the desired biomolecule; adding one or more reagents to the implantable device, wherein the reagents are necessary for the plurality of cells to produce the desired biomolecule; and
producing the desired biomolecule.
28. The method of claim 27, wherein the cells are pancreas cells and the desired biomolecule is insulin.
29. A method for growing cells or tissues for transplant in a live mammal using the system of claim 1, comprising:
maintaining the growth of the plurality of cells in the live mammal using the implantable device, wherein the implantable device is implanted in a live mammal such that at least a portion of the observation module is not embedded within the live mammal and wherein data of the content within the implantable device are measured using an optical equipment, and wherein cells of the plurality of cells are selected from the group consisting of stem cells, embryonic stem cells, adult stem cells and non-stem cells;
measuring, via the observation module, data of the cells of the plurality of cells to determine whether the cells are suitable for transplant;
removing the cells from the implantable device if the cells are suitable for transplant; and
transplanting the cells in a mammal.
PCT/US2013/023296 2012-01-27 2013-01-25 Implantable devices and applications and use thereof WO2013112936A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201261591751P 2012-01-27 2012-01-27
US61/591,751 2012-01-27
US201261592103P 2012-01-30 2012-01-30
US61/592,103 2012-01-30
US201261618485P 2012-03-30 2012-03-30
US61/618,485 2012-03-30

Publications (1)

Publication Number Publication Date
WO2013112936A1 true WO2013112936A1 (en) 2013-08-01

Family

ID=48873976

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/023296 WO2013112936A1 (en) 2012-01-27 2013-01-25 Implantable devices and applications and use thereof

Country Status (1)

Country Link
WO (1) WO2013112936A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2223720C2 (en) * 2002-02-26 2004-02-20 Колганов Андрей Сергеевич Device for implanting cellular culture possessing excretory function
JP2007007414A (en) * 2005-06-29 2007-01-18 Lifescan Inc Multi-compartment delivery system

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2223720C2 (en) * 2002-02-26 2004-02-20 Колганов Андрей Сергеевич Device for implanting cellular culture possessing excretory function
JP2007007414A (en) * 2005-06-29 2007-01-18 Lifescan Inc Multi-compartment delivery system

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MATTHEW J. FARRAR ET AL.: "Chronic in vivo imaging in the mouse spinal cord using an implanted chamber", NAT METHODS, vol. 9, no. 3, 22 January 2012 (2012-01-22), pages 297 - 302, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3429123> [retrieved on 20130418] *

Similar Documents

Publication Publication Date Title
Bhattacharjee et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts
AU2016204936B2 (en) Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
Machingal et al. A tissue-engineered muscle repair construct for functional restoration of an irrecoverable muscle injury in a murine model
US11873514B2 (en) Method of screening for a substance that acts on a cell mass
CN101918532B (en) For culture systems and the method for vitro test immunogenicity and immunologic function
KR102152492B1 (en) Method for culturing 3D lung cancer organoid and preparing patient-derived xenograft model using thereof
CN106795225A (en) Alginates and application for the modification of anti-fibrosis material
Jungebluth et al. Tracheal tissue engineering in rats
US11559607B2 (en) Living devices for replacement of organs or specific organ functions, methods and uses of the same
Xiao et al. Hyaluronic-acid based hydrogels for 3-dimensional culture of patient-derived glioblastoma cells
Gosztyla et al. A comparison of sterilization techniques for production of decellularized intestine in mice
US10695379B2 (en) Cell population seeding in dermal matrices for endocrine disorder management
Kivrak Pfiffner et al. A new in vivo magnetic resonance imaging method to noninvasively monitor and quantify the perfusion capacity of three-dimensional biomaterials grown on the chorioallantoic membrane of chick embryos
WO2013112936A1 (en) Implantable devices and applications and use thereof
WO2006057444A1 (en) Method of automatically evaluating differentiation degree of cells
Portillo‐Esquivel et al. z‐Wire: A Microscaffold That Supports Guided Tissue Assembly and Intramyocardium Delivery for Cardiac Repair
WO2021038038A1 (en) Multi-cell culture device and kit
WO2020243832A1 (en) Tissue constructs with biocompatible linear scaffolds, methods of making and uses thereof
Modulevsky et al. Plant scaffolds support motor recovery and regeneration in rats after traumatic spinal cord injury
Chen et al. Optical tissue window: a novel model for optimizing engraftment of intestinal stem cell organoids
Fisher The Cytoseeder TM—A Microneedle Device for Seeding Tissue Engineered Scaffolds
Duek et al. In vitro and in vivo cell tracking of PKH26-labeled osteoblasts cultured on PLDLA scaffolds
CN101912636B (en) Miniature skin structure, preparation method and use thereof
De Robertis et al. Mouse Melanoma Model in Tumor Vaccines and Immunotherapy Research
Archer Applications of MRI in Tissue Engineering: Environmental Control and Noninvasive Culture Surveillance

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13740570

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13740570

Country of ref document: EP

Kind code of ref document: A1