TEMPERATURE-CONTROLLED MULTI-MATERIAL OVERPRINTING

A three-dimensional (3D) bioprinting method and system are disclosed. The method includes disposing/immersing a printing platform or surface into a first bioink, such as a bioink resin, curing one or more layer of the first bioink resin onto the printing platform or surface, and removing the printing platform or surface from the first bioink resin. The process is repeated with a second bioink resin such that the second bioink resin is cured on top of the one or more layer of first bioink resin, and can be further repeated with a third or even fourth bioink resin. By varying constituents of one or more or each bioink resin (such as living cell type or polymer), complex, multilayered tissues can be engineered. A system capable of performing the method is also disclosed.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application is a Divisional application of U.S. patent application Ser. No. 17/011,767, filed Sep. 3, 2020, which relies on the disclosure of and claims priority to and the benefit of the filing date of U.S. Provisional Patent Application No. 62/897,188 filed Sep. 6, 2019, which applications are hereby incorporated by reference herein in their entireties.

BACKGROUND OF THE INVENTION Field of the Invention

The present invention is directed to the field of three-dimensional (3D) bioprinting. More particularly, embodiments of the invention are directed to systems and methods which utilize digital light processing (DLP) and multiple bioink resin tanks containing one or more photopolymerizable resins to provide for multi-material overprinting on a printing surface.

Description of Related Art

In order to replicate complex tissue structures such as organs, it is desirable to build tissue strata having constituents and properties which differ from previous layers. Current technologies for 3D bioprinting including extrusion, inkjet, acoustic/ultrasound, laser, and stereolithography technologies fall short of this goal. Thus, new technologies for 3D bioprinting are urgently needed.

SUMMARY OF THE INVENTION

Toward this end, a method of three-dimensional (3D) bioprinting is disclosed, which method includes immersing a printing platform or surface into a first bioink resin, curing one or more layer of the first bioink resin onto the printing platform or surface, and removing the printing platform or surface from the first bioink resin. The process is repeated with a second bioink resin such that the second bioink resin is cured on top of the one or more layer of first bioink resin, and can be further repeated with a third or even fourth bioink resin. By varying the composition of one or more or each bioink resin such as living cell type or polymer, complex, multilayered tissues can be engineered.

A system for three-dimensional (3D) bioprinting is also disclosed which is capable of performing the above method. The system includes a printing platform or surface, first and second container, one or more source of electromagnetic radiation, one or more actuator capable of horizontal, vertical, and/or rotary movement of the printing platform or surface and/or first container and/or second container and/or one or more source of electromagnetic radiation, and a controller capable of sending commands to the one or more source of electromagnetic radiation and one or more actuator.

Embodiments include Aspect 1, which is a method of three-dimensional (3D) bioprinting comprising immersing a printing platform or surface into a first bioink resin, curing one or more layer of the first bioink resin on the printing platform or surface, and removing the printing platform or surface from the first bioink resin.

Aspect 2 is a method of three-dimensional (3D) bioprinting comprising mechanically lowering a printing platform or surface into a first container irradiating the printing platform or surface with electromagnetic energy, and mechanically raising the printing platform or surface from the first container.

Aspect 3 is the method of Aspect 1, further comprising immersing the printing platform or surface into a second bioink resin; and curing one or more layer of the second bioink resin.

Aspect 4 is the method of Aspect 2, further comprising mechanically lowering the printing platform or surface into a second container.

Aspect 5 is the method of Aspect 4, further comprising irradiating the printing platform or surface with electromagnetic energy.

Aspect 6 is the method of any one of Aspects 2, 4, or 5, wherein the first container comprises a first bioink resin and the second container comprises a second bioink resin.

Aspect 7 is the method of Aspect 6, wherein the electromagnetic energy is capable of curing one or more layer of the first bioink resin and/or second bioink resin.

Aspect 8 is the method of any of Aspects 1-7, wherein the first bioink resin is the same as the second bioink resin.

Aspect 9 is the method of any of Aspects 1-8, wherein the first bioink resin is different from the second bioink resin.

Aspect 10 is the method of any of Aspects 1-9, wherein curing one or more layer of the first and/or second bioink resin comprises polymerizing one or more monomer within the first and/or second bioink resin.

Aspect 11 is the method of any of Aspects 1-10, further comprising irradiating the printing platform or surface or a portion thereof with electromagnetic energy to initiate curing of the first and/or second bioink resin and/or polymerization of the one or more monomer.

Aspect 12 is the method of any of Aspects 1-11, wherein the electromagnetic energy comprises light.

Aspect 13 is the method of any of Aspects 1-12, wherein curing one or more layer of the first and/or second bioink resin comprises irradiating the first and/or second bioink resin with light from one or more digital light processing (DLP) projector.

Aspect 14 is the method of any of Aspects 1-13, wherein the light is ultraviolet, visible, or infrared.

Aspect 15 is the method of any of Aspects 1-14, wherein curing one or more layer of the first and/or second bioink resin comprises photopolymerization of a monomer to form one or more polymer.

Aspect 16 is the method of any of Aspects 1-15, wherein the one or more polymer is a biopolymer.

Aspect 17 is the method of any of Aspects 1-16, wherein the one or more polymer is a synthetic polymer.

Aspect 18 is the method of any of Aspects 1-17, wherein the polymer formed from the first bioink resin is the same as the polymer formed from the second bioink resin.

Aspect 19 is the method of any of Aspects 1-18, wherein the polymer formed from the first bioink resin is different from the polymer formed from the second bioink resin.

Aspect 20 is the method of any of Aspects 1-19, wherein photopolymerization is initiated by one or more photo-initiator present in the first and/or second bioink resin.

Aspect 21 is the method of any of Aspects 1-20, wherein photopolymerization is controlled by one or more photo-absorber present in the first and/or second bioink resin.

Aspect 22 is the method of any of Aspects 1-21, wherein the first bioink resin and/or second bioink resin comprises one or more living cells.

Aspect 23 is the method of any of Aspects 1-22, wherein the living cells of the first bioink resin are the same as the living cells of the second bioink resin.

Aspect 24 is the method of any of Aspects 1-23, wherein the living cells of the first bioink resin are different from the living cells of the second bioink resin.

Aspect 25 is the method of any of Aspects 1-24, wherein the first bioink resin and/or second bioink resin comprises one or more therapeutic agents or particles.

Aspect 26 is the method of any of Aspects 1-25, wherein the one or more therapeutic agents or particles of the first bioink resin are the same as the one or more therapeutic agents or particles of the second bioink resin.

Aspect 27 is the method of any of Aspects 1-26, wherein the one or more therapeutic agents or particles of the first bioink resin are different from the one or more therapeutic agents or particles of the second bioink resin.

Aspect 28 is the method of any of Aspects 1-27, wherein the one or more therapeutic agents or particles comprise a biologic.

Aspect 29 is the method of any of Aspects 1-28, wherein the one or more therapeutic agents or particles comprise a small molecule.

Aspect 30 is the method of any of Aspects 1-29, further comprising immersing the printing platform or surface into a third bioink resin and curing one or more layer of the third bioink resin.

Aspect 31 is the method of any of Aspects 1-30, wherein the immersing and removing of the printing platform or surface or mechanically lowering and mechanically raising of the printing platform or surface is by way of one or more actuator.

Aspect 32 is the method of any of Aspects 1-31, wherein the one or more actuator is/are capable of rotary and/or vertical motion.

Aspect 33 is the method of any of Aspects 1-32, further comprising consecutively positioning the first bioink resin and second bioink resin or first container and second container below the printing platform or surface.

Aspect 34 is the method of any of Aspects 1-33, wherein the consecutive positioning is by way of one or more actuator.

Aspect 35 is the method of any of Aspects 1-34, wherein the one or more actuator is capable of rotary and/or horizontal motion.

Aspect 36 is the method of any of Aspects 1-35, wherein immersing the printing platform or surface into the first bioink resin comprises mechanically lowering the printing platform or surface into the first container.

Aspect 37 is the method of any of Aspects 1-36, wherein curing the one or more layer of the first bioink resin on the printing platform or surface comprises irradiating the printing platform or surface with electromagnetic energy.

Aspect 38 is the method of any of Aspects 1-37, wherein removing the printing platform or surface from the first bioink resin comprises mechanically raising the printing platform or surface from the first container.

Aspect 39 is the method of any of Aspects 1-38, wherein immersing the printing platform or surface into the second bioink resin comprises mechanically lowing the printing platform or surface into the second container.

Aspect 40 is the method of any of Aspects 1-39, wherein the first container is mechanically removed and the second container is mechanically positioned below the printing platform or surface prior to lowering.

Aspect 41 is the method of any of Aspects 1-40, wherein the printing platform or surface is inflatable and optionally further comprising inflating or deflating the printing platform or surface.

Aspect 42 is the method of any of Aspects 1-41, further comprising controlling the temperature of the printing platform or surface or first and second bioink resin or first and second container by way of one or more temperature control unit.

Aspect 43 is a system for three-dimensional (3D) bioprinting comprising a printing platform or surface, first and second containers, one or more source of electromagnetic radiation, one or more actuator capable of horizontal, vertical, and/or rotary movement of the printing platform or surface and/or the first container and/or second container and/or the one or more source of electromagnetic radiation, and a controller capable of sending commands to the one or more source of electromagnetic radiation and the one or more actuator.

Aspect 44 is the system of Aspect 43, or a method, wherein the system is configured to perform or the method comprises the steps of:

a) positioning the first container below the printing platform or surface; and/or

b) mechanically lowering the printing platform or surface into the first container; and/or

c) irradiating the printing platform or surface with electromagnetic energy; and/or

d) mechanically raising the printing platform or surface from the first container; and/or

e) positioning the second container below the printing platform or surface; and/or

f) mechanically lowering the printing platform or surface into the second container; and/or

g) irradiating the printing platform or surface with electromagnetic energy; and/or

h) mechanically raising the printing platform or surface from the second container.

Aspect 45 is the system or method of Aspect 44, wherein the system is configured to perform or the method comprises the steps of any of steps a through h consecutively, such as steps a and b, steps a through c, steps a through d, steps a through e, steps a through f, steps a through g, or steps a thorough h.

Aspect 46 is the system of claim 43, wherein the one or more actuator is configured to raise and lower the printing platform or surface by way of vertical and/or rotary motion and position the first container or second container below the printing platform or surface by way of horizontal and/or rotary motion.

Aspect 47 is the system of any of Aspects 43-46, wherein the one or more actuator comprises one or more linear or rotary actuators.

Aspect 48 is the system of any of Aspects 43-47, wherein the one or more actuators are pneumatic, hydraulic, electric and/or mechanical actuators.

Aspect 49 is the system of Any of Aspects 43-48, wherein the system is configured to perform the method of any preceding claim.

Aspect 50 is the system of any of Aspects 43-49, wherein the one or more source of electromagnetic energy is one or more light source, optionally one or more digital light processing (DLP) projector.

Aspect 51 is the system of any of Aspects 43-50, wherein the controller comprises one or more processor and a non-transitory computer readable storage medium comprising one or more 3D files having instructions capable of being read by the processor.

Aspect 52 is the system of any of Aspects 43-51, wherein the electromagnetic energy is light, such as UV, visible, or infrared light.

Aspect 53 is the system of any of Aspects 43-52, wherein the one or more digital light processing (DLP) projector are positioned or positionable above, below, and/or at one or more sides of the printing platform or surface and/or first and/or second container.

Aspect 54 is the system of any of Aspects 43-53, wherein the printing platform or surface is capable of inflation or deflation.

Aspect 55 is the system of any of Aspects 43-54, further comprising a tank of compressed air or inert gas or air or inert gas compression mechanism.

Aspect 56 is the system of any of Aspects 43-55, further comprising one or more temperature control unit in operable communication with the printing platform or surface and/or first and/or second containers and/or a platform or stage holding the first and/or second containers.

Aspect 57 is the system or method of any of Aspects 1-56, which incorporates structure or methods step(s) for extruding bioink.

BRIEF DESCRIPTION OF THE DRAWINGS

The accompanying drawings illustrate certain aspects of embodiments of the present invention, and should not be used to limit the invention. Together with the written description the drawings serve to explain certain principles of the invention.

FIG. 1 is a schematic diagram showing a print surface and the progression of different bioinks photocured on the surface with a side projector according to an embodiment.

FIG. 2A is a schematic diagram showing a top view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting.

FIG. 2B is a schematic diagram showing a top view of another embodiment of a multi-bioink resin tank system for 3D bio-overprinting.

FIG. 3A is a schematic diagram showing a side view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting with one projector.

FIG. 3B is a schematic diagram showing a side view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting with two projectors.

FIG. 4 is a schematic diagram showing multi-projector printing and the progression of different bioinks photocured on the printing surface according to an embodiment.

FIG. 5 is a schematic diagram of a print surface with an extrusion function according to an embodiment.

DETAILED DESCRIPTION OF VARIOUS EMBODIMENTS OF THE INVENTION

Reference will now be made in detail to various exemplary embodiments of the invention. It is to be understood that the following discussion of exemplary embodiments is not intended as a limitation on the invention. Rather, the following discussion is provided to give the reader a more detailed understanding of certain aspects and features of the invention.

As used herein, “digital light processing”, or “DLP” is a 3D printing process in which a projector is used to emit electromagnetic energy such as light to cure a resin.

As used herein, “computed axial lithography”, or “CAL” is a volumetric additive manufacturing process which relies on DLP to deliver a computed 3D exposure dose to a rotating volume of photopolymerizable resin, thereby producing a 3D object.

As used herein, a “resin” or “bioink resin” is a reagent containing one or more photopolymerizable monomers, photo-initiators, photo-absorbers, living cells, and other constituents which together contribute to the formation of bioprinted tissue.

As used herein, “DLP/CAL printing” or “DLP/CAL” is a process which incorporates DLP and CAL and two or more bioink resin tanks to form multi-layered tissue structures.

As used herein, “printing platform”, “printing surface”, “overprinting surface”, and the like are used interchangeably to indicate a substrate upon which one or more bioink resin is cured.

According to one embodiment, a temperature controlled multi-bioink resin tank system for 3D bio-overprinting through DLP/CAL includes two or more tunable temperature controlled bioink resin tanks which are used consecutively to form a multi-layered tissue which is overprinted on a surface based on instructions stored in 3D files or software and tasks executed by a controller. The multi-layer tissue is formed by overprinting on a printing surface submerged in the bioink resin tank through one or more DLP projectors which cure the resin on the surface, which projectors can be positioned from the top, side and/or beneath the bioink resin tank or any angle therebetween. The one or more DLP projectors can be stationary or can be moveable by one or more actuators. The overprinting surface can be an inflatable, soft, or solid 3D shape, or combination thereof, and can include a rubber polymer, plastic, or metal, or combination thereof. The overprinting surface can be moveable by one or more actuators capable of rotary or vertical movement, and can have an extrusion function. The extrusion function can be implemented for example by way of one or more ports or orifices disposed in communication with one or more print surface and channels, such as the orifice being disposed on the top of the print surface and the channel(s) being disposed underneath the print surface. The channels can deliver one or more bioink, such as extrudable bioinks, including gels or polymers, which are capable of being extruded, for example into a photopolymerizable gel. For example, one or more materials can be dispensed by way of a sprayer, or other device for dispensing materials without direct contact with the printed structures and/or the print surface. In one embodiment, an air-brush type sprayer can be used that can spray additional cells, drugs, nanofibers, etc. between layers or around the printed structures. Such dispensing/extrusion function, shown in FIG. 5, will be elaborated on further below. The two or more tunable temperature controlled bioink resin tanks can be used consecutively as a solution rinse step or solution absorb step during the process. The two or more tunable temperature controlled bioink resin tanks can mechanically rotate into position below the overprinting surface which is raised and lowered in one or more or each bioink resin tank to form a multi-layered tissue through DLP/CAL printing. Further, the two or more bioink resin tanks can have tunable temperature control for biologics, cells, biopolymers, synthetic polymers, therapeutics or any combination thereof. The temperature control for one or more or each bioink resin tank can be in the range of 4 to 60 degrees Celsius. The temperature controlled bioink resin tank(s) can include a condensation absorber. The two or more bioink resin tanks can include one or more bioinks with one or more natural or synthetic monomers capable of polymerization by way of electromagnetic irradiation, one or more types of living cells, one or more therapeutic agents or particles, and one or more photo-initiators or photo-absorbers. The completed bio-overprinting tissue structure can be removed from the surface by way of deflation according to some embodiments. The completed bio-overprinting tissue structure can also be used as a bioreactor with inflating or deflating mechanical forces on the bioprinted tissue according to some embodiments. The system can include a clean chamber function using positive pressure of filtered air or inert gas.

The temperature controlled multi-bioink resin tank system includes a controller and one or more DLP/CAL projector, as well as one or more actuators to control motion of a print platform or surface, the bioink resin tanks, and/or one or more DLP/CAL projector such that different bioink resins can be cured on the print surface consecutively to form a multi-layered tissue on the print surface. The controller commands the one or more DLP/CAL projector and the actuators to allow for bioprinting on the print surface.

The electromagnetic energy from the DLP/CAL projector can have a wavelength ranging from 100 nm to 2,500 nm, including from 250 nm to 450 nm, or from 300 nm to 425 nm, or from 330 nm to 420 nm, or from 350 nm to 390 nm, or from 365 nm to 405 nm, or from 330 and 460 nm, or from 370 and 440 nm, or from 405 nm to 500 nm, or from 500 nm to 800 nm, or from 700 nm to 2,500 nm.

The electromagnetic energy from the DLP/CAL projector can include UV light with wavelengths ranging from 100 nm to 400 nm, including UV-A light (320-400 nm), UV-B light (290-320 nm), and UV-C light (100-290 nm). The electromagnetic energy can include infrared light with wavelengths ranging from 780 nm to 1 mm, including IR-A (780 nm-1.4 μm), IR-B (1.4-3 μm) and IR-C, also known as far-IR (3 μm-1 mm). The electromagnetic energy can include visible light with wavelengths ranging from 400 nm to 700 nm.

The intensity of the electromagnetic energy from the DLP/CAL projector can range from 0.1-40 J/cm2 such as from 0.1-1 J/cm2, 1-5 J/cm2, 5-10 J/cm2, 10-15 J/cm2, 15-20 J/cm2, 20-25 J/cm2, 25-30 J/cm2, 30-35 J/cm2, or 35-40 J/cm2. The frequency of the electromagnetic energy used for irradiation can be varied. For example, ultraviolet light has frequencies that range from 8×1014 Hz to 3×1016 Hz. If infrared light is used, the frequency can range from 300 GHz to 450 THz.

The source of the electromagnetic energy from the DLP/CAL projector can be a laser, LED, incandescent, or fluorescent light source. Depending upon the desired wavelength, the laser can be a solid-state laser, gas laser, excimer laser, dye laser, or semiconductor laser.

The controller can perform operations and processes described or depicted herein through one or more computer processor. For example, the one or more computer processor can issue commands to the DLP/CAL projector to irradiate the overprinting surface with a specified wavelength, intensity, and location. Further, the one or more computer processor can issue commands to the actuators to control horizontal, vertical, or rotary motion of the bioink resin tanks, overprinting surface, or one or more DLP/CAL projector as well as inflation and deflation of the print surface (if such inflatable surface is implemented). The one or more computer processor commands a sequence of movements such that a different bioink from one or more or each tank can be used to photopolymerize a layer on the overprinting surface.

The one or more computer processor commands the DLP/CAL projector and actuators through instructions programmed on one or more computer files which are read by the one or more computer processors. Embodiments can include a non-transitory computer readable storage medium comprising one or more computer files (e.g. “3D files”) comprising a set of computer-executable instructions for performing one or more of the processes and operations described herein and/or depicted in the drawings, such as irradiation of the overprinting surface by the DLP/CAL projector or movement of the actuators. The 3D files can be created through Computer Aided Design (CAD) software, or equivalent 3D modeling software, which allow a user to determine the 3D structure of the overprinted tissue by creating tissue structure models. In embodiments, the files can be stored contiguously or non-contiguously on the computer-readable medium. Further, embodiments include a computer program product comprising the computer files, either in the form of the computer-readable medium comprising the computer files and, optionally, made available to a consumer through packaging, or alternatively made available to a consumer through electronic distribution. As used herein, a “computer-readable medium” includes any kind of computer memory such as floppy disks, conventional hard disks, CD-ROMS, Flash ROMS, non-volatile ROM, electrically erasable programmable read-only memory (EEPROM), and RAM.

As used herein, the terms “computer-executable instructions”, “code”, “software”, “program”, “application”, “software code”, “computer readable code”, “software module”, “module” and “software program” are used interchangeably to mean software instructions that are executable by a processor. The computer-executable instructions can be organized into routines, subroutines, procedures, objects, methods, functions, or any other organization of computer-executable instructions that is known or becomes known to a skilled artisan in light of this disclosure, where the computer-executable instructions are configured to direct a computer or other data processing device to perform one or more of the specified processes and operations described herein. The computer-executable instructions can be written in any suitable programming language, non-limiting examples of which include C, C++, C#, Objective C, Swift, Ruby/Ruby on Rails, Visual Basic, Java, Python, Perl, PHP, and JavaScript.

In other embodiments of the invention, files comprising the set of computer-executable instructions can be stored in computer-readable memory on a single computer or distributed across multiple computers. A skilled artisan will further appreciate, in light of this disclosure, how the invention can be implemented, in addition to software, using hardware or firmware. As such, as used herein, the operations of the invention can be implemented in a system comprising any combination of software, hardware, or firmware.

Embodiments of the invention include one or more computers or devices loaded with a set of the computer-executable instructions described herein. The computers or devices can be a general-purpose computer, a special-purpose computer, or other programmable data processing apparatus to produce a particular machine, such that the one or more computers or devices are instructed and configured to carry out the processes and operations described herein. The computer or device performing the specified processes and operations can comprise at least one processing element such as a central processing unit (i.e. processor) and a form of computer-readable memory which can include random-access memory (RAM) or read-only memory (ROM). The computer-executable instructions can be embedded in computer hardware or stored in the computer-readable memory such that the computer or device can be directed to perform one or more of the processes and operations depicted in the drawings and/or described herein. The computer-executable instructions can provide an operator interface for the system controller which can be a graphical user interface (GUI) which can be presented on a display of a general-purpose or special-purpose computer with a processor, computer-readable memory, and standard I/O interfaces such as a universal serial bus (USB) port and a serial port, a disk drive, a CD-ROM drive, and/or one or more user interface devices including a keyboard, keypad, mouse, control panel, touch screen display, microphone, etc. which allow a user to operate or program the controller.

The actuators controlling the movement of system components can include rotary actuators, such as for mechanical rotation of the bioink resin tanks or the overprinting surface, or linear actuators such as for mechanical raising and lowering of the overprinting surface from and to the interior of the bioink resin tanks. The actuators can be pneumatic, hydraulic, electric or mechanical actuators.

The system can include one or more temperature control units which are capable of regulating the temperature of the bioink resin tanks. In some embodiments, the temperature of the overprinting surface is independently controlled through a temperature control unit. The temperature control units can include a heater in communication with the bioink resin tanks, or a heat sink such as circulating coolant. The temperature control units are capable of regulating the temperature of the bioink resin tanks in the range of 4 to 60 degrees Celsius.

The system can include one or more tanks of pressurized air or inert gas such as nitrogen or helium for inflation of the overprinting surface or for cleaning any component of the system.

The system and its components can be powered via 60V or 120V AC current, or other voltages according to the single-phase voltage standard that is used in particular countries or regions. In general, this can be in the range of 100-127 volts or 220-240 volts.

One or more bioink resin tank can include one or more natural, semi-synthetic or synthetic monomers which, upon irradiation with electromagnetic energy for example from one or more DLP/CAL projector, can form a polymer such as any biodegradable or non-biodegradable, optionally “biocompatible polymer,” which when introduced into a living system is compatible with living tissue and/or the living system (e.g., not substantially toxic, injurious, or leading to immunological rejection). Any target polymer can be used, including any monomers, co-polymers, and block co-polymers used for preparing any such target polymer.

One or more bioink resin tank can include one or more natural monomers which, upon irradiation with electromagnetic energy for example from one or more DLP/CAL projector, form a polymer or biopolymer such as gelatin, alginate, fibrin, hyaluronic acid (or hyaluronan), agarose, decellularized extracellular matrix, silk, natural rubber, glycogens, chitin, amylopectin, cellulose, alginate, wool, amber, keratin, collagen, starch, DNA, shellac, growth factors, Matrigel®, poly (acrylic acid), polypeptide-DNA, anticoagulants (including heparin and coumarin), polysaccharide, dextran, casein, albumin, ovalbumin, fibronectin, keratin, pectin, or elastin. Particular examples of photopolymerizable bioinks that can be used include gelatin methacrylate, collagen methacrylate, and PEGDA.

One or more bioink resin tank can include one or more semi-synthetic or synthetic monomers which, upon irradiation with electromagnetic energy for example from one or more DLP/CAL projector, can form a polymer or biopolymer, for example, including polymethylmethacrylate, polyaryletherketones (PAEKs), including polyetheretherketone (PEEK) and polyaryletherketone-etherketoneketone (PEKEKK), polyurethane, poly(L-lactide), poly(D,L-lactide), poly(L-co-D,L-lactide), polyglycolide, poly(lactide-co-glycolide), poly(hydroxylbutyrate), poly(hydroxyvalerate), tyrosine-derived polycarbonate, polyanhydride, polyorthoester, polyphosphazene, poly(dioxanone), poly(ε-caprolactone), and polyglyconate, such as described in U.S. Pat. No. 9,662,821, which is hereby incorporated by reference herein in its entirety. Other similar polymers known in the art may be used and various combinations of polymers may be included in the composition to adjust the properties of the composition as desired.

Polymers for tissue engineering and/or drug delivery can also be used. Electrically conducting polymers such as polyaniline, polypyrrole, polythiophene, and their derivatives (mainly aniline oligomer and poly(3,4-ethylenedioxythiophene)), or combinations thereof and combinations with other polymers can be used.

Polymers can include α-hydroxycarboxylic acids and copolymers thereof, including PGA, PLA and copolymers thereof, polyethylene oxide/polyethylene terephthalates, copolymers of lactic or glycolic acid, poly(alkylene glycols) of various molecular weights, biodegradable and biocompatible polycaprolactones, polyhydroxybutyrates and copolymers of polyesters, polycarbonates, polyanhydrides and poly(ortho esters), bisphenol-A based polyphosphoesters, including poly(bisphenol-A phenylphosphate), poly(bisphenol-A ethylphosphate), poly(bisphenol-A ethylphosphonate), poly(bisphenol-A phenylphosphonate), poly[bis(2-ethoxy)hydrophosphonic terephthalate], and copolymers of bisphenol-A based poly(phosphoesters), copolymers of polyethylene oxide/polyethylene terephthalate, polymers of tyrosine-derived diphenol compounds, polycarbonates, polyiminocarbonates, polyarylates, polyurethanes, polyethers, and the like.

Some bioinks which can be used with an extrusion function and/or for curing include hydrogels based on RGD-conjugated polysaccharide bioinks with or without fibrin described in U.S. Published Application No. 2019/0160203, modified cellulose nanofibrils with extracellular matrix components described in U.S. Published Application No. 2019/0209738, and nanocellulose bioinks described in U.S. Published Application No. 2017/0368225, each of which is hereby incorporated by reference herein.

Bioinks that can be used include a double-network bioink comprising a non-crosslinkable thickener and a cross-linkable polyethylene glycol-based network that interpenetrate each other. Such bioinks can comprise (a) a biocompatible or non-biocompatible thickener; (b) a polyethylene glycol based crosslinkable network; a photoinitiator; and/or optionally, additives to impart different characteristics; wherein the thickener and polyethylene glycol based crosslinkable network form a structure comprising two interpenetrating networks. In embodiments, the bioink can comprise a thickener chosen from one or more of: polyethylene oxide; polypropylene oxide; nanofibrillar cellulose; nanocrystalline cellulose; gelatin; collagen; glucomannan; alginate; k-carrageenan; bentonite clay; and/or xanthan gum. In embodiments, the cross-linkable polyethylene glycol can comprise one or more reactive groups, including: acrylate; thiol; maleimide; and/or biotin. In embodiments, the polyethylene glycol crosslinker can exhibit structures or blends that are: linear; branched; 4-arm; 8-arm; and/or hyperbranched. In embodiments, the additives can comprise monoacrylate PEG with functionalization of the following: fluorescent groups such as fluorescein, rhodamine, or dansyl; sulfonate groups; amine groups; phosphate groups; lipid groups; and/or CNT binding. In embodiments, the photoinitiator can comprise one or more of Irgacure 2959; LAP; Eosin-Y; and/or Avidin. An exemplary double-network bioink composition according to embodiments of the invention can include (a) one or more thickener present in an amount ranging from 0.1% to 20% w/v of the composition, (b) one or more PEGDA crosslinker present in an amount ranging from 0.1% to 10% w/v of the composition, (c) one or more photoinitiator present in an amount ranging from 0.05% to 1% w/v of the composition, and (d) optionally one or more supplementary proteins present in an amount ranging from 0.01% to 10% w/v of the composition, with the remainder comprising water and salts. By varying the type and/or amount of any one or more of the thickener, PEGDA crosslinker and/or photoinitiator, the mechanical stiffness, the diffusivity, the elasticity, binding capacity, etc. of the construct can be varied. The elasticity and stiffness of the cross-linked bioink can be controlled by changing the blending ratio between the thickener, cross-linking polymer, and the concentration of the photoinitiator.

Bioinks also include biogum-hydrogel bioinks (for example comprising a xanthan gum-based bioink, and one or more biomaterials derived from mammalian, plant, microbial or synthetically derived hydrogels) and/or botanical gum-hydrogel bioinks (for example comprising a botanical gum-based bioink, and one or more biomaterials derived from mammalian, plant, microbial or synthetically derived hydrogels). In embodiments, the bioink compositions can be provided with cells, preferably human cells. In embodiments, the bioink compositions can comprise a ratio of biogum versus biomaterial, or a ratio of botanical gum versus biomaterial, in the interval from 5:95 to 95:5. In embodiments, the biomaterial hydrogel can be at least one of the following constituents for cross-linking purposes and/or to contribute to rheological properties of the bioink, such as hydrocolloids or thickening and gelling agents: collagen type I, collagen and its derivatives, gelatin methacryloyl, gelatin and its derivatives, fibrinogen, thrombin, elastin, alginates, agarose and its derivatives, glycosaminoglycans such as hyaluronic acid and its derivatives, chitosan, low and high methoxy pectin, gellan gum, diutan gum, glucomannan gum, carrageenans, nanofibrillated cellulose, microfibrillated cellulose, crystalline nanocellulose, carboxymethyl cellulose, methyl and hydroxypropylmethyl cellulose, bacterial nanocellulose, or a combination of these constituents. In embodiments, the bioink compositions can configured such that the xanthan-gum bioink has a concentration in the interval from 0.5 to 10% (w/v) or the botanical gums bioink has a concentration in the interval from 0.5 to 50% (w/v). In embodiments, the bioink comprises additional biopolymers for cross-linking purposes and/or to contribute to rheological properties of the bioink, such as hydrocolloids or thickening and gelling agents, for example, alginates, hyaluronic acid and its derivatives, agarose and its derivatives, chitosan, fibrin, gellan gum, silk nanofibrillated cellulose, microfibrillated cellulose, crystalline nanocellulose, bacterial nanocellulose, carrageenans, elastin, collagen and its derivatives as well as gelatin and its derivatives. In embodiments, the concentration of cells, such as human or animal cells, is in the interval from 0.1 million/ml to 150 million/ml. In embodiments, the cells are of human or porcine origin, preferably human origin. In embodiments, the composition is provided in physiological conditions, such as having a pH-value for the composition in the interval from 5-8, preferably 7 and/or an osmolarity of the composition is in the interval from 275 to 300 mOsm/kg, preferably 295 mOsm/kg.

Exemplary bioinks which can be used include, but are not limited to, the following available from CELLINK (Gothenburg, Sweden):

TABLE I Exemplary Bioinks Bioink Components CELLINK A Alginate CELLINK A-RGD Alginate coupled with L-arginine-Glycine- L-aspartic Acid peptide CELLINK Bioink Alginate & Nanofibrillar cellulose CELLINK BONE CELLINK & Tricalcium phosphate CELLINK FIBRIN CELLINK & Fibrinogen CELLINK LAMININK+ CELLINK & Laminin blend CELLINK LAMININK 111 CELLINK & Laminin a1b1y1 CELLINK LAMININK 121 CELLINK & Laminin a1b2y1 CELLINK LAMININK 411 CELLINK & Laminin a4b1y1 CELLINK LAMININK 521 CELLINK & Laminin a5b2y1 CELLINK RGD CELLINK & Alginate coupled with L- arginine-Glycine-L-aspartic Acid peptide CELLINK SKIN CELLINK & Fibrinogen Coll 1 Collagen type 1 ColMA Collagen methacrylate Bio Conductink Gelatin methacrylate & Carbon nanotubes GelMA Gelatin methacrylate GelMA A Gelatin methacrylate & Alginate GelMA C Gelatin methacrylate, Nanofibrillar cellulose & Alginate GelMA HA Gelatin methacrylate & Methacrylated hyaluronic acid GelMA HIGH High-concentration gelatin methacrylate. GelXA Gelatin methacrylate, Xanthan Gum & Alginate GelXA BONE Gelatin methacrylate, Xanthan Gum, Alginate, Tricalcium phosphate & Hydroxyapatite GelXA FIBRIN Gelatin methacrylate, Xanthan Gum, Alginate & Fibrinogen GelXA LAMININK+ Gelatin methacrylate, Xanthan Gum, Alginate & Laminin blend GelXA LAMININK 111 Gelatin methacrylate, Xanthan Gum, Alginate & Laminin a1b1y1 GelXA LAMININK 121 Gelatin methacrylate, Xanthan Gum, Alginate & Laminin a1b2y1 GelXA LAMININK 411 Gelatin methacrylate, Xanthan Gum, Alginate & Laminin a4b1y1 GelXA LAMININK 521 Gelatin methacrylate, Xanthan Gum, Alginate & Laminin a5b2y1 GelXA SKIN Gelatin methacrylate, Xanthan Gum, Alginate & Fibrinogen GelXG Gelatin methacrylate & Xanthan Gum CELLINK PCL Polycaprolactone CELLINK PLURONICS Poloxamer CELLINK START Polypropylene gel CELLINK START X CELLINK START. Photo-crosslinkable polymer. CELLINK SUPPORT Nanofibrillar cellulose CELLINK XPLORE Cellulose nanocrystals. Alginate. Coloring.

The monomers (natural or synthetic) can be provided in the bioink resins at an appropriate concentration, such as 0.1% to 20% w/v or higher, such that they react to polymerize upon exposure of the bioink resin to light.

One or more or each bioink resin tank can include one or more photo-initiators within the bioink(s) which can include, for example, free radical photoinitiators, cationic photoinitiators, and anionic photoinitiators. The photoinitiator forms a free radical, cation, or anion which subsequently reacts and catalyzes a polymerization reaction among the monomers within the bioink. Examples of photoinitiators include, but are not limited to benzophenone, benzoin-ether, 2-(dimethylamino)ethanol (DMAE), hydroxyacetophenones, 2-hydroxy-2-methyl-1-phenylpropan-1-one and, hydroxyl-phenyl-ketone, Irgacure® 2959, 2-hydroxy-4′-(2-hydroxyethoxy)-2-methylpropiophenone, (2-hydroxy-1-[4-(2-hydroxyethoxy) phenyl]-2-methyl-1-propanone; lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP); (2,2′-azobis[2-methyl-N-(2-hydroxyethyl)propionamide]; 2-isocyanotoethyl methacrylate; benzoyl benzylamine; camphorquinone; thiol-norbornene (thiol-ene); riboflavin; lucirin-TPO; Rose Bengal/furfuryl; ethyl eosin; methacrylic anhydride; 2,2-dimethoxy-2-phenyl acetophenone; and Eosin Y.

One or more or each bioink resin tank can include one or more photo-absorbers within the bioink(s) which can include, for example, compounds such as tartrazine, curcumin, anthocyanin, quinoline yellow, as well as gold nanoparticles. The photo-absorber(s) is included in the bioink(s) to limit or control the polymerization reaction.

The photo-initiators and/or photo-absorbers can be free within the bioink solution or can be bound to the one or more monomers, such as through covalent bonding. The photo-initiators and photo-absorbers can be added to the bioink resins at concentration ratios which produce a desired rate of polymerization.

One or more or each bioink resin tank can include one or more types of living cells within the bioink(s) which can be or include, for example, cells from tissues such as liver, kidney, heart, lung, gastrointestinal, muscle, skin, bone, cartilage, vascularized tissues, blood vessels, ducts, ear, nose, esophagus, trachea, and eye. These can include endothelial cells, skin cells such as keratinocytes, melanocytes, Langerhans' cells, and Merkel cells, connective tissue cells such as fibroblasts, mast cells, plasma cells, macrophages, adipocytes, and leukocytes, bone tissue cells such as osteoblasts, osteoclasts, osteocytes, and osteoprogenitor (or osteogenic) cells, cartilage cells such as chondrocytes and chondroblasts, muscle cells such as smooth muscle cells, skeletal muscle cells, cardiac muscle cells, any cells having muscle fibers such as type I (slow twitch), type IIa and type IIb (fast twitch), nerve cells such as multipolar neurons, bipolar neurons, unipolar neurons, sensory neurons, interneurons, motor neurons, neurons of the brain (e.g. Golgi cells, Purkinje cells, pyramidal cells), glial cells such as oligodendrocytes, astrocytes, ependymal cells, Schwann cells, microglia, and satellite cells, liver cells such as hepatocytes, biliary epithelial cells (cholangiocytes), stellate cells, Kupffer cells, and liver sinusoidal endothelial cells, kidney cells such as glomerulus parietal cells, glomerulus podocytes, proximal tubule brush border cells, Loop of Henle thin segment cells, thick ascending limb cells, kidney distal tubule cells, collecting duct principal cells, collecting duct intercalated cells, and interstitial kidney cells, pancreatic cells such as islets cells, alpha cells, beta cells, delta cells, PP cells, endocrine gland cells such as pancreatic cells, hypothalamus cells, pituitary cells, thyroid cells, parathyroid cells, adrenal cells, pineal body cells, and ovarian cells and testicular cells, exocrine gland cells such as sweat gland cells, salivary gland cells, mammary gland cells, ceruminous gland cells, lacrimal gland cells, sebaceous gland cells, and mucous gland cells, epithelial cells such as squamous cells, cuboidal cells, and columnar cells arranged in architectures such as simple, stratified, and pseudostratified, primary cells, immortalized cells, stem cells such mesenchymal stem cells, hematopoietic stem cells, embryonic stem cells, pluripotent stem cells, multipotent stem cells, totipotent stem cells, as well as xenogeneic cells, autologous cells, and allogenic cells, and any combination thereof. The cells can first be grown in suspension or on cell or tissue culture plates as is known in the art, then harvested and added to the bioink resin prior to printing.

One or more or each bioink resin tank can include one or more therapeutic agents or particles within the bioink(s), which can have a molecular weight in the range of less than 900 Daltons for small molecules to up to 1000 kDa for biologics. The concentration of the small molecule or biologic therapeutics can be chosen according to published studies which reveal effective concentrations, such as pre-clinical or clinical pharmacology data. The small molecules can be any small molecule used in the treatment of any disease, including diseases of the eye, ear, nose, throat, mouth, lung, heart, liver kidney, spleen, pancreas, gastrointestinal system, circulatory system, reproductive system, central nervous system, immune system, musculoskeletal system, and skin. The small molecules can be found or identified in publicly available drug references (see Remington, The Science and Practice of Pharmacy. Easton, Pa.: Mack Pub. Co., 1995; Goodman, L. S., Brunton, L. L., Chabner, B., & Knollmann, B. C. (2011). Goodman & Gilman's pharmacological basis of therapeutics. New York: McGraw-Hill; O'Neil, M. J. (2006). The Merck index: An encyclopedia of chemicals, drugs, and biologicals. Whitehouse Station, N.J.: Merck; United States Pharmacopeia and National Formulary). Rockville, Md.: United States Pharmacopeial Convention; 2016; and Center for Drug Evaluation and Research (U.S.), & Center for Drug Evaluation and Research (U.S.). (1985). Orange book: Approved drug products with therapeutic equivalence evaluations. Rockville, Md.: U.S. Dept. of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Science, Office of Generic Drugs); these and any edition available at the time of this disclosure are hereby incorporated by reference herein in their entireties.

The one or more therapeutic agents or particles within the bioink(s), can be or include, for example, a biologic such as any bioactive protein or peptide, such as growth factors which include, but are not limited to native or modified PDGF, FGF-2, EGF, epiregulin, TGF-alpha, keratinocyte growth factor, keratinocyte growth factor-2 (KGF-2), granulocyte-macrophage colony-stimulating factor (GM-CSF), TGF-beta, insulin-like growth factor I (IGF-I), insulin-like growth factor II (IGF-II), nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and human growth hormone (HGH), cytokines which include but are not limited to IL-1 (IL-1α and -β), IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-16, IL-18, IFN-α, IFN-β, IFN-γ, TNF-α, and TNF-β, and chemokines which include, but are not limited to, any of four subclasses of chemokines (CCL, CXCL, CX 3 CL, and XCL), including CCL2, CCL3 and CCLS, CXCL1, CXCL2 and CXCL8. The biologics can include any of the above, as well as, for example, an antibody, antibody fragment, antibody-drug conjugate, peptide, peptide-drug conjugate, protein, polypeptide, fusion protein, multivalent binding protein, blood and blood product, nucleic acid, nucleotide, oligonucleotide, antisense oligonucleotide, short interfering RNA (siRNA), micro-interfering RNA (miRNA); small, temporal RNA (stRNA); short, hairpin RNA (shRNA), aptamer, ribozyme, viral vector (e.g. adenovirus, adeno-associated virus (AAV), retrovirus, and lentivirus), and non-viral vector (e.g. plasmid). Non-limiting examples of biologics products include adalimumab (Humira), rituximab (Rituxan), etanercept (Enbrel), trastuzumab (Herceptin), bevacizumab (Avastin), infliximab (Remicade), insulin glargine (Lantus), pegfilgrastim (Neulasta), interferon beta-1a (Avonex), ranibizumab (Lucentis), and epoetin alfa (Epogen). Additional biologics can be found in the U.S. Food and Drug Administration's Purple Book, which is publicly available on the U.S. FDA website and hereby incorporated by reference.

The temperature controlled multi-bioink resin tank system is capable of building complex tissue structures, such as branched or unbranched vascular structures, branched or unbranched pulmonary structures, branched or unbranched duct-like structures, bladder-shaped structures, and layered structures.

The temperature controlled multi-bioink resin tank system is capable of use in combination with other 3D bioprinting technologies, including extrusion, inkjet, acoustic/ultrasound, laser, and stereolithography technologies.

The temperature controlled multi-bioink resin tank system can be used in a clean room to maintain sterile conditions or can be incorporated into clean chamber technology such as described in U.S. Patent Application Publication No. US 20180326665A1, incorporated herein by reference in its entirety.

FIG. 1 is a schematic diagram showing a print surface and the progression of different bioinks photocured on the surface with a side projector according to an embodiment. The print surface (1) is vertically lowered into bioink resin tank one and the first section of the sphere is polymerized through DLP/CAL onto the print surface (2). Upon completion, the print surface is raised and resin tank two moves into position with the print surface. The print surface is lowered into bioink resin tank two and polymerized through DLP/CAL onto the previous bioprinted layer (3). Upon completion, the print surface is raised and resin tank three moves into position with the print surface. The print surface is lowered into bioink resin tank three and polymerized through DLP/CAL on the previous bioprinted layer (4). Upon completion, a three-cell type multi-layered bioprinted spherical structure is created on the print surface.

FIG. 2A is a schematic diagram showing a top view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting which includes a controller (1) that commands and DLP/CAL projector (2), vertical motion of a print surface (3), and horizontal rotation of a temperature-controlled platform or stage (4) with multiple temperature-controlled bioink resin tanks (5,6,7). One or more or each bioink resin tank can move into position with the DLP projector and under the print surface consecutively to form a multi-layered tissue on the inflated surface. Movement of one or more components of the system can be relative to one or more other component(s) of the system. For example, one or more light source can move and/or one or more print surface can move, and/or one or more bioink tank can move, and/or one or more platform on which one or more bioink tank is positioned can move.

FIG. 2B is a schematic diagram showing a top view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting which includes a controller (1) which commands multiple functions of the system. The controller commands the side projector (2), the horizontal rotation of the temperature-controlled platform or stage (3), the vertical and circular motion of the print surface (4), and the multiple temperature-controlled bioink resin tanks with circular motion (5). One or more or each bioink resin tank moves into position with the projector and under the print surface consecutively to form a multi-layered tissue on the surface.

FIG. 3A is a schematic diagram showing a side view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting with one projector. The system includes a controller (1) that commands a DLP projector (2), the horizontal rotation of a temperature-controlled platform or stage (3) with multiple temperature-controlled bioink resin tanks (4), and the print surface (5,6,7) with vertical and circular rotational motion and an inflatable feature (7). The controller and the system allow for multi-layer multi-material bioprinting on the inflatable print surface (8).

FIG. 3B is a schematic diagram showing a side view of an embodiment of a multi-bioink resin tank system for 3D bio-overprinting with two projectors. The controller (1) commands multiple functions of the system. The controller commands the side projector (2), the horizontal rotation of the temperature-controlled platform or stage (3), the bottom projector (4), the vertical and circular motion of the print surface (5,6), and the temperature-controlled bioink resin tanks with circular motion (7). The system allows for photo-polymerization of a bioinks on the print surface (8).

FIG. 4 is a schematic diagram showing multi-projector printing and the progression of different bioinks photocured on the surface. The print surface (1) is vertically lowered into bioink resin tank one and the first section is polymerized with the bottom projector onto the print surface (2). Upon completion, the print surface is raised and resin tank two moves into position with the print surface. The print surface is lowered into bioink resin tank two and polymerized with the side projector onto the previous bioprinted layer (3).

FIG. 5 is a schematic diagram of a print surface with an extrusion function. The print surface (1) is vertically lowered into a bioink resin tank and a sphere is polymerized through DLP/CAL onto the print surface (2). Upon completion, the print surface extrusion function is initiated and a bioink is extruded out of the print surface and into the polymerized sphere at specific points. In embodiments, one or more materials can be dispensed by way of a sprayer, or other device for dispensing materials without direct contact with the printed structures and/or the print surface. For example, an air-brush type sprayer can be used that can spray additional cells, drugs, nanofibers, etc. between layers or around the printed structures.

Example 1

A 3D file is created containing a model of a hollow Y-branched vascular tube (50×50×10 mm) with three layered sections. Each of the three sections are 2 mm in width and represent a different bioink that will be consecutively photopolymerized on the print surface that mimics the model. The mimicking surface is a rubber polymer that is inflated and vertically lowered into the temperature-controlled multi-bioink resin tanks and rotates in a circular motion. The bioprinting software controller recognizes each model section and makes adjustments to each tank position, the print surface, and the projector based on the model coordinates and bioprinting progress. Three individual bioink resin tanks are each set and maintained at 4 degrees C. during the bioprinting. Bioink resin tank one is loaded with a photopolymerizable collagen solution, a photo-initiator, a photo-absorber, and human smooth muscle cells. Bioink resin tank two is loaded with a photopolymerizable collagen solution, a photo-initiator, a photo-absorber, and human fibroblast cells. Bioink resin tank three is loaded with a photopolymerizable collagen solution, a photo-initiator, a photo-absorber, and human endothelial cells. The print surface is inflated and is vertically lowered into bioink resin tank one and polymerized through DLP/CAL onto the print surface until 2 mm thickness is achieved. Upon completion, the print surface is raised and resin tank two moves into position with the print surface. The print surface is lowered into bioink resin tank two and polymerized through DLP/CAL onto the previous bioprinted layer until 2 mm thickness is achieved. Upon completion, the print surface is raised and resin tank three moves into position with the print surface. The print surface is lowered into bioink resin tank three and polymerized through DLP/CAL on the previous bioprinted layer until 2 mm thickness is achieved. Upon completion, a three-cell type multi-layered bioprinted construct is engineered on the print surface. After printing is complete the print surface is deflated and the bioprinted multi-layered structure is rinsed and incubated with cell culture media.

Example 2

Same as Example 1, but after printing is complete the print surface with the bioprinted multi-layered structure is rinsed and placed into cell culture media. The inflate and deflate function of the system is used as a mechanical force bioreactor on the bioprinted multi-layered structure.

Example 3

A 3D file is created containing a model of a sphere (50×50×10 mm) with three layered sections. Each of the three sections represents a different bioink that will be consecutively photopolymerized on the print surface. The print surface and bioink tank both rotate in a circular motion during the photo-curing which is achieved by the side projector. The bioprinting software controller recognizes each model section and makes adjustments to the print surface, the bioink tank positions, and the side projector based on the model coordinates and bioprinting progress.

The three individual bioink resin tanks are each set and maintained at 4 degrees C. during the bioprinting. Bioink resin tank one is loaded with a photopolymerizable collagen solution, a photo-initiator, a photo-absorber, and human smooth muscle cells. Bioink resin tank two is loaded with a photopolymerizable collagen solution, a photo-initiator, a photo-absorber, and human fibroblast cells. Bioink resin tank three is loaded with a photopolymerizable collagen solution, a photo-initiator, a photo-absorber, and human endothelial cells. The print surface is vertically lowered into bioink resin tank one and the first section of the sphere is polymerized through DLP/CAL onto the print surface. Upon completion, the print surface is raised and resin tank two moves into position with the print surface. The print surface is lowered into bioink resin tank two and polymerized through DLP/CAL onto the previous bioprinted layer. Upon completion, the print surface is raised and resin tank three moves into position with the print surface. The print surface is lowered into bioink resin tank three and polymerized through DLP/CAL on the previous bioprinted layer. Upon completion, a three-cell type multi-layered bioprinted spherical structure is engineered on the print surface (FIG. 1). After printing is complete the bioprinted multi-layered sphere is rinsed and incubated with cell culture media.

The present invention has been described with reference to particular embodiments having various features. In light of the disclosure provided above, it will be apparent to those skilled in the art that various modifications and variations can be made in the practice of the present invention without departing from the scope or spirit of the invention. One skilled in the art will recognize that the disclosed features may be used singularly, in any combination, or omitted based on the requirements and specifications of a given application or design. When an embodiment refers to “comprising” certain features, it is to be understood that the embodiments can alternatively “consist of” or “consist essentially of” any one or more of the features. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention.

It is noted in particular that where a range of values is provided in this specification, each value between the upper and lower limits of that range is also specifically disclosed. The upper and lower limits of these smaller ranges may independently be included or excluded in the range as well. The singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. It is intended that the specification and examples be considered as exemplary in nature and that variations that do not depart from the essence of the invention fall within the scope of the invention. Further, all of the references cited in this disclosure are each individually incorporated by reference herein in their entireties and as such are intended to provide an efficient way of supplementing the enabling disclosure of this invention as well as provide background detailing the level of ordinary skill in the art.

Claims

1. A system for three-dimensional (3D) bioprinting, comprising:

a printing platform or surface;
one or more container;
one or more source of ultraviolet, visible, or infrared light;
one or more actuator capable of horizontal, vertical, and/or rotary movement of the printing platform or surface, the one or more container and/or the one or more source of ultraviolet, visible, or infrared light; and
a controller capable of sending commands to the one or more source of ultraviolet, visible, or infrared light and the one or more actuator in a manner sufficient to build a 3D structure.

2. The system of claim 1, wherein the system is configured to perform the steps of:

a) positioning a first container below the printing platform or surface;
b) lowering the printing platform or surface into the first container;
c) exposing the printing platform, surface or portion thereof to the source of ultraviolet, visible, or infrared light in the presence of a first bioink in a manner to form one or more first layer of a 3D structure;
d) raising the printing platform or surface from the first container;
e) positioning a second container below the printing platform or surface;
f) lowering the printing platform or surface into the second container;
g) rinsing the 3D structure or exposing the printing platform, surface or portion thereof to the source of ultraviolet, visible, or infrared light in the presence of a second bioink in a manner to form one or more subsequent layer of a 3D structure;
h) raising the printing platform or surface from the second container; and
i) optionally repeating steps e) through h) with one or more additional substances and optionally one or more additional containers to further rinse or build upon the 3D structure.

3. The system of claim 2, wherein the one or more actuator is configured to:

raise and lower the printing platform or surface by way of vertical and/or rotary motion;
position the one or more container below the printing platform or surface by way of horizontal and/or rotary motion.

4. The system of claim 2, wherein the one or more source of ultraviolet, visible, or infrared light is one or more digital light processing (DLP) projector positioned or capable of being positioned above, below, and/or at one or more sides of the printing platform or surface and/or one or more of the containers.

5. The system of claim 1, wherein the printing platform or surface is capable of inflation and/or deflation.

6. The system of claim 1, wherein the 3D structure comprises a bio-overprinted surface capable of acting as a bioreactor for tissue stimulation.

7. The system of claim 2, further comprising:

one or more temperature control unit in operable communication with: the printing platform or surface; one or more of the containers; and/or a platform or stage on which one or more of the containers is disposed.

8. The system of claim 2, further comprising one or more device configured for extruding or spraying one or more materials, cells, drugs, nanofibers and/or therapeutic agents between and/or on one or more of the first or subsequent layers of the 3D structure.

9. The system of claim 1, wherein the one or more actuator is a pneumatic, hydraulic, electric, and/or mechanical actuator.

10. The system of claim 1, wherein the controller comprises one or more processor and a non-transitory computer readable storage medium comprising one or more 3D files having instructions capable of being read by the processor.

11. The system of claim 1, further comprising a tank of compressed air or inert gas or air or a gas compression mechanism.

12. A system for bioprinting, comprising:

a printing platform or surface;
first and second containers;
one or more source of electromagnetic radiation;
one or more actuator capable of horizontal, vertical, and/or rotary movement of the printing platform or surface, the first and/or second containers, and/or the one or more source of electromagnetic radiation; and
a controller capable of sending commands to the one or more source of electromagnetic radiation and the one or more actuator in a manner sufficient to build a structure.

13. The system of claim 12, wherein the system is configured to perform the steps of:

a) exposing the printing platform, surface or portion thereof to the source of electromagnetic radiation in the presence of a first bioink in a manner to form one or more first layer of a structure;
b) rinsing the structure or exposing the printing platform, surface or portion thereof to the source electromagnetic radiation in the presence of a second bioink in a manner to form one or more subsequent layer of a structure; and
c) optionally repeating steps a) and b) with one or more additional substances and optionally one or more additional containers to further rinse or build upon the structure.

14. The system of claim 12, wherein the one or more source of electromagnetic radiation is one or more light source.

15. The system of claim 12, wherein the one or more source of electromagnetic radiation is one or more digital light processing (DLP) projector.

16. The system of claim 12, wherein the printing platform or surface is capable of inflation and/or deflation.

17. The system of claim 15, wherein the one or more digital light processing (DLP) projector is positioned or capable of being positioned above, below, and/or at one or more sides of the printing platform or surface and/or one or more of the containers.

18. The system of claim 12, further comprising one or more device configured for extruding or spraying one or more materials, cells, drugs, nanofibers and/or therapeutic agents between and/or on one or more layers of the structure.

19. The system of claim 12, wherein the controller comprises one or more processor and a non-transitory computer readable storage medium comprising one or more 3D files having instructions capable of being read by the processor.

20. The system of claim 12, further comprising a tank of compressed air or inert gas or air or a gas compression mechanism.

Patent History
Publication number: 20220105676
Type: Application
Filed: Dec 17, 2021
Publication Date: Apr 7, 2022
Inventors: Christen J. Boyer (Germantown, MD), Hector Martinez (Gothenburg), Erik Gatenholm (Gothenburg)
Application Number: 17/554,789
Classifications
International Classification: B29C 64/129 (20060101); B29C 64/188 (20060101); B29C 64/241 (20060101); B29C 64/236 (20060101); B29C 64/232 (20060101); B29C 64/264 (20060101); B29C 64/205 (20060101); B29C 64/255 (20060101); B29C 64/245 (20060101);