EP4182437A1 - Systèmes microfluidiques pour bioréacteurs multiples et leurs applications - Google Patents

Systèmes microfluidiques pour bioréacteurs multiples et leurs applications

Info

Publication number
EP4182437A1
EP4182437A1 EP21841908.3A EP21841908A EP4182437A1 EP 4182437 A1 EP4182437 A1 EP 4182437A1 EP 21841908 A EP21841908 A EP 21841908A EP 4182437 A1 EP4182437 A1 EP 4182437A1
Authority
EP
European Patent Office
Prior art keywords
fluidic
channel
bus
port
actuator
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21841908.3A
Other languages
German (de)
English (en)
Inventor
David K. Schaffer
Ronald S. Reiserer
Michael D. GEUY
John P. Wikswo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vanderbilt University
Original Assignee
Vanderbilt University
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 Vanderbilt University filed Critical Vanderbilt University
Publication of EP4182437A1 publication Critical patent/EP4182437A1/fr
Pending legal-status Critical Current

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • B01L3/502738Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip characterised by integrated valves
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/44Multiple separable units; Modules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/58Reaction vessels connected in series or in parallel
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M29/00Means for introduction, extraction or recirculation of materials, e.g. pumps
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/02Adapting objects or devices to another
    • B01L2200/026Fluid interfacing between devices or objects, e.g. connectors, inlet details
    • B01L2200/027Fluid interfacing between devices or objects, e.g. connectors, inlet details for microfluidic devices
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/06Fluid handling related problems
    • B01L2200/0621Control of the sequence of chambers filled or emptied
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0803Disc shape
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure
    • B01L2300/0887Laminated structure
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/04Moving fluids with specific forces or mechanical means
    • B01L2400/0475Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure
    • B01L2400/0487Moving fluids with specific forces or mechanical means specific mechanical means and fluid pressure fluid pressure, pneumatics
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2400/00Moving or stopping fluids
    • B01L2400/06Valves, specific forms thereof
    • B01L2400/0633Valves, specific forms thereof with moving parts
    • B01L2400/0644Valves, specific forms thereof with moving parts rotary valves

Definitions

  • the invention relates generally to fluidic systems, and more particularly to microfluidic systems including valves for multiple bioreactors, and applications of the same.
  • organs-on-chips are designed to study the physiology of a single organ, and use either height differences in reservoir fluid levels, syringe pumps, on-chip or off- chip peristaltic pumps, or pressurized reservoirs to cause culture media to flow through single- or dual-chamber bioreactors.
  • Many chips have been single-pass, perfused by the pressure from liquid in a pipette tip or a syringe body connected to the chip directly or by a tube.
  • Experiments by others involving recirculation of single-organ or coupled-organ chips typically use rocking gravity perfusion or on-chip pumps.
  • the fluidic device includes a fluidic chip having a fluidic network comprising a plurality of fluidic channels in fluidic communication with a plurality of input ports, at least one output port, and at least one sensing port; and an actuator configured to engage with the fluidic network to control each fluidic channel to switch between an open state in which fluidic flow through said fluidic channel is permitted and a closed state in which no fluidic flow through said fluidic channel is permitted, so as to selectively collect fluid from multiple inputs via the plurality of input ports, and direct either all of the multiple inputs to the at least one output port, or all but a single selected input to the at least one output port and the single selected input to the at least one sensing port to which an analytical instrument is operably connected.
  • the plurality of input ports is operably coupled with a plurality of fluidic modules, wherein in operation, the plurality of fluidic modules is individually perfused, and all outputs of the plurality of fluidic modules are directed to the at least one output port, or an output of any one of the plurality of fluidic modules is directed to the at least one sensing port, while outputs of all other fluidic modules are directed to the at least one output port.
  • the plurality of fluidic modules comprises bioreactors, wells, organs- on-chips, chemostats, or a combination of them.
  • the fluidic chip has a body in which the fluidic network is formed, and a fluidic chip registration means formed on the body for aligning the fluidic chip with a support structure.
  • the fluidic chip registration means is configured such that multiple fluidic chip orientations are allowed while maintaining automatic and precise mechanical alignment to the support structure.
  • the fluidic chip is configured such that one or more plug-in accessories are addable in or removable from the fluidic chip.
  • the fluidic chip is formed of an elastic material such that compression of the actuator on the body causes at least one of the channels to be occluded.
  • the plurality of fluidic channels comprises a first fluidic bus, a second fluidic bus, and a plurality of intermediate channels, each intermediate channel being in fluidic communication with at least one of the plurality of input ports and connected to the first fluidic bus and/or the second fluidic bus.
  • the actuator comprises a plurality of actuating elements disposed on the body of the fluidic chip with each actuating element at a location that is over an intermediate channel and is located between a respective port and one of the first fluidic bus and the second fluidic bus to which said intermediate channel is connected, such that compression of said actuating element on the body causes fluidic flow between said respective port and said one of the first fluidic bus and the second fluidic bus through said intermediate channel to be occluded.
  • the locations of the plurality of actuating elements on the body of the fluidic chip comprise first locations and second locations, wherein the first locations comprise each actuating element location over a respective intermediate channel between a respective port and the first fluidic bus, and the second locations comprise each actuating element location over a respective intermediate channel between a respective port and the second fluidic bus.
  • the actuator further comprises an actuator head for selectively compressing or relaxing each of the plurality of actuating elements.
  • the actuator head comprises an outer actuator head having an outer groove corresponding to one of the first locations of the plurality of actuating elements on the body, wherein the outer groove, when aligned with the corresponding one of the first locations, relieves the corresponding actuating element so that the corresponding port is connected to the first fluidic bus; and an inner actuator head sleeved in the outer actuator head, having an inner groove corresponding to one of the second locations of the plurality of actuating elements on the body, wherein the inner groove, when aligned with the corresponding one of the second locations, relieves the corresponding actuating element so that the corresponding port is connected to the second fluidic bus, wherein one of the outer actuator head and the inner actuator head is a driving actuator head driven by a motor, and the other of the outer actuator head and the inner actuator head is a driven actuator head driven by said driving actuator head.
  • each of the outer actuator head and the inner actuator head has a circular-segment pocket with a near-360° sweep, wherein the actuator head further comprises a single limiting element, whose motion is constrained by the pockets, allowing the driving actuator head and the driven actuator head to rotate or remain stationary independently until the limiting element contacts opposing ends of both pockets, at which point both actuator heads rotate as one, wherein when the direction of the motor is then reversed, the motion of each of the actuator heads becomes independent again.
  • the actuator head comprises a first relief pocket having two ends and a gap defined therebetween, and a second relief aligned with the gap along an intermediate channel, wherein the first relief pocket and the gap are corresponding to the first locations of the plurality of actuating elements on the body of the fluidic chip, and the second relief is corresponding to one of the second locations of the plurality of actuating elements on the body of the fluidic chip, such that in operation, one of the actuating elements on the first locations is pressed by the gap and the others of the actuating elements on the first locations are relaxed by the first pocket, and one of the actuating elements on the second locations is relaxed by the second relief and the others of the actuating elements on the second locations are pressed by the surface of the actuator head, thereby directing the single selected input from the input port connected to said intermediate channel with which the gap and the second relief are aligned to the at least one sensing port through the second fluidic bus, while directing the inputs from all of the other input ports to the at least one output port through the
  • a rotation of the actuator head at a predetermined angle selects which port is connected to the at least one sensing port, and ensures that all of the other ports are connected to the at least one output port.
  • the first fluidic bus comprises two separate sections, each of which is connected to a respective common output port, thereby allowing some of the fluidic channels to have a different common output port so that the inputs from the input ports do not have to mix from every channel.
  • the fluidic network further comprises one or more additional ports for flushing the first fluidic bus and/or the second fluidic bus.
  • the fluidic device is a direct-access valve or a random-access valve.
  • the invention in another aspect, relates to a fluidic device comprising: a fluidic chip having a fluidic network comprising a plurality of channel modules, each channel module being in fluidic communication with a pair of input ports, at least one make-up media port, and at least one sensing port; and an actuator configured to engage with the fluidic network to control each channel module to switch between a run mode in which the pair of input ports is fluidically connected to each other, and an analysis mode in which one of the pair of input ports is fluidically connected to the at least one make-up media port, while the other of the pair of input ports is fluidically connected to the at least one sensing port to which an analytical instrument is operably connected.
  • the pair of ports of each channel module is operably coupled with a fluidic module and a recirculating pump, such that when said channel module is in the run mode, the fluidic module is fluidically connected to the recirculating pump in a circulating loop, and when said channel module is in the analysis mode, make-up media from the at least one make-up media port is pumped into the fluidic module, and output media from the fluidic module is delivered to the at least one sensing port.
  • the fluidic device is configured such that each fluidic module is individually perfusable with its output media directed to the at least one sensing port without disturbing the flow of the others.
  • the fluidic module comprises a bioreactor, wells, an organ-on-chip, chemostats, or a combination of them.
  • the fluidic chip has a body in which the fluidic network is formed, and a fluidic chip registration means formed on the body for aligning the fluidic chip with a support structure.
  • the fluidic chip registration means is configured such that multiple fluidic chip orientations are allowed while maintaining automatic and precise mechanical alignment to the support structure.
  • the fluidic chip is configured such that one or more plug-in accessories are addable in or removable from the fluidic chip.
  • the fluidic chip is formed of an elastic material such that compression of the actuator on the body causes at least one of the channels to be occluded.
  • the fluidic network further comprises a first fluidic bus and a second fluidic bus, and wherein each channel module is connected between the first fluidic bus and the second fluidic bus.
  • each channel module has an intermediate channel connected between the first fluidic bus and the second fluidic bus; a first channel connected to the intermediate channel at a first position and one of the pair of input ports; and a second channel connected to the intermediate channel at a second position and the other of the pair of input ports, wherein the first position is between the first fluidic bus and the second position, and the second position is between the first position and the second fluidic bus.
  • the actuator comprises a plurality of actuating elements disposed on the body of the fluidic chip, such that compression of an actuating element on the body causes fluidic flow through a corresponding channel portion at which said actuating element is located to be occluded, wherein each of three actuating elements of the plurality of actuating elements are over the intermediate channel of a respective channel module at first, second, and third locations in first, second, and third channel portions of the intermediate channel, respectively, wherein the first channel portion is between the first fluidic bus and the first position, the second channel portion is between the second position and the second fluidic bus, and the third channel portion is between the first and second positions.
  • the actuator further comprises an actuator head for selectively compressing or relaxing each of the plurality of actuating elements.
  • the actuator head comprises an outer relief for controlling access to the at least one sensing port, an inner relief for controlling access to the at least one make-up media port, and a middle relief pocket having two ends and a gap defined therebetween.
  • all of the first and second actuating elements are pressed by the surface of the actuator head, while all of the third actuating elements are relaxed by the middle relief pocket, so that each channel module is in the run mode.
  • the first and second actuating elements on said channel module are relaxed by the first and second reliefs, respectively, and the third actuating element on said channel module is pressed by the gap, all of the first and second actuating elements on the other channel modules are pressed by the surface of the actuator head, while all of the third actuating elements on the other channel modules are relaxed by the middle relief pocket, so that said channel module is in the analysis mode and all of the other channel modules are in the run mode.
  • a rotation of the actuator head at a predetermined angle selects which channel module is in the analysis mode.
  • replacement fluid can be injected into the output line of the isolated module without disturbing the flows of the other fluidic modules.
  • the fluidic network further comprises one or more additional ports for flushing the first fluidic bus and/or the second fluidic bus.
  • the fluidic device is a direct-access valve or a random-access valve.
  • FIGS. 1 A-1F show schematically examples of single bioreactor perfusion topologies.
  • FIGS. 2A-2D show schematically examples of multiple bioreactor sensing valves.
  • FIGS. 3A-3B show schematically a circular, through-plate valve, according to embodiments of the invention.
  • FIGS. 4A-4B show schematically another circular, through-plate valve, according to embodiments of the invention.
  • FIGS. 5A-5G show schematically an analytical valve, according to embodiments of the invention.
  • FIGS. 6A-6C show schematically a lagging actuator random-access valve, according to embodiments of the invention.
  • FIGS. 7A-7C show schematically different implementations of a pump, according to embodiments of the invention.
  • FIGS. 8A-8C show schematically a universal valve, according to embodiments of the invention.
  • FIGS. 9A-9J show schematically a sensing valve.
  • FIG. 10 shows schematically an alternative sensing valve, according to embodiments of the invention.
  • FIGS. 11A-11M show schematically a cut-in valve, according to embodiments of the invention.
  • FIG. 12 shows schematically a cut-in valve with individual make-up inputs, according to embodiments of the invention.
  • FIGS. 13A-13D show schematically a random-access cut-in valve, according to embodiments of the invention.
  • first, second, third, etc. may be used herein to describe various elements, components, regions, layers and/or sections, these elements, components, regions, layers and/or sections should not be limited by these terms. These terms are only used to distinguish one element, component, region, layer or section from another element, component, region, layer or section. Thus, a first element, component, region, layer or section discussed below can be termed a second element, component, region, layer or section without departing from the teachings of the invention.
  • relative terms such as “lower” or “bottom” and “upper” or “top,” may be used herein to describe one element's relationship to another element as illustrated in the figures. It will be understood that relative terms are intended to encompass different orientations of the device in addition to the orientation shown in the figures. For example, if the device in one of the figures is turned over, elements described as being on the “lower” side of other elements would then be oriented on the “upper” sides of the other elements. The exemplary term “lower” can, therefore, encompass both an orientation of lower and upper, depending on the particular orientation of the figure. Similarly, if the device in one of the figures is turned over, elements described as “below” or “beneath” other elements would then be oriented “above” the other elements. The exemplary terms “below” or “beneath” can, therefore, encompass both an orientation of above and below.
  • “around,” “about,” “substantially” or “approximately” shall generally mean within 20 percent, preferably within 10 percent, and more preferably within 5 percent of a given value or range. Numerical quantities given herein are approximate, meaning that the terms “around,” “about,” “substantially” or “approximately” can be inferred if not expressly stated.
  • the terms “comprise” or “comprising,” “include” or “including,” “carry” or “carrying,” “has/have” or “having,” “contain” or “containing,” “involve” or “involving” and the like are to be understood to be open-ended, i.e., to mean including but not limited to.
  • the phrase “at least one of A, B, and C” should be construed to mean a logical (A or B or C), using a non-exclusive logical OR.
  • the term “and/or” includes any and all combinations of one or more of the associated listed items.
  • FIGS. 1 A-1F show five different configurations of reservoir/bioreactor assemblies that are in common use.
  • FIG. 1 A shows gravity-reservoir, single-chamber bioreactor assembly 100, which comprises a supply reservoir 106 containing supply media 107 and connected by tubing 108 to single-chamber bioreactor 109.
  • the effluent media is delivered by tubing to selector valve 120 that delivers sample to on-line media analyzer 121 whose analyzed media 127 is sent to waste.
  • the media not sent to the analyzer 121 is passed on to collection reservoir 116 containing collected media 117 that can be analyzed off-line or sent to waste.
  • FIG. IB shows a gravity-reservoir, dual-chamber bioreactor assembly 101 that uses two input reservoirs 106 to perfuse dual-chamber bioreactor 118 whose media is sent either to a pair of analyzers 121 or to separate waste containers 116. .
  • FIG. 1C shows a pumped, dual-reservoir single-chamber bioreactor assembly 102 that utilizes pump 130 to return collected media 117 to the supply reservoir 106 via return tubing 138.
  • FIG. ID shows a pumped, single-reservoir single-chamber bioreactor assembly 103 for which the pump 130 directly delivers media to the bioreactor 109.
  • FIG. IE shows an assembly 104 comprising a plurality of N gravity-reservoir, single chamber bioreactors, each of which has its own supply and collection reservoirs, selector valve, and analyzer.
  • FIG. IF shows a similar assembly 105 comprising a plurality of N pumped, single reservoir, single-chamber bioreactors with only one reservoir for each bioreactor.
  • FIG. 2A shows an assembly 204 comprising a plurality of N gravity-reservoir, single chamber bioreactors, where each bioreactor 209 has a supply reservoir 206 containing supply media 207 and connected by tubing 208 to the common multichannel selector valve 222 with a common collection reservoir 216 containing collected media 217 that can be analyzed off-line or sent to waste.
  • the multichannel selector valve 222 is also connected by sample analysis port/line 220 to on-line media analyzer 221 whose analyzed media 227 is sent to waste.
  • FIG. 2B shows a similar assembly 204 that differs from that in FIG. 2A by the use of a multichannel selector valve 223 with N separate collection reservoirs 216 and a single sample analysis port and line 220.
  • FIG. 2C shows an assembly 205 comprising a plurality of N pumped, single-reservoir, single-chamber bioreactors 209 with N tubes 208 connected to the N-channel selector valve 223 with separate collection reservoirs 216.
  • a multi-channel pump 231 is comprised of N pumps 230 operating in parallel to return collected media 217 from each collection reservoir 216 directly to the corresponding bioreactor 209 via return tubing 238.
  • N bioreactors N sets of tubing 208 and 238 (which could be configured as ribbon tubing), and an N-channel pump, with all of the N media streams being analyzed by on-line media analyzer 221.
  • FIG. 2D shows a configuration similar to that of FIG. 2C, except that the N-channel selector valve 224 connects not only to the separate collection reservoirs, but also via media make-up line 228 to a make-up media reservoir 225 filled with make-up media 226 to replace the analyzed media 227 that is sent to waste when a particular reservoir is being sampled for analysis by analyzer 221.
  • FIGS. 3 A-3B are perspective and plan views of a circular through-plate 25-channel valve fluidic chip 301, showing an actuated surface, working channels 333, registration/alignment protrusions 306, and interface ports 302, according to one embodiment of the invention.
  • FIG. 4A shows an enclosed valve cartridge 400 according to one embodiment of the invention.
  • This configuration utilizes the position data provided by motor encoder 439 to align a valve actuator 416 in order to open specific channels in the valve fluidic chip 401.
  • the valve actuator 416 is a cylinder made from acetal resin or other material. Topography on the lower face of the valve actuator 416, such as a groove 417, pockets, or similar features, displaces balls 418 as the actuator 416 is rotated.
  • the ball cage 419 constrains movements of the balls 418 to the vertical axis via holes 420 within which the balls reside.
  • the ball cage 419 is constrained against interior faces of surrounding standoff plates/tabs/flanges 406, thereby preventing rotational movement of the ball cage 419 and the balls 418.
  • FIG. 4B shows an expanded, exploded view of a 25-channel valve subassembly according to one embodiment of the invention.
  • FIGS. 5A-5G show the operation and utility of the multi-port, multi-throw analytical valve to control the connection of the output flow of one or more perfused microbioreactors, chemostats, or organ chips 540 to a metabolic or other sensor 550 or a waste reservoir 551.
  • FIGS. 5 A and 5B show the operation and utility of the multi-port, multi-throw analytical valve to control the connection of the output flow of one or more perfused microbioreactors, chemostats, or organ chips 540 to a metabolic or other sensor 550 or a waste reservoir 551.
  • the output flow of the bioreactor(s) is never blocked.
  • the output of the organ(s) 540 passes through the sensor 550 by the position of the valve 575, and the common fluidic channels of the valve 575 and the pump 580 are rinsed by having the pump 580 withdraw rinse media from reservoir 590 and direct it towards the waste reservoir 551.
  • Other calibration solutions 560 could be pumped to the waste reservoir 551 as well, depending upon the position of valve 570. Further, as shown in FIG.
  • valve 575 in order to calibrate the metabolic sensor 550, which is prone to drift, the valve 575 is switched to the calibration mode and valve 570 is used to select calibration media (CAL 02 shown) from one of several calibration media reservoirs 560 to perform the calibration operation, the calibration media is delivered to the sensor 550 by the pump 580, and the output(s) of the organ(s) 540 is directed towards the waste 551 by the action of the valve 575.
  • the organ(s) may be perfused by gravity, pneumatic pressure, or a pump (not shown) such that the organ is always continuously perfused.
  • FIG. 5C shows a single microfluidic analytical valve 500 that can implement the various modes described in FIGS. 5A and 5B with a single actuator and motor.
  • the analytical valve 500 includes a fluidic chip 501 with protrusions 521 that serve as tubing ports and anchor the fluidic chip 501 to the support plate 514, and microfluidic channels in the fluidic network 520 are sealed within the fluidic chip 501.
  • the actuator 502, with ball-actuating grooves 530 in an actuating surface 533, is driven by a motor shaft 505, and includes actuating elements 503, a ball cage 504, and an off-board pump (not shown).
  • the actuator 502 rotates to change the state of the valve as the caged actuating elements (in this embodiment balls) slide against the actuator.
  • the analytical valve 500 collects an analyte from the bioreactor (not shown) through a bioreactor input channel, and sends it to a waste reservoir (not shown) through a waste output channel, while also collecting a rinse solution and directing it first through an internal fluidic channel 510, the off-board pump (not shown), through another internal fluidic channel 511, and finally to the sensor (not shown) through a sensor output channel.
  • a second state as shown in FIG.
  • a calibration solution is selected (e.g., CAL 08) through a corresponding calibration input channel and directed to the sensor through the sensor output channel, while the bioreactor input channel is interconnected to the waste output channel to send the effluent from the bioreactor to waste.
  • the effluent is directed from the bioreactor to waste, while all other conduits are closed and idle.
  • the bioreactor input channel is switched and interconnected to the sensor output channel to direct the effluent from the bioreactor to the sensor for analysis, while all other conduits are idle.
  • a rinse solution is directed through a rinse input channel to pass the length of channels 512 and 513 and sent to waste, while the effluent is collected from the bioreactor and sent to the sensor.
  • the analytical valve 500 is designed such that fluid being pumped into the fluidic chip 501 from the bioreactor as effluent has an outlet at all times (either sent to the sensor, to waste, or to both), as not to cause a dead-end scenario that might rupture the fluidic chip 501 or interrupt perfusion of a sensitive organ chip.
  • 5C-5G is determined by the available circumference of the through-plate fluidic chip 501 and the underlying tubing-port protrusions 521 that anchor the fluidic chip 501 to the support plate 514, and the minimum spacing between individual channels in the fluidic network 520.
  • a typical valve has 1 inlet/outlet port and 25 outlet/inlet ports, depending upon the chosen flow direction.
  • different designs of the fluidic network 520 could use some of the 23 channels to service two or more independent bioreactors whose output is either sent to a sensor or waste.
  • Both of the valves shown in FIGS. 4A-4B and 5A-5G are serial access, i.e., the channels are opened and closed in sequence, so that if the valve has channel 4 open but instead it is necessary to open channel 6, with these valves the actuator must be turned until channel 4 is closed, followed by channel 5 being opened, then channel 5 closed, and finally channel 6 is opened.
  • FIGS. 6A-6C show how a lagging-actuator random-access (also called direct-access) valve 600 utilizes a two-part actuator with control over rotational direction and range to randomly access any channel in the valve 600, according to certain embodiments of the invention.
  • the rotary valves described above are all serial access valves, with the ports being addressed one after another as the actuator is rotated. As long as the fluidic channels were not pressurized and the actuator was rotated quickly, there would be minimal fluid displaced from channels during rotation.
  • the lagging-actuator random-access valve has the ability to go from one valve position to a distant one without having to transiently open and close each intermediate channel. As shown in FIG.
  • a valve chip 600 has multiple actuating element (ball) locations 601 operating on each of the input/output fluidic channels 602 connected to a common fluidic channel 603.
  • Each input/output fluidic channel 602 is normally pinched closed by two radially adjacent balls 620 and 621 (see FIG. 6C).
  • a given input/output channel 602 pinched by at least one ball 620 or 621 is closed.
  • the compressive force applied to both corresponding balls 620 and 621 must be relieved concurrently. In other words, if either or both balls 620 and 621 are not actuated from their normally-closed position, the channel 602 remains closed.
  • FIG. 6B shows a lagging-actuator assembly 610, including an inner (driven) actuator 611 and an outer (driving) actuator 612, as coaxially aligned on a motor shaft 613.
  • the outer actuator 612 is locked to the motor shaft 613 by a set screw (not shown) in the screw hole 618.
  • a limiting pin 614 is affixed to the outer actuator 612, and its motion is constrained within the bounds of a limiting pocket 615 on the inner actuator 611.
  • the limiting pin 614 is driven to the clockwise extreme end of the limiting pocket 615, and the reverse is also true.
  • the arc-length of the limiting pocket 615 establishes an intended backlash or "lag" between the outer actuator 612 and the inner actuator 611.
  • the limiting pin 614 contacts clockwise the extreme end of the limiting pocket 615, and the outer actuator 612 and the inner actuator 611 begin to rotate as a whole, with an outer groove 617 on the outer actuator 612 and an inner groove 616 on the inner actuator 611 unaligned. With the grooves 616 and 617 unaligned, all input/output channels 602 see compression from at least one ball 620 and 621 at all times.
  • the inner actuator 611 remains stationary (by friction and/or presence of the actuator element 620 within the inner groove 616) until the limiting pin 614 contacts the other end of the limiting pocket 615, and the inner actuator 611 and the outer actuator 612 rotate as a whole with the grooves 616 and 617 axially aligned.
  • both pinch points established by the balls 620 and 621 are relieved concurrently, and the target channel 602 opens to flow.
  • FIG. 6C shows a lagging-actuator assembly 610 as seen by the fluidic chip 600 as well as corresponding cross-sectional views, in multiple steps (from left to right) required for opening target channel 602 to flow.
  • the inner ball 620 and the outer ball 621 remain pressed into the fluidic chip, thereby blocking flow.
  • the lagging-actuator assembly 610 rotates from its previous position, no channel has both balls released concurrently.
  • the outer groove 617 reaches and aligns with the outer ball 621.
  • the outer groove 617 has rotated past alignment with the outer ball 621, while the inner groove 616 reaches and aligns with the inner ball 620.
  • rotation reverses until the outer groove 617 also aligns with the outer ball 621 while the inner (driven) actuator 611 remains stationary. Both balls have now retracted to open the target channel 602.
  • the actuator assembly 610 may spin along a same direction continuously (rather than moving to a designated channel and stopping) with the grooves unaligned, causing the balls 620 and 621 to sequentially relax momentarily. As the pinched channels 602 momentarily relax, the fluid previously displaced by respective balls 620 and 621 returns to fill the void. As the actuator assembly 610 continues to rotate, the balls 620 and 621 once again pinch the channel 602, and the corresponding fluid is again displaced. The result is a pulsation of fluid through the conduit within and/or connected to the channel 602, and this pulsation could be used at the end of the conduit to mix or agitate a reservoir of liquid in which it is submerged.
  • One of the objectives of the invention is to develop valves that would allow a portion of the effluent from any and/or all of a plurality of bioreactors or chemostats to be sent to a common analytical instrument while the effluent from all other bioreactors or chemostats flows without interruption.
  • the invention relates to a fluidic device.
  • the fluidic device includes a fluidic chip having a fluidic network comprising a plurality of fluidic channels in fluidic communication with a plurality of input ports, at least one output port, and at least one sensing port; and an actuator configured to engage with the fluidic network to control each fluidic channel to switch between an open state in which fluidic flow through said fluidic channel is permitted and a closed state in which no fluidic flow through said fluidic channel is permitted, so as to selectively collect fluid from multiple inputs via the plurality of input ports, and direct either all of the multiple inputs to the at least one output port, or all but a single selected input to the at least one output port and the single selected input to the at least one sensing port to which an analytical instrument is operably connected.
  • the plurality of input ports is operably coupled with a plurality of fluidic modules, wherein in operation, the plurality of fluidic modules is individually perfused, and all outputs of the plurality of fluidic modules are directed to the at least one output port, or an output of any one of the plurality of fluidic modules is directed to the at least one sensing port, while outputs of all other fluidic modules are directed to the at least one output port.
  • the outputs from all other fluidic modules flow without interruption.
  • the plurality of fluidic modules comprises bioreactors, chemostats, wells, organs-on-chips, or a combination of them.
  • the fluidic chip has a body in which the fluidic network is formed, and a fluidic chip registration means formed on the body for aligning the fluidic chip with a support structure.
  • the fluidic chip registration means is configured such that multiple fluidic chip orientations are allowed while maintaining automatic and precise mechanical alignment to the support structure.
  • the fluidic chip is configured such that one or more plug-in accessories are addable in or removable from the fluidic chip.
  • the fluidic chip is formed of an elastic material such that compression of the actuator on the body causes at least one of the channels to be occluded.
  • the plurality of fluidic channels comprises a first fluidic bus, a second fluidic bus, and a plurality of intermediate channels, each intermediate channel being in fluidic communication with at least one of the plurality of input ports and connected to the first fluidic bus and/or the second fluidic bus.
  • the actuator comprises a plurality of actuating elements disposed on the body of the fluidic chip with each actuating element at a location that is over an intermediate channel and is located between a respective port and one of the first fluidic bus and the second fluidic bus to which said intermediate channel is connected, such that compression of said actuating element on the body causes fluidic flow between said respective port and said one of the first fluidic bus and the second fluidic bus through said intermediate channel to be occluded.
  • the locations of the plurality of actuating elements on the body of the fluidic chip comprise first locations and second locations, wherein the first locations comprise each actuating element location over a respective intermediate channel between a respective port and the first fluidic bus, and the second locations comprise each actuating element location over a respective intermediate channel between a respective port and the second fluidic bus.
  • the actuator further comprises an actuator head for selectively compressing or relaxing each of the plurality of actuating elements.
  • the actuator head comprises an outer actuator head having an outer groove corresponding to one of the first locations of the plurality of actuating elements on the body, wherein the outer groove, when aligned with the corresponding one of the first locations, relieves the corresponding actuating element so that the corresponding port is connected to the first fluidic bus; and an inner actuator head sleeved in the outer actuator head, having an inner groove corresponding to one of the second locations of the plurality of actuating elements on the body, wherein the inner groove, when aligned with the corresponding one of the second locations, relieves the corresponding actuating element so that the corresponding port is connected to the second fluidic bus, wherein one of the outer actuator head and the inner actuator head is a driving actuator head driven by a motor, and the other of the outer actuator head and the inner actuator head is a driven actuator head driven by said driving actuator head.
  • each of the outer actuator head and the inner actuator head has a circular-segment pocket with a near-360° sweep
  • the actuator head further comprises a single limiting element, whose motion is constrained by the pockets, allowing the driving actuator head and the driven actuator head to rotate or remain stationary independently until the limiting element contacts opposing ends of both pockets, at which point both actuator heads rotate as one, wherein when the direction of the motor is then reversed, the motion of each of the actuator heads becomes independent again.
  • the actuator head comprises a first relief pocket having two ends and a gap defined therebetween, and a second relief aligned with the gap along an intermediate channel, wherein the first relief pocket and the gap are corresponding to the first locations of the plurality of actuating elements on the body of the fluidic chip, and the second relief is corresponding to one of the second locations of the plurality of actuating elements on the body of the fluidic chip, such that in operation, one of the actuating elements on the first locations is pressed by the gap and the others of the actuating elements on the first locations are relaxed by the first pocket, and one of the actuating elements on the second locations is relaxed by the second relief and the others of the actuating elements on the second locations are pressed by the surface of the actuator head, thereby directing the single selected input from the input port connected to said intermediate channel with which the gap and the second relief are aligned to the at least one sensing port through the second fluidic bus, while directing the inputs from all of the other input ports to the at least one output port through
  • a rotation of the actuator head at a predetermined angle selects which port is connected to the at least one sensing port, and ensures that all of the other ports are connected to the at least one output port.
  • the first fluidic bus comprises two separate sections, each of which is connected to a respective common output port, thereby allowing some of the fluidic channels to have a different common output port so that the inputs from the input ports do not have to mix from every channel.
  • the fluidic network further comprises one or more additional ports for flushing the first fluidic bus and/or the second fluidic bus.
  • the fluidic device is a direct-access valve or a random-access valve.
  • the invention in another aspect, relates to a fluidic device comprising: a fluidic chip having a fluidic network comprising a plurality of channel modules, each channel module being in fluidic communication with a pair of input ports, at least one make-up media port, and at least one sensing port; and an actuator configured to engage with the fluidic network to control each channel module to switch between a run mode in which the pair of input ports is fluidically connected to each other, and an analysis mode in which one of the pair of input ports is fluidically connected to the at least one make-up media port, while the other of the pair of input ports is fluidically connected to the at least one sensing port to which an analytical instrument is operably connected.
  • the pair of ports of each channel module is operably coupled with a fluidic module and a recirculating pump, such that when said channel module is in the run mode, the fluidic module is fluidically connected to the recirculating pump in a circulating loop, and when said channel module is in the run mode, make-up media from the at least one make-up media port is pumped into the fluidic module, and output media from the fluidic module is delivered to the at least one sensing port.
  • the fluidic device is configured such that each fluidic module is individually perfusable with its output media directed to the at least one sensing port without disturbing the flow of the others.
  • the fluidic module comprises a bioreactor, chemostats, wells, an organ-on-chip, or a combination of them.
  • the fluidic chip has a body in which the fluidic network is formed, and a fluidic chip registration means formed on the body for aligning the fluidic chip with a support structure.
  • the fluidic chip registration means is configured such that multiple fluidic chip orientations are allowed while maintaining automatic and precise mechanical alignment to the support structure.
  • the fluidic chip is configured such that one or more plug-in accessories are addable in or removable from the fluidic chip.
  • the fluidic chip is formed of an elastic material such that compression of the actuator on the body causes at least one of the channels to be occluded.
  • the fluidic network further comprises a first fluidic bus and a second fluidic bus, and wherein each channel module is connected between the first fluidic bus and the second fluidic bus.
  • each channel module has an intermediate channel connected between the first fluidic bus and the second fluidic bus; a first channel connected to the intermediate channel at a first position and one of the pair of input ports; and a second channel connected to the intermediate channel at a second position and the other of the pair of input ports, wherein the first position is between the first fluidic bus and the second position, and the second position is between the first position and the second fluidic bus.
  • the actuator comprises a plurality of actuating elements disposed on the body of the fluidic chip, such that compression of an actuating element on the body causes fluidic flow through a corresponding channel portion at which said actuating element is located to be occluded, wherein each of three actuating elements of the plurality of actuating elements are over the intermediate channel of a respective channel module at first, second and third locations in first, second and third channel portions of the intermediate channel, respectively, wherein the first channel portion is between the first fluidic bus and the first position, the second channel portion is between the second position and the second fluidic bus, and the third channel portion is between the first and second positions.
  • the actuator further comprises an actuator head for selectively compressing or relaxing each of the plurality of actuating elements.
  • the actuator head comprises an outer relief for controlling access to the at least one sensing port, an inner relief for controlling access to the at least one make-up media port, and a middle relief pocket having two ends and a gap defined therebetween.
  • all of the first and second actuating elements are pressed by the surface of the actuator head, while all of the third actuating elements are relaxed by the middle relief pocket, so that each channel module is in the run mode.
  • the first and second actuating elements on said channel module are relaxed by the first and second reliefs, respectively, and the third actuating element on said channel module is pressed by the gap, all of the first and second actuating elements on the other channel modules are pressed by the surface of the actuator head, while all of the third actuating elements on the other channel modules are relaxed by the middle relief pocket, so that said channel module is in the analysis mode and all of the other channel modules are in the run mode.
  • a rotation of the actuator head at a predetermined angle selects which channel module is in the analysis mode.
  • replacement fluid can be injected into the output line of the isolated module without disturbing the flows of the other fluidic modules.
  • the fluidic network further comprises one or more additional ports for flushing the first fluidic bus and/or the second fluidic bus.
  • the fluidic device is a direct-access valve or a random-access valve.
  • the multichannel microfluidic valves enable efficient scaling of the perfusion systems depicted in FIGS. 1 A-1F.
  • Each of the valve configurations is comprised of a network of fluidic microchannels within a deformable, elastomeric polymer or similar material. Rigid actuating elements superimposed upon certain critical regions of the network(s), when displaced by an actuator, either pinch the region closed and block passage of fluid therethrough, or relax, thereby allowing the channel to open and permitting fluid to pass.
  • the valves are connected to external constructs (pumps, reservoirs, sensors) via tubing or other conduits attached to integrated ports.
  • Perfusate circulation within the systems of FIG. 1 A-1F may be gravity-driven, as shown in FIGS. 1 A, IB, and IE, or may be actively accomplished using pumps, as shown in FIGS. 1C, ID, and IF.
  • the pumps can be a single-channel spiral pump 710 shown in FIG. 7A according to one embodiment of the invention, which includes a single spiral channel 711. With recirculation, media flows continuously through the circuit (the single spiral channel 711) at whatever rate is required to recapitulate a physiological residence time in each fluidic model.
  • a two-channel pump 720 shown in FIG. 7B can be used, according to one embodiment of the invention, which includes two spiral channels 721 and 722.
  • FIG. 3B shows a 25-channel valve that can be used to deliver a selected media or drug to a bioreactor or chemostat
  • the downstream network 731 collects the media that perfuses each of the 12 bioreactors and is collected in N reservoirs 117, and with a single, time-shared pump 130 returns the media to the appropriate supply reservoir 106, which also serves to control media oxygenation.
  • the channels within this valve 730 are opened in port-pairs; that is, for example, Port 5 would be connected to the outlet port via its corresponding common channel at the same time that Port 17 is opened to the collection port via its respective common channel.
  • Ports 12 and 24 are activated concurrently, as shown in FIG. 7C, and so on. For any given active port-pair, the remaining ports are closed to flow, and the connected modules would see no net movement of fluid therethrough.
  • valve 730 enables time-division multiplexing to control the relative perfusion of each of N ⁇ 12 modules.
  • the 13th port in the valve can be used to flush the common channel if desired.
  • the two-channel spiral pump 720 shown in FIG. 7B ensures that whatever volume of media is withdrawn from a chamber for sampling is made up with media withdrawn from a replacement reservoir.
  • a 1 x25 port valve at the output of any pump could direct the sample to any well in a 24-well plate, with the 25th port being used to flush the pump and valve to prevent mixing of samples from different chambers.
  • FIGS. 8A-8C show a universal valve chip 800 as used with a lagging actuator assembly 810, a combination which allows independent selection of both collection source and destination sink using a single valve construct with a single motor, according to embodiments of the invention.
  • Two fluidic buses (common channels) 801 and 802 are connected by an offboard pump 803.
  • a shunt (not shown) may be installed in lieu of the pump 803.
  • FIG. 8C A schematic diagram of the fluidic control network shown in FIG. 8A is clarified by FIG. 8C.
  • bus 801/802 may serve as a collection conduit or an output conduit, while the other bus 802/801 serves as output or collection, respectively.
  • bus 801 is the collection conduit and the bus 802 is the output conduit, but these could be switched.
  • Ports 808 and 809 may serve as flush ports to clear common channels (buses) 801 and 802, or may be used as additional analysis ports. Additional ports and corresponding connecting channels may be added to (or removed from) the buses 801 and 802.
  • the lagging actuator assembly 810 functions similarly to the lagging actuator assembly 610 shown in FIG. 6B, and is used to select one collection port 806 and one output port 807. This is accomplished when the actuator groove 813 in the driving actuator 811 aligns with the actuating element 804, and the actuating groove 814 in the driven actuator 812 aligns with the actuating element 805.
  • those actuating elements 804 and 805 relax, and the corresponding port 806/807 is connected to the common channel of its respective bus 801/802, and thereby selected ports 806 and 807 become connected to each other.
  • one actuating element 804/805 in each bus 801/802 opens while all other actuating elements remain pressed into their corresponding regions, and those channels remain pinched closed.
  • the lagging actuator 810 has ample backlash such that any permutation of port-pair interconnections can be achieved.
  • This backlash is accomplished by circular-segment pockets 816 with a near-360° sweep, and a single limiting ball 815, whose motion is constrained by the pockets 816, allowing the driving actuator 811 and the driven actuator 812 to rotate or remain stationary independently until the limiting ball 815 contacts opposing ends of both pockets 816, at which point both actuator parts 811 and 812 rotate as one.
  • the motion of each actuator 811/812 becomes independent again.
  • the sum of the pocket arc-lengths equals the backlash of the actuator assembly (not accounting for ball diameter).
  • the outer actuator 811 is the driving actuator, and time-division multiplexing of solution in conduits attached to the outer bus 801 may be readily achieved, as described previously.
  • the driving actuator and the driven actuator may manifest such that the inner actuator 812 drives the outer actuator 811. By this means, time-division multiplexing may be readily achieved on the inner fluidic bus 802.
  • the actuator assembly concept may be extrapolated to include more than the two actuating “rings,” which would address additional fluidic buses.
  • the mechanical function of such embodiments would operate similar to the combination lock mechanism on an old-fashioned safe; that is, the primary actuator would drive the secondary actuator, which in turn would drive subsequent subordinate actuators in a cascading fashion.
  • FIGS. 9A-9J provide details of a sensing valve assembly 920 according to embodiments of the invention, in which the fluidic chip 900 collects fluid from multiple (N ⁇ 24 as shown) inputs via ports such as 903 and 906, and directs either all of its inputs to a common output port 908, or all but a single selected input 906 to a common output 908, while fluid entering outlier port 906 is directed to an isolated “sensing” output 909, to which an analytical instrument is connected, as 221 shown in FIG. 2C.
  • the face of the actuator 910 presses the plurality of actuating elements 904 and 905 into the underlying elastomeric polymer, thereby pinching the corresponding channel closed, except in the regions where the outer relief pocket 913 has all outer actuating elements 904 relaxed, and where the inner actuating relief 914 has the inner actuators 905 relaxed.
  • the gap 915 in the outer relief pocket 913 aligns with an outer actuating element 904, that element closes the connection of the port at that location to the outer fluidic bus 901 and hence a common output port 908, while the inner relief 914 at that angle allows that inner actuating element 905 to relax, thereby connecting the port at that angle to the inner fluidic bus 902 and hence a sensing port 909.
  • rotation of the actuator 910 selects which port is connected to the sensing port 909 and ensures that all of the other ports are connected to the common output port 908.
  • the bioreactor media line that is to be analyzed is selected by rotating the actuator 910 to a position in which the actuating elements 904 and 905 corresponding to the selected target port 906 are switched such that the selected target port 906 becomes isolated from the common output 908 and opened to the sensing output 909, as exemplified in FIG. 9A. Meanwhile, all of the other ports 903 remain connected to the common output port 908 and isolated from the sensing port 909. As shown in FIG. 9C, the actuating elements 904 and 905 are constrained by a cage 921, and the fluidic chip 900 is constrained by a baseplate 922.
  • FIGS. 9D-9I show the actuator position (FIGS. 9D, 9F and 9H) and the channel and port status (FIGS. 9E, 9G and 91) for three actuator positions (240°, 168°, and 48°).
  • the position of the inner relief 914 is such that the two inner actuating elements 990 at positions 16 and 17 remain compressed, and all of the outer actuating elements 904 are relaxed, so that all ports are connected to the common channel 901.
  • input #12 (991) is connected to the sensing/analysis port 909 because the inner actuating element 905 at that position is relaxed while the corresponding outer one 904 is compressed.
  • FIGS. 9H and 91 input #4 is connected to the analysis port 909, from which all other inputs are isolated.
  • FIG. 9J shows sequential connection and disconnection of adjacent ports can be accomplished by the rotation of the actuator 910 in increments of 12°. Note that while this valve allows a single bioreactor to be sampled at a given time, it also prevents any backpressure on the organs not being sampled, as their perfusate is directed towards waste rather than being blocked, which could increase the pressure in the reactor.
  • FIG. 10 shows a minor modification of the valve 900, wherein the outer fluidic bus is divided into two separate sections 901 A and 901B, each of which is connected to a respective common output 908A or 908B.
  • This allows some of the channels to have a different normal common channel so that the outputs do not have to mix from every channel.
  • Such modification allows one to send solutions to the analytical line 909 (calibration solutions) that should not get mixed with the solution going to the other (organ) channels.
  • FIGS. 11 A-l 1M show a “cut-in” or “insert” valve according to embodiments of the invention.
  • FIGS. 11 A and 1 IB explain the concept of the “cut-in” or “insert” valve that supports a run mode in which a bioreactor is connected to a recirculating pump, and an analysis mode in which make-up media is pumped into the bioreactor, displacing media that is delivered to an analyzer, as discussed above for FIG. 2D.
  • For each module of the valve there are four connections and three actuating elements.
  • the actuating element 1105 is relaxed so that the recirculating pump can push media through the bioreactor, while the actuating elements 1104 and 1106 compress their corresponding channels, thereby isolating this bioreactor-pump module from both the analyzer and the media supply.
  • the actuating element 1105 In the analysis mode, the actuating element 1105 is in compression and the actuating elements 1104 and 1106 are relaxed, thereby allowing the make-up media to be drawn from its reservoir and the bioreactor effluent delivered to the analyzer, as shown in FIG. 1 IB.
  • FIGS llC and 1 ID show the through-plate fluidic channel layout 1100 along with the actuating elements 1104-1106 that accomplish these modes for a total of 12 bioreactors.
  • a rotation of the actuator by the appropriate angle allows the sequential sampling of each bioreactor.
  • the topology enables serial access, and the valve has a low dead volume in each fluidic module loop. The majority of this valve’s dead volume is in the analytical loop, and the clearing of that dead volume is limited by the amount of fresh media that can be added to sweep the analytical loop. In other embodiments, an extra input port uses fresh media to flush the channels.
  • FIG. 1 IE provides details of the actuating recesses of the actuator 1110, with an inner actuating relief 1114 controlling access to the media reservoir, an outer actuating relief 1116 controlling access to the analyzer, and the combination of a recess 1113 and a gap 1115 controlling the recirculation or isolation of the two halves of the circuit.
  • FIG. 1 IF shows the channel layout of the fluidic chip 1100.
  • An outer fluidic bus 1133 is connected to an analyzer port 1134.
  • An inner fluidic bus 1135 is connected to a make-up media port 1136.
  • the input to the recirculating pump is connected to a port 1137, while the return from the bioreactor is connected to a port 1138.
  • FIG. 11G provides details of the cut-in valve subassembly 1120, in which the fluidic chip 1100 interfaces with multiple (N ⁇ 12 as shown) bioreactors via ports 1137 and 1138 shown in FIG. 1 IF.
  • the face of an actuator 1110 presses the plurality of the outer, middle, and inner actuating elements 1104, 1105, and 1106, respectively, into the underlying elastomeric polymer (fluidic chip) 1100, thereby pinching the corresponding channels closed, except in the regions where the middle relief pocket 1113 has all of the middle actuating elements 1105 relaxed; where the inner actuating relief 1114 has an inner actuator 1106 relaxed; and the outer actuating relief 1116 has an outer actuating element 1104 relaxed.
  • the bioreactor media line that is to be analyzed is selected by rotating the actuator 1110 to a position in which the actuating elements 1104 and 1106, corresponding to the selected bioreactor target port, are switched open, and the actuating element 1105 is closed, such that the make-up media enters the valve through port 1136, and by means of the recirculating pump displaces media from the selected bioreactor that is then pushed out through the analysis port 1134.
  • the actuating elements 1104, 1105, and 1106 are constrained by the cage 1121, and the fluidic chip 1100 is constrained by the baseplate 1122.
  • FIGS. 1 lH-11M show the actuating element position (FIGS. 11H, 11 J, and 11L) with black dots as closed and clear-center as open, and the corresponding channel and port status (FIGS. Ill, 1 IK, and 11M) for three actuator positions (0°, 18°, and 42°).
  • actuating element position FIGS. 11H, 11 J, and 11L
  • FIGS. Ill, 1 IK, and 11M the corresponding channel and port status
  • valve allows a single bioreactor to be sampled at a time, it also prevents any backpressure on the bioreactors not being sampled, as their perfusate is recirculated rather than being blocked, which could alter the pressure in the bioreactor.
  • FIG. 12 shows a modification of the valve network 1100, wherein the single make-up media port 1136 is divided into multiple media ports 1140, each of which is connected to a respective fluidic module, which allows switched insertion of analytical devices or other fluidic modules into any fluid line and does not cause any cross contamination between sampled sites.
  • valve designs shown in FIGS. 11 A-l 1M and 12 provide a great deal of advantages in controlling and sensing bioreactors, including chemostats and microchemostats.
  • the valve allows switched insertion of analytical devices or other fluidic modules into any fluid line (currently designed for Tygon 0.060 O.D., but many other tubing sizes are possible) and does not cause cross contamination between sampled sites.
  • This could allow a single analytical instrument to take measurements on isolated systems or take samples from different locations within a single system. Sampling sites are completely independent, which allows them to be arranged either on parallel legs of a perfusion system or in series between experimental points along a path or any desired combination.
  • Sampled devices are completely independent or may be arranged either in parallel or in series with each other or any desired combination.
  • the actuator design also allows easy configuration for a variety of ball position layouts and can accommodate valve designs for various motor sizes (the current design has a 24 mm diameter actuator that may be used with NEMA 8 motors). Actuation on the radial centerline makes scaling and reconfiguration simple.
  • Series measurements along an experimental path allow measurements of both inputs and outputs using a single instrument and allows direct comparisons of input fluid composition vs output fluid composition throughout an experimental sequence.
  • the design simply breaks the fluid path and inserts the sampling loop, which could either be closed for recirculation (other fluidic devices or non-destructive analysis only, e.g., optical density, colorimetric pH, etc.) or connected to a fluid replenish reservoir to add fresh media to replace the volume extracted during sampling. With properly sized compression zones and fluidic channels, this design is zero dead volume in the normal flow loop.
  • the individual channel design allows easy configuration for a variety of channel numbers, which enables it to be used on various motor sizes without additional engineering time to redesign the fluidic channels. Utilizing the radial inline orientation of the compression balls allows this design to be used with the radial cylinder random-access actuator concept.
  • valve provides isolation within its own design, it cannot provide any isolation for organs connected in parallel through external connections, and another valve would be required for that function.
  • FIGS. 6A-6C and 8A-8C can be used to create a random-access cut-in valve, which is shown in FIGS. 13A-13D according to certain embodiments of the invention.
  • Sequential operation is accomplished when running in the counterclockwise direction. Random-access operation is achieved by rotating in the clockwise direction by 216 degrees (120 steps on a standard stepper motor), then rotating to the desired position, and then by reversing back 216 degrees in the counterclockwise direction.
  • valves disclosed herein can be implemented with the cartridge pumps and valves described in this disclosure and earlier patents, or with integrated fluidic chips also described in other patents. If desired, physiological flow pulsations can be provided by periodic changes in pump speed. These advances provide the necessary parallelism to sample with a common instrument various configurations of multiple organs-on-chips, tissue chips, bioreactors, or fluidic modules.
  • Patent No. 9,874,285 B2 (January 23, 2018).
  • Patent No. 10,078,075 B2 (September 18, 2018).
  • NCATS Supports Award-Winning Technology for Drug Development: NIH; 2018

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Clinical Laboratory Science (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Sustainable Development (AREA)
  • Dispersion Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Automatic Analysis And Handling Materials Therefor (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Abstract

L'invention concerne un dispositif fluidique qui comprend une puce fluidique ayant un réseau fluidique comprenant une pluralité de canaux fluidiques en communication fluidique avec une pluralité d'orifices d'entrée, au moins un orifice de sortie et au moins un orifice de détection ; et un actionneur conçu pour venir en prise avec le réseau fluidique pour commander chaque canal fluidique pour commuter entre un état ouvert, dans lequel un écoulement fluidique à travers ledit canal fluidique est permis, et un état fermé, dans lequel aucun écoulement fluidique à travers ledit canal fluidique n'est permis, de façon à collecter sélectivement un fluide à partir de multiples entrées par l'intermédiaire de la pluralité de ports d'entrée et diriger l'une ou l'autre des entrées multiples vers ledit au moins un orifice de sortie ou tout sauf une seule entrée sélectionnée audit au moins un orifice de sortie et l'entrée sélectionnée unique audit au moins un orifice de détection auquel un instrument analytique est connecté de manière fonctionnelle.
EP21841908.3A 2020-07-17 2021-07-19 Systèmes microfluidiques pour bioréacteurs multiples et leurs applications Pending EP4182437A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063053388P 2020-07-17 2020-07-17
PCT/US2021/042141 WO2022016136A1 (fr) 2020-07-17 2021-07-19 Systèmes microfluidiques pour bioréacteurs multiples et leurs applications

Publications (1)

Publication Number Publication Date
EP4182437A1 true EP4182437A1 (fr) 2023-05-24

Family

ID=79555012

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21841908.3A Pending EP4182437A1 (fr) 2020-07-17 2021-07-19 Systèmes microfluidiques pour bioréacteurs multiples et leurs applications

Country Status (2)

Country Link
EP (1) EP4182437A1 (fr)
WO (1) WO2022016136A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11746317B2 (en) 2019-06-28 2023-09-05 Vanderbilt University Massively parallel, multiple-organ perfusion control system

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8546128B2 (en) * 2008-10-22 2013-10-01 Life Technologies Corporation Fluidics system for sequential delivery of reagents
US10078075B2 (en) * 2011-12-09 2018-09-18 Vanderbilt University Integrated organ-on-chip systems and applications of the same
WO2020041342A2 (fr) * 2018-08-20 2020-02-27 Vanderbilt University Systèmes de cartouche, pompes capacitives et soupapes à jets multiples et systèmes de soupape de pompe et leurs applications
WO2014138203A2 (fr) * 2013-03-05 2014-09-12 Board Of Regents, The University Of Texas System Dispositifs microfluidiques pour le traitement rapide et automatique de populations d'échantillons
WO2015153449A1 (fr) * 2014-03-31 2015-10-08 Redshift Systems Corporation Analyseur de fluide avec modulation pour des liquides et des gaz

Also Published As

Publication number Publication date
WO2022016136A1 (fr) 2022-01-20

Similar Documents

Publication Publication Date Title
US10781809B2 (en) Peristaltic micropump and related systems and methods
US10577574B2 (en) Interconnections of multiple perfused engineered tissue constructs and microbioreactors, multi-microformulators and applications of the same
CN110564614B (zh) 多个灌注工程组织构建和微型生化分析仪的互联,多个微型配方设计模块及其应用
US10309545B2 (en) Fluid control device
US10908149B2 (en) Devices for fluid management
US11565256B2 (en) Microfluidic systems, pumps, valves, fluidic chips thereof, and applications of same
WO2022016136A1 (fr) Systèmes microfluidiques pour bioréacteurs multiples et leurs applications
US10124335B2 (en) Integrated fluidic module
EP3802777A1 (fr) Bioréacteurs microfluidiques à compartiments multiples, valves cylindriques rotatives et leurs applications
US20230271181A1 (en) Microfluidic systems for multiple bioreactors and applications of same
US20120138833A1 (en) Multi-Function Eccentrically Actuated Microvalves and Micropumps
US11465144B2 (en) Cartridge systems, capacitive pumps and multi-throw valves and pump-valve systems and applications of same
US20210198607A1 (en) Multicompartment microfluidic bioreactors, cylindrical rotary valves and applications of same
US20230338949A1 (en) Microfluidic systems for multiple bioreactors and applications of same
WO2022016143A1 (fr) Systèmes microfluidiques pour bioréacteurs multiples et leurs applications
CN103586094A (zh) 一种具有环流混合结构的微流控芯片及环流混合方法
US7032605B1 (en) Dual piston rotary valve
US11746317B2 (en) Massively parallel, multiple-organ perfusion control system
WO2022229873A1 (fr) Système de dosage pour fluides
EP3984569A1 (fr) Dispositif de diagnostique in vitro avec separateur de plasma integre
Curtis kk tt| kk kkk kk kkkkkkS (58) Field of Classification Search
CN116809130A (zh) 微流控芯片及其用接头、接口和微流控系统
WO2023104805A1 (fr) Appareil pour l'alimentation d'un milieu liquide dans un système fluidique comprenant une agitation magnétique

Legal Events

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

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

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

Free format text: ORIGINAL CODE: 0009012

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230117

AK Designated contracting states

Kind code of ref document: A1

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

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