US20140127733A1 - Ex vivo microfluidic analysis of biologic samples - Google Patents

Ex vivo microfluidic analysis of biologic samples Download PDF

Info

Publication number
US20140127733A1
US20140127733A1 US14/070,444 US201314070444A US2014127733A1 US 20140127733 A1 US20140127733 A1 US 20140127733A1 US 201314070444 A US201314070444 A US 201314070444A US 2014127733 A1 US2014127733 A1 US 2014127733A1
Authority
US
United States
Prior art keywords
channel
fluid
posts
biological sample
sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/070,444
Other languages
English (en)
Inventor
Soner Altiok
Alla Epshteyn
Angela Holton
David Landis
Abagail Spencer
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.)
Moffit Cancer Center
Charles Stark Draper Laboratory Inc
Original Assignee
Moffit Cancer Center
Charles Stark Draper Laboratory Inc
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 Moffit Cancer Center, Charles Stark Draper Laboratory Inc filed Critical Moffit Cancer Center
Priority to US14/070,444 priority Critical patent/US20140127733A1/en
Assigned to THE CHARLES STARK DRAPER LABORATORY, INC. reassignment THE CHARLES STARK DRAPER LABORATORY, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPENCER, ABIGAIL, HOLTON, Angela, LANDIS, DAVID, EPSHTEYN, ALLA
Publication of US20140127733A1 publication Critical patent/US20140127733A1/en
Priority to US14/586,577 priority patent/US9879308B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • 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
    • C12M23/02Form or structure of the vessel
    • C12M23/16Microfluidic devices; Capillary tubes
    • 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
    • B01L3/502761Containers 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 specially adapted for handling suspended solids or molecules independently from the bulk fluid flow, e.g. for trapping or sorting beads, for physically stretching molecules
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • 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
    • B01L2200/0647Handling flowable solids, e.g. microscopic beads, cells, particles
    • B01L2200/0668Trapping microscopic beads
    • 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
    • B01L2400/08Regulating or influencing the flow resistance
    • B01L2400/084Passive control of flow resistance
    • B01L2400/086Passive control of flow resistance using baffles or other fixed flow obstructions
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2510/00Detection of programmed cell death, i.e. apoptosis

Definitions

  • This invention relates to culturing and monitoring a biological sample.
  • a fluidic apparatus includes a substrate, a channel located within the substrate, and a concave retaining barrier located within the channel.
  • the channel is configured to receive a biological sample and to carry a fluid flow across the biological sample.
  • the concave retaining barrier is configured to retain the biological sample.
  • the geometry of the concave retaining barrier is configured to allow the fluid flowing through the channel to induce interstitial flow of the fluid through the biological sample as to perfuse the biological sample with the fluid while maintaining shear rates at the biological sample within a range of between about 0.375 and 0.500 dynes/cm 2 .
  • the concave retaining barrier includes a plurality of posts extending from the floor of the channel to the top of the channel.
  • the plurality of posts includes between three to ten free-standing posts, which extend the full height of the channel and at least one embedded post extending out from a channel sidewall.
  • the concave retaining barrier includes five free-standing posts and two embedded posts.
  • the adjacent posts are spaced apart from one another to have a lateral separation across of the between about 20 ⁇ m-about 30 ⁇ m. In some implementations, the shortest distance between any two adjacent posts is between about 65 ⁇ m-85 ⁇ m. In some implementations, the outer most free-standing post of the plurality of posts is spaced between about 65 ⁇ m-85 ⁇ m away from a nearest channel sidewall.
  • the fluidic apparatus also includes a fluid supply configured to flow a fluid through the channel.
  • the fluid supply includes a closed loop fluid supply configured to reroute fluid already flowed through a distal end of the channel back through a proximal end of the channel.
  • the fluidic apparatus also includes a fluid extractor configured to extract a sample of the fluid after it has flowed through to channel.
  • the fluidic apparatus also includes an optical sensor for optically monitoring a state of a biological sample located within the retaining barrier.
  • the biological sample is between about 100 and about 500 microns in diameter.
  • a method of assessing the impact of a pharmacological agent on an ex vivo tissue sample includes introducing a tissue sample having a diameter of between about 100-500 microns into a concave retaining barrier formed in a flow channel, introducing a fluid including the pharmacological agent to the channel such that the fluid flows through the channel across the tissue sample, collecting a portion of the fluid, and analyzing the collected fluid portion for evidence of pharmacological impact on the tissue sample.
  • the channel and concave retaining barrier are configured to induce interstitial flow of the fluid through the tissue sample, such that the fluid perfuses through the tissue sample.
  • the evidence of pharmacological impact includes evidence of cell death.
  • the method also includes capturing an image of the tissue sample after flowing the fluid through the channel and analyzing the captured image for evidence of pharmacological impact on the tissue sample.
  • the fluid flows through the channel at a rate of about 500 microliters per hour.
  • the tissue sample includes a tissue sample collected using fine-needle biopsy (FNBA).
  • the concave retaining barrier includes a plurality of posts extending from the floor of the channel to the top of the channel.
  • the plurality of posts includes between three to ten free-standing posts extending the full height of the channel and at least one embedded post extending out from a channel sidewall.
  • the method includes introducing a second tissue sample having a diameter of between about 100-500 microns into a second concave retaining barrier formed in a second flow channel, introducing a second fluid including a second pharmacological agent to the channel such that the fluid flows through the channel across the second tissue sample, collecting a portion of the second fluid, and analyzing the collected fluid portion for evidence of pharmacological impact on the second tissue sample, and comparing the pharmacological impacts of the first and second pharmacological agents.
  • the selection of the first or second pharmacological agents for treatment of an animal is based on the comparison of the pharmacological impacts.
  • the animal is a source of the first and second tissue samples.
  • FIG. 1A is a diagram showing an exemplification of a micro-scale microfluidic apparatus and its retaining element.
  • FIG. 1B is a diagram showing an exemplification of a channel.
  • FIG. 1C is a diagram showing an exemplification of dimensions of a free standing post.
  • FIG. 1D is a diagram showing an exemplification of a retaining element.
  • FIG. 1E is a diagram of a close-looped microfluidic system.
  • FIG. 1F is a diagram of a microfluidic system.
  • FIG. 2A is a perspective view of an example fluidic apparatus and biological sample.
  • FIG. 2B is a perspective view of an example microfluidic systems device.
  • FIG. 2C is a view of a biological sample collected using fine needle aspirate biopsy retained within an example fluidic apparatus.
  • FIG. 3 is a model of the fluid dynamics of an example fluidics apparatus.
  • FIG. 4 is a table including example tumor samples subjected to a florescence stain.
  • FIG. 5 is a flow chart showing an exemplary method for assaying a biological sample.
  • FIG. 6 is a flow chart showing an exemplary method for determining the personalized treatment for a patient.
  • FIG. 7A is a table illustrating the effect of MK-1775 and gemcitabine in a xenograft mouse model in vivo.
  • FIG. 7B is a table illustrating percent change of initial tumor volume as a function of time.
  • FIG. 7C provides images of tumor response in a microfluidic apparatus ex vivo.
  • FIG. 8 provides an expanded view of an extracted ion chromatogram.
  • the present disclosure includes a cost effective method that monitors patient driven pharmacologic and metabolic responses in an ex vivo micro-scale fluidic apparatus utilizing fine-needle aspiration biopsy (FNAB) samples or other tissue samples through which a test agent can perfuse.
  • FNAB fine-needle aspiration biopsy
  • the present technology is directed to a standardized tumor microenvironment model to individualize cancer therapy selection using a microfluidics assay susbtrate that utilizes microscopic tumor tissue obtained by fine needle aspiration biopsy (FNAB) or other extraction method to analyze tumor response to conventional and targeted anti-cancer drugs.
  • FNAB fine-needle aspiration biopsy
  • Biomolecular sensors for high throughput real-time monitoring of drug response are optionally integrated with this platform. While the disclosure focuses on a single channel apparatus for ease of explanation, other apparatuses having multiple channels for high throughput multi-drug analysis at various drug concentrations can also be used. The apparatuses can also be used for multi-tissue interactions within a complex system of pharmacodynamic/pharmacokinetic/proteomic/metabolomic and for genomic processes that are influenced by multiple organ systems in vivo to generate a reproducible, repeatable clinical model for patient specific responses to trials of chemotherapies.
  • the fluidic apparatus may include a substrate, a channel located within the substrate, a retaining element located within the channel for retaining the biological sample, a supply apparatus for supplying a fluid to the channel, a retrieval apparatus for retrieving fluid from the channel, and a visual field for providing visual observation of the channel.
  • a three-dimensional (3-D) micro-scale fluidics apparatus is provided in FIGS. 1A-D .
  • FIG. 1A is a diagram of a micro-scale fluidics apparatus 100 with five channels 101 .
  • the micro-scale fluidics substrate has one channel, or two channels, or three channels, or four channel, or five channels, or six channels.
  • the micro-scale fluidics substrate has up to or more than ten channels.
  • a high-throughput micro-scale fluidics may be produced containing a hundred or more channels.
  • the biological sample inserted into each channel can be the same for each channel, different for each channel, or a combination thereof.
  • FIG. 1B is a diagram of a single channel 101 .
  • the channel 101 is about 10 mm in length (l) and about 615 ⁇ m in width (w). In various implementations, the channel 101 is between about 5 mm to about 20 mm long, between about 400 ⁇ m to about 1000 ⁇ m wide, and between about 75 ⁇ m to about 500 ⁇ m high.
  • the channel 101 has a fluid inlet 105 , a fluid outlet 104 , and a concave retaining barrier 106 .
  • the concave retaining barrier 106 in the channel 101 includes five free standing posts 102 and a pair of embedded posts 103 in the sidewalls of the channel 101 .
  • the free standing posts 102 and embedded posts 103 are arranged in a V-configuration that points in the direction of the flow of fluid through the channel 101 .
  • the concave retaining barrier 106 is used to hold a biological sample in place.
  • the concave retaining barrier 106 includes more than five free standing posts.
  • the free standing posts 102 of the concave retaining barrier 106 are configured in a semi-circle formation, U-shape configuration, or any other shape concave with respect to the direction of flow through the channel 101 .
  • the multiple concave retaining barriers are lined up along the length of the channel creating a series of concave retaining barriers.
  • the multiple concave retaining barriers are lined up across the width of the channel forming a line of concave retaining barriers.
  • each of the concave retaining barriers 106 in a channel 101 are identical.
  • the retaining elements are formed from posts having different dimensions and spacing to trap tissue samples of different sizes. For example, the posts and paths between the posts may shrink progressively down the length of the channel to capture progressively smaller tissue samples.
  • the free standing posts 102 are about 150 ⁇ m long 203 , about 75 ⁇ m wide 204 , and about 125 ⁇ m high 107 .
  • the free standing posts are between about 50 ⁇ m to about 200 ⁇ m long, between about 50 ⁇ m to about 100 ⁇ m wide, and between about 50 ⁇ m to about 500 ⁇ m high.
  • the embedded posts 103 are about 150 ⁇ m long and about 125 ⁇ m high.
  • the embedded posts are between about 25 ⁇ m to about 175 ⁇ m long and between about 100 ⁇ m to about 150 ⁇ m high.
  • the free standing posts 102 have between about a 2:1 to about a 3:1 length to width ratio.
  • the free standing posts 102 and/or the embedded posts 103 are as high as the channel 101 .
  • the height of the channel has up to about a 1:2 ratio compared to the diameter of the biological sample.
  • FIG. 1D is a diagram showing exemplary spacing of the embedded posts 103 A and 103 B and free standing posts 102 A- 102 E of the concave retaining barrier 106 .
  • the free standing posts 102 and embedded posts 103 are aligned such that the leading ends of complimentary posts are aligned, e.g., the leading ends of 102 A and 102 E are aligned.
  • the leading edge of one free standing post is aligned with the trailing edge of the next free standing post in the channel 101 , e.g., the leading edge of 102 B may be aligned with the trailing edge of 102 C.
  • the distance 205 between the sidewalls of the freestanding posts 102 A and 102 E closest to the channel sidewall is about 70 ⁇ m. In some implementations, the distance 108 between the sidewalls of the freestanding posts 102 A and 102 E closest to the channel sidewall is between about 60 ⁇ m to about 80 ⁇ m.
  • the diagonal distance 110 between the closest points of the freestanding posts 102 A and 102 E closest to the embedded posts 103 A and 103 B is about 75 ⁇ m. In some other implementations, the diagonal distance 110 between the closest points of the freestanding posts 102 A and 102 E closest to the embedded posts 103 A and 103 B is between about 65 ⁇ m to about 85 ⁇ m.
  • the lateral spacing 112 between the outer walls of free standing posts 102 A and 102 E and the outer walls of embedded posts 103 A and 103 B is about 25 ⁇ m. In some implementations, the lateral distance 112 between outer walls of free standing posts and the outer walls of embedded posts is between about 20 ⁇ m to 40 ⁇ m.
  • the lateral spacing 111 between the walls of adjacent free standing posts is about 25 ⁇ m. In some implementations, the lateral distance 111 between the outer walls of adjacent free standing posts is between about 20 ⁇ m to 40 ⁇ m.
  • the diagonal distance 109 between the closest points of two adjacent free standing posts e.g., 102 A to 102 B, 102 B to 102 C, 102 C to 102 D, and 102 D to 102 E, is about 75 ⁇ m. In some other implementations, the diagonal distance 109 between the closest points of two adjacent free standing posts is between about 65 ⁇ m to 85 ⁇ m.
  • the geometry of the channel 101 and the spacing of the components of the concave retaining barrier 106 are selected for their beneficial characteristics with relation to culturing tissue samples having sizes on the order of 300 ⁇ m in diameter and smaller.
  • the post sizing and spacing provides for fluid dynamics, including flow rate, shear rate, and pressure, that approximate in vivo conditions.
  • culturing the biological sample in the channel environment allows for the biological sample to remain intact without the triggering of shear related cellular responses.
  • the flow rate is about 100 ⁇ l/hr, or about 250 ⁇ l/hr, or about 500 ⁇ l/hr, or about 1000 ⁇ l/hr, or ranges between any two of these values.
  • the geometry and flow rate are selected to achieve shear stress on the sample of about 0.350, or about 0.375, or about 0.400, or about 0.425, or about 0.450, or about 0.500 dyne/cm 2 or any value between any two of these values, which are physiologically relevant and which do not pose a significant disturbance to the system or sample.
  • the geometry and flow rates are selected to provide a shear rate of between about 0.375 and 0.500 dyne/cm 2 .
  • the flow rate and geometry are selected to provide a shear rate of about 0.389 dyne/cm 2 .
  • the flow and shear rates within the channel 101 are affected by the width of the channel 202 and the spacing of the free standing posts 102 and embedded posts 103 .
  • a wide channel reduces interstitial flow to the biological sample, as the fluid is able to move around the sample, thus reducing contact and perfusion.
  • a narrow channel can increase shear stress, which can lead to shear-induced activation of gene expression. Shear-induced activation of gene expression can lead to a biochemical adhesion cascade in the biological sample and tissue transformation. Additionally, some studies have shown that in changing the shear stress a tissue experiences over time the extracellular matrix proteins become altered changing the tumors potential for cell invasion, angiogenesis and tissue remodeling. Mechanotransduction induced changes in the biological sample can have adverse effects on the assaying of the biological sample. Each of these conditions is preferably avoided to effectively replicate the tissue response in vivo to the agent being tested as a snapshot in time of the patient's cancer.
  • the embedded posts 103 also help maintain improved flow parameters.
  • a channel 101 without embedded posts 103 i.e., in which the channel walls are smooth and spaced apart from the nearest free standing post 102 , decreases interstitial flow throughout the biological sample and drug perfusion as the space between the two outer most free standing posts 102 and the channel side wall provides a substantial path, which allows the fluid to flow around the biological sample.
  • using conventional micromachining and injection molding processes, which would be applied for high volume manufacture maintaining a narrow enough gap to obtain a desirable flow between the outer most free standing posts 102 and a smooth channel wall is impractical, if not impossible.
  • the use of the embedded posts 103 substantially mitigates this issue.
  • the biological samples are tumor or tissue samples.
  • the biological samples are micron-scale samples.
  • the biological samples are between about 100 ⁇ m and about 500 ⁇ m in diameter, and in some implementations less than about 300 ⁇ m.
  • the biological samples are fine needle aspirate biopsy samples (FNABs).
  • the fluidic apparatus can be produced methods known in the art.
  • methods for producing the fluidic apparatus include injection molding, micro-machining, heat and/or pressure embossing, and photolithography.
  • the fluidic apparatus is substantially smooth along all surfaces, e.g., the bottom and sides of the flow channel, the free standing posts, and the embedded posts. This helps prevent vortices and eddy currents in the flow, as well as prevents undesirable cellular interaction with the surfaces of the channel and concave retaining barrier.
  • the fluidic apparatus can be fabricated from known materials in the art.
  • the fluidic apparatus is transparent, allowing for real-time visual analysis of the biological sample.
  • the materials selected for the fluidic apparatus is substantially transparent (i.e., greater than 50%) to light in the visible portion of the spectrum and down to wavelengths of about 200 nm or smaller.
  • the fluidic apparatus is made of optical glass or glass-like material.
  • Exemplary optical glass materials include, but are not limited to, N-FK51A, N-PK52A, N-PK51, N-FK5, P-BK7, P-SK58A, P-SK57, P-SK57Q1, P-SK60, N-KZFS2, P-LAK35, P-LAF37, N-KZFS4, N-KZFS4HT, N-LAF33, N-KZFS11, P-LASF51, P-LASF47, P-LASF50, N-KZFS5, N-KZFS8, N-LASF46B, P-SF8, P-SF69, P-SF67, and P-SF68 (available from Schott, Inc).
  • the fluidic apparatus includes a cover forming a top to the channel 101 .
  • the cover is made from the same material as the fluidic apparatus.
  • the fluidic apparatus is made from polymers.
  • exemplary polymers include, but are not limited to, polymethyl methacrylate (PMMA), polyamide (PA), polycarbonate, polystyrene, polydimethylsiloxane (PDMS), and cyclic olefin copolymer (COC).
  • FIGS. 1E and 1F are diagrams showing exemplary microfluidic systems of the present disclosure for the analysis of ex vivo treatment of biological samples with drugs or therapeutic agents.
  • FIG. 1E is an exemplary continuous closed-loop system 200 .
  • a supply apparatus 201 pumps fluid to the channel (or channels) of the fluidic apparatus 100 through a single fluid inlet (or multiple fluid inlets) and re-cycles the fluid received from the fluid outlet (or multiple fluid outlets), such that the biological sample is continuously perfused with the fluid.
  • the sample may be continuously perfused in a closed loop over a period of 7 days to determine the lifetime of FNAB samples in 3-D microenvironment platform.
  • the sample is continuously perfused in a closed loop over a period of 12-24 hours, or 12-36 hours, or 12-48 hours, or 24-72 hours.
  • FIG. 1F is another exemplary continuous closed-loop system 250 .
  • a supply apparatus 201 pumps fluid to the channel (or channels) of the fluidic apparatus 100 through a single fluid inlet (or multiple fluid inlets) such that the biological sample is continuously perfused with the fluid.
  • a retrieval apparatus 203 collects the fluid that has passed over/through the biological sample. In some implementations, the retrieval apparatus 203 provides the fluid to an analysis apparatus 204 to determine any pharmacologic and a metabolic responses associated with the sample and/or the fluid. In some implementations, the retrieval apparatus only collects a sample of the fluid and returns the remainder to the supply apparatus to provide a closed-loop fluidic system.
  • the microfluidic systems of FIGS. 1E or 1 F can also include a visualization apparatus 202 , which can be configured to provide for visual observation using, for example, an inverted tissue culture microscope.
  • a visualization apparatus 202 can be configured to provide for visual observation using, for example, an inverted tissue culture microscope.
  • a Zeiss Axio Observer with ApoTome technology may be used for gross morphological measurements to determine cell growth and death in the microfluidics channels and resulting architecture and presence of differentiation.
  • the visualization apparatus 202 may also include image sensors for capturing optical information from the sample forw wavelengths of light ranging from the visible spectrum down to about 200 nm.
  • FIGS. 2A and 2B show illustrative views of additional examples of a micro-scale fluidic apparatus for culturing and monitoring, ex vivo, pharmacologic and metabolic response in a biological sample.
  • the apparatus may include a substrate, a channel located within the substrate for receiving the biological sample, a retaining element located within the channel for retaining the biological sample, a supply apparatus for supplying a fluid to the channel, a retrieval apparatus for retrieving fluid from the channel, and a visual field for providing visual observation of the channel.
  • a substrate a substrate, a channel located within the substrate for receiving the biological sample, a retaining element located within the channel for retaining the biological sample, a supply apparatus for supplying a fluid to the channel, a retrieval apparatus for retrieving fluid from the channel, and a visual field for providing visual observation of the channel.
  • the substrate may be fabricated from a polydimethylsiloxane (PDMS) with PDMS posts (or any of the other material identified above) on a glass substrate or other transparent cover slip for ease of microscopy.
  • PDMS polydimethylsiloxane
  • An example biological sample may be collected by FNAB using a 23-27 gauge needles, which typically provides a microscopic tumor fragments in the size of less than 500 micrometer ( ⁇ m), and in some cases less than 300 ⁇ m, inner diameter in a 3-D structure.
  • An example biological sample of a tumor cell may include tumor cells that intimately interact with the stromal components such as fibroblasts, endothelial cells, and immune cells.
  • a biological sample can include one or more cell that is metabolically active or expected to be metabolically active.
  • the one or more cell may also include a tumor cell.
  • the one or more cell may also include a non-tumor cell.
  • the one or more cell may also include a plurality of cells including at least one tumor cell and at least one non tumor cell.
  • the channel may be sized and configured to provide for the culture of a biological sample retrieved using fine-needle aspiration.
  • the channels in the devices shown in FIGS. 2A and 2B are about 550 ⁇ m wide by about 120 ⁇ m in height and about 1 cm in length.
  • the channels are sized and configured to provide for the growth of a three-dimensional cell culture of the biological sample.
  • the channels may be sized and configured to provide for growth of the biological sample on a multi-channel 3-D microfluidic cell culture chip.
  • the substrate shown in FIG. 2B includes a second channel for receiving a second biological sample.
  • the second biological sample may be from the same, or different, tumor as included in the first channel.
  • a second supply apparatus may supply a second fluid to the second channel.
  • the second fluid may include the same, or different, fluid provided to the first sample in the first channel.
  • the channels include concave retaining barriers, similar to the concave retaining barriers shown in FIGS. 1B-1D , located within the channel for retaining the biological samples.
  • the retaining element may include a series of posts extending up from the floor of the channel.
  • FIG. 2C is a micrograph of an example of a concave retaining barrier holding a tissue sample.
  • the concave retaining barrier includes a series of posts in a V-shaped configuration pointed in the direction of the fluid flow.
  • the posts in the example, measure about 25 ⁇ m wide and about 100 ⁇ m in height, with about 25 ⁇ m lateral distance between posts.
  • FIG. 3 shows a model of the fluid dynamics of the channel incorporated into the fluidic apparatus shown in FIG. 2A .
  • the model depicts both the fluid velocity and pressure assuming a flow rate 100 ⁇ l/hour.
  • a biological sample retrieved from the patient is provided to the fluidic apparatus.
  • the biological sample may be retrieved using fine-needle aspiration and include a sample that is about or less than about 500 ⁇ m (or in some implementations 300 ⁇ m) in diameter.
  • An example sample includes at least one of a tumor cell and a stroma cell.
  • the culture of the biological sample is provided in the channel of the fluidic apparatus.
  • the culture may be a three-dimensional cell culture.
  • the cells may be culture a multi-channel 3-D microfluidic cell culture chip.
  • a fluid is supplied to the biological sample inside the channel.
  • the fluid may include, for example, a culture medium, a vehicle, and a drug.
  • the fluid may be supplied by a continuous closed-loop system.
  • the sample can be observed at a visual field provided on the fluidic apparatus.
  • the culture and the sample may be observed at the visual field using an inverted tissue culture microscope.
  • a biomolecular sensor may be applied to the sample.
  • a florescence stain may be applied to the sample.
  • fluorescence-based assays such as LIVE/DEAD® assays (Invitrogen, Carlsbad, Calif.) may be used to evaluate cell death and viability.
  • the Hoechst stain demonstrates nuclear structures and the compressed view shows the phase contrast views of the tumor FNAB samples in the microfluidic apparatus. Using this data, characteristic cell death metabolic profiles can be correlated along with microscopic analysis to indicate drug sensitivity by analyzing cultured media perfused through apparatus in contact with tumor. Metabolic analysis also includes methods for control versus drug response in the example microfluidic apparatus.
  • the methods, apparatuses, and systems described herein are may also be used to predict patient tumor response to chemotherapy and analyzes drug processing in tumor and other tissue ex vivo.
  • the present system can be used for drug development, biomarker discovery and personalized therapy for individual patients to identify the most effective drug/drug combinations in a short period of time.
  • the methods, apparatuses, and systems may include flowing a fluid past the biological sample having at least one viable cell or cell that is expected to be viable, retrieving the fluid, and analyzing the fluid to determine at least one of a pharmacologic and a metabolic response of the sample.
  • a pharmacologic response can be, for example, caused by flowing a fluid comprising a pharmacologic agent over the biologic sample.
  • the response of the sample to the agent can be determined by analyzing the fluid for compounds, factors, or the like produced or elicited by a cell of the biologic sample by its interaction with the pharmacologic agent.
  • the response can also be determined by visual analysis of the biologic sample, for example under the microscope, alone or in combination with detection of the fluid compounds, factors, or the like.
  • a pharmacologic agent is a chemotherapeutic agent.
  • a metabolic response can be, for example, caused by flowing a fluid comprising an agent that causes a metabolic response in the biologic sample over the biologic sample.
  • the response of the sample to the agent can be determined by analyzing the fluid for compounds, factors, metabolic products, or the like produced or elicited by a cell of the biologic sample by its interaction with the agent.
  • the response can also be determined by visual analysis of the biologic sample alone or in combination with detection of the fluid compounds, factors, or the like.
  • An example, metabolic agent is glucose. Alternatively, or additionally, the analysis of the fluid indicates that the cell or cells of the sample are not viable.
  • FIG. 5 is flowchart showing a non-limiting, exemplary method 300 for analyzing a biological sample using the disclosed microfluidic apparatus.
  • a biological sample is obtained (step 301 ).
  • the biological sample is a tissue sample or tumor sample on the order of about 300 ⁇ m in diameter or smaller.
  • the biological sample is obtained from FNAB or other extraction methods suitable for obtaining similarly sized tissue or tumor samples.
  • the biological sample is inserted into the concave retaining barrier (step 302 ) located in a channel of a fluidic apparatus, such as the concave retaining barrier shown in FIG. 1C .
  • the biological sample is exposed to a flowing fluid (step 303 ).
  • the fluid contains a drug or a compound.
  • the biological sample is subject to continuous fluid flow in a closed loop for about 2-4 hours, about 4-8 hours, about 8-12 hours, about 12-24 hours, about 12-36 hour, or about 12-48 hours, or about 24-72 hours.
  • the flow fluid is collected and analyzed (step 304 ).
  • the flow fluid is analyzed for metabolites or other cellular byproducts related to drug or compound treatment.
  • the flow fluid is collected about every 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, or 48 hours.
  • the biological sample is subjected to visual analysis (step 305 ). Analysis of the flow fluid and visual analysis of the biological sample are used to determine the efficacy of the drug or compound (step 306 ).
  • An example system includes a three-dimensional (3-D) microfluidics platform for short-term culture of tumor and stroma cells obtained from FNAB, with the goal of using these for drug sensitivity testing by assessing full tumor response to chemotherapy by incorporating microscopic, pharmacodynamic, pharmacokinetic, proteomic, metabolomic and nucleic acid (including but not limited to DNA, mRNA and miRNA) analysis of tumor microenvironment and perfused media.
  • This FNAB microfluidics example system allows analysis of normal and disease-state tissue to drugs as a single tissue fragment or in a multi-tissue system on a chip format.
  • This platform can be used for: drug development and drug testing for efficacy; biomarker discovery and validation in micofluidics; personalized therapy whereby tumor cells/tissue from individual patients can be tested for the most effective drug/drug combinations in a short period of time; personalized tumor microenvironment pharmacokinetic studies; and toxicology assays using liver and other non-tumoral tissue fragments.
  • FIG. 6 is a flow chart 400 showing how potential therapeutic agents are identified.
  • one or more biological samples are obtained from a patient (step 401 ).
  • the biological samples are then inserted into the retaining barriers of different channels in a fluidic apparatus (step 402 ).
  • the channels are then subjected to fluid flow (step 403 ).
  • the fluid in each channel contains a different therapeutic agent and/or different concentrations of the same therapeutic agent.
  • the a fluid can have one or more therapeutic agents.
  • the biological sample is subject to continuous fluid flow in a closed loop for about 2-4 hours, about 4-8 hours, about 8-12 hours, about 12-24 hours, about 12-36 hour, or about 12-48 hours, or about 24-72 hours.
  • the flow fluid is collected and analyzed (step 404 ).
  • the flow fluid is analyzed for metabolites or other cellular byproducts related to the therapeutic agent or combination of agents.
  • the flow fluid is collected about every 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, or 48 hours.
  • the biological sample is subjected to visual analysis (step 405 ).
  • a microscope or other optical sensor is used for visual and/or optical analysis. Analysis of the flow fluid and visual analysis of the biological sample are used to determine the efficacy of each therapeutic agent or combination of agents (step 406 ). Based on the results, a therapeutic course of treatment tailored specifically for the patient is determined (step 406 ).
  • a micro-scale fluidic apparatus was used to investigate the response of fine needle biopsy samples (FNBA) of osteosarcoma to Gemcitabine and MK-1775, either alone or in combination for their effects in cell death and tumor shrinkage.
  • FNBA fine needle biopsy samples
  • FIGS. 2A and 2C The structure of a fluidic apparatus is shown in FIGS. 2A and 2C .
  • Each fluidic apparatus included a single channel, and the center of the channel contained a series of seven free standing post, collectively called the retaining element, which were used to retain the FNBA osteosarcoma sample (see FIG. 3A ). Fluid flows into the channel by an inlet port and out of the channel by an outlet port.
  • FNAB osteosarcoma samples were taken from xenograft mouse with patient-derived osteosarcoma using a 25-gauge needle. FNABs were processed and selected for viability by H&E staining procedures before use and individually loaded one into each retaining element and imaged by microscopy and measured for size (all FNABs assayed were ⁇ 300 ⁇ m in diameter).
  • FNAB samples were divided and treated as follows: two FNAB samples were treated with media only (control) (RPMI with 10% FBS, 300 mg/L L-glutamine, 2000 mg/L D-glucose, 2000 mg/L sodium bicarbonate); two FNAB samples were treated with 1 ⁇ M MK-1775 (SelleckChem, Houston, Tex.), a Wee 1 inhibitor, in cell culture media; two FNAB samples were treated with 3 ⁇ M gemcitabine (Eli Lilly, Indianapolis, Ind.), anti-cancer drug, in cell culture media; and two FNAB samples were treated with a combination of 1 ⁇ M MK-1775 and 3 ⁇ M gemcitabine in cell culture media.
  • the fluidic apparatuses were connected to their respective syringe pumps.
  • the FNAB samples were first primed with several passes of culture media. After priming, each FNAB sample was subject to constant laminar flow at 100 ⁇ l/hr for 72 hours with their respective treatment. Media was collected every 24 hours and analyzed for caspase 3 and 7 and lactate dehydrogenase (LDH).
  • LDH lactate dehydrogenase
  • each FNAB sample was microscopically visualized with Hoechst stain for nuclear counter stains by intercalating DNA and Sytox Green for cell death.
  • Xenograft mice bearing human osteosarcoma tumor were treated with vehicle, gemcitabine, MK-1775, or a combination of gemcitabine and MK-1775 for 15 days. Treatments were administered on day 1, 3, 8, and 10. FNAB samples were taken before (day 0) and at 6 and 24 hours after treatment. Cell extracts were prepared from the FNAB samples to assess expression levels of CDC2Y15, a target for MK-1775, by Western blot. After day 15, tumors were analyzed by T 2 -weighted MRIs and paraffin sections of tumors were prepared to assess microscopic features of cell death by H&E-staining and to assess levels of cleaved-caspase 3 .
  • FIGS. 7A-B show the effect of MK-1775, gemcitabine, or combination of MK-1775 and gemcitabine in a xenograft mouse model in vivo.
  • FIG. 7C shows the effect of MK-1775, gemcitabine, or combination of MK-1775 and gemcitabine using the fluidic apparatus model ex vivo.
  • FIG. 7A shows that MK-1775 and its combination with an anti-cancer drug gemcitabine lead to cell death in a patient-derived osteosarcoma xenograft mouse model in vivo.
  • Column (a) shows that in vivo treatment of osteosarcoma xenograft mice with MK-1775 and combination treatment reduces expression levels of CDC2Y15 as compared to vehicle and gemcitabine treatments.
  • Column (b) shows reduced tumor volume at day 15 in MK- 1775-treated and combination-treated animals as compared to vehicle and gemcitabine treatments.
  • Column (c) shows that MK-1775 and combination-treated animals have decreased DNA staining and high immunoreactivity to caspase 3 as compared to vehicle and gemcitabine treatments.
  • FIG. 7B shows that the percent change from the initial tumor volume (i.e., day 0) as a function of time, displaying the significant increase in tumor growth from day 0 to 15 in the control group versus the other treatment groups.
  • the symbol (*) indicates statistical significance (p ⁇ 0.05).
  • FIG. 7C shows the tumor response in microfluidic apparatus ex vivo.
  • the results show that MK-1775 and its combination with gemcitabine lead to cell death in tumor fragments ex vivo.
  • the FNAB samples were fully penetrated by drugs. Tumor cells were stained with HCS LIVE/DEAD® Green Kit (Life Technologies, Carlsband, Calif.) and images were obtained using inverted fluorescence microscope to assess dead (green) and alive (blue) cells.
  • results show that the ex vivo treatment of osteosarcoma tumor samples with potential therapeutic agents using the fluidic apparatus produced similar results as in vivo treatment of osteosarcoma xenograph mice. Both models were able to identify that MK-1775 treatment or the combination of MK-1775 and gemcitabine treatment were able to induce apoptosis of the cancer cells. However, the ex vivo treatment was able to identify the efficacy of the agent or agents in a 72 hour period versus the 15 days of treatment the in vivo model require (the 15 days does not account for the time required for the nude mice to grow xenograft tumors).
  • FIG. 8 provides an expanded view of an extracted ion chromatogram (XIC) from a potential metabolite highlighted against the background sample.
  • Perfused fluid was collected from the fluidic apparatus as described in the above experiment performed with the FNAB samples ( FIGS. 2A-C ) to determine whether tumor-mediated drug processing and metabolism can be analyzed. Fluidic samples were prepared by filtration and analyzed by mass spectrometry.
  • FIG. 8 demonstrates that tumor fragments obtained by FNAB from a patient-derived osteosarcoma xenograft model can incorporate drugs and convert the drugs into active and metabolized forms.
  • Mass spectrometry data shows active metabolism of gemcitabine into its active form, triphosphate, and metabolism of MK-1775 into its metabolized forms.
US14/070,444 2012-11-01 2013-11-01 Ex vivo microfluidic analysis of biologic samples Abandoned US20140127733A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/070,444 US20140127733A1 (en) 2012-11-01 2013-11-01 Ex vivo microfluidic analysis of biologic samples
US14/586,577 US9879308B2 (en) 2012-11-01 2014-12-30 Apparatus for ex vivo microfluidic analysis of biologic samples

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261721183P 2012-11-01 2012-11-01
US14/070,444 US20140127733A1 (en) 2012-11-01 2013-11-01 Ex vivo microfluidic analysis of biologic samples

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/586,577 Division US9879308B2 (en) 2012-11-01 2014-12-30 Apparatus for ex vivo microfluidic analysis of biologic samples

Publications (1)

Publication Number Publication Date
US20140127733A1 true US20140127733A1 (en) 2014-05-08

Family

ID=49585625

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/070,444 Abandoned US20140127733A1 (en) 2012-11-01 2013-11-01 Ex vivo microfluidic analysis of biologic samples
US14/586,577 Active 2035-03-09 US9879308B2 (en) 2012-11-01 2014-12-30 Apparatus for ex vivo microfluidic analysis of biologic samples

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/586,577 Active 2035-03-09 US9879308B2 (en) 2012-11-01 2014-12-30 Apparatus for ex vivo microfluidic analysis of biologic samples

Country Status (4)

Country Link
US (2) US20140127733A1 (ja)
EP (1) EP2914379A1 (ja)
JP (1) JP2015536141A (ja)
WO (1) WO2014071255A1 (ja)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018187181A1 (en) * 2017-04-03 2018-10-11 The Charles Stark Draper Laboratory, Inc. Microfluidic system for evaluation of chemotherapeutic and immunotherapeutic drugs
US10254280B2 (en) * 2015-08-10 2019-04-09 Micronit Microtechnologies B.V. Channel for trapping particles to be fed to said channel with a fluid
US20210162416A1 (en) * 2019-12-02 2021-06-03 The Charles Stark Draper Laboratory Inc. Multiwell dynamic model for a tumor-immune microenvironment
WO2021185873A1 (en) * 2020-03-16 2021-09-23 Imperial College Innovations Limited Device for perfusion and preservation of tissue specimens ex vivo

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109097243A (zh) * 2017-06-21 2018-12-28 曦医生技股份有限公司 生物微粒捕捉芯片组
US11760966B2 (en) 2019-06-14 2023-09-19 University Of Connecticut Multigel tumor-on-a-chip system

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6942873B2 (en) * 2000-09-25 2005-09-13 The Board Of Trustees Of The University Of Illinois Microfabrication of membranes containing projections and grooves for growing cells
US20110086427A1 (en) * 2009-10-12 2011-04-14 Faris Ronald A Microfluidic device for cell culture
US20110256574A1 (en) * 2008-08-08 2011-10-20 Agency For Science, Technology And Research Microfluidic Continuous Flow Device

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6001307A (en) * 1996-04-26 1999-12-14 Kyoto Daiichi Kagaku Co., Ltd. Device for analyzing a sample
WO2004051231A1 (ja) * 2002-11-29 2004-06-17 Nec Corporation 分離装置および分離方法
WO2004101743A2 (en) * 2003-05-06 2004-11-25 Bellbrook Labs, Llc Three dimensional cell cultures in a microscale fluid handling system
US20090136982A1 (en) * 2005-01-18 2009-05-28 Biocept, Inc. Cell separation using microchannel having patterned posts
US8389277B2 (en) * 2007-10-11 2013-03-05 Agency For Science, Technology And Research Forming cell structure with transient linker in cage
US20110136162A1 (en) * 2009-08-31 2011-06-09 Drexel University Compositions and Methods for Functionalized Patterning of Tissue Engineering Substrates Including Bioprinting Cell-Laden Constructs for Multicompartment Tissue Chambers
US9261496B2 (en) 2010-09-29 2016-02-16 Massachusetts Institute Of Technology Device for high throughput investigations of multi-cellular interactions
CN106902903B (zh) * 2011-02-15 2020-03-24 海默索尼克斯有限公司 用于评估止血的装置、系统和方法
EP3498818A1 (en) * 2011-02-28 2019-06-19 President and Fellows of Harvard College Cell culture system
US9074204B2 (en) * 2011-05-20 2015-07-07 Fluidigm Corporation Nucleic acid encoding reactions
EP3722001A1 (en) * 2013-11-22 2020-10-14 The General Hospital Corporation Microfluidic methods and systems for isolating particle clusters
US20160018365A1 (en) * 2014-06-13 2016-01-21 Masoud Agah Functionalized Metal Oxides As A Stationary Phase And A Surface Template For Micro Gas Chromatography Separation Columns

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6942873B2 (en) * 2000-09-25 2005-09-13 The Board Of Trustees Of The University Of Illinois Microfabrication of membranes containing projections and grooves for growing cells
US20110256574A1 (en) * 2008-08-08 2011-10-20 Agency For Science, Technology And Research Microfluidic Continuous Flow Device
US20110086427A1 (en) * 2009-10-12 2011-04-14 Faris Ronald A Microfluidic device for cell culture

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FNBA. Procedures/diagnostic tests: interventional radiology. Clinical Center National Institutes of Health. 2010;1-5. *
Kim et al. In-situ synthesized and patterned nanowire arrays in microfluidic channel for particle trapping and cell lysis applications. 15th International Conference on Miniaturized Systems for Chemistry and Life Sciences. 2011;1789-1791. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10254280B2 (en) * 2015-08-10 2019-04-09 Micronit Microtechnologies B.V. Channel for trapping particles to be fed to said channel with a fluid
WO2018187181A1 (en) * 2017-04-03 2018-10-11 The Charles Stark Draper Laboratory, Inc. Microfluidic system for evaluation of chemotherapeutic and immunotherapeutic drugs
US11628437B2 (en) 2017-04-03 2023-04-18 The Charles Stark Draper Laboratory, Inc. Microfluidic system for evaluation of chemotherapeutic and immunotherapeutic drugs
US20210162416A1 (en) * 2019-12-02 2021-06-03 The Charles Stark Draper Laboratory Inc. Multiwell dynamic model for a tumor-immune microenvironment
WO2021185873A1 (en) * 2020-03-16 2021-09-23 Imperial College Innovations Limited Device for perfusion and preservation of tissue specimens ex vivo

Also Published As

Publication number Publication date
US9879308B2 (en) 2018-01-30
JP2015536141A (ja) 2015-12-21
EP2914379A1 (en) 2015-09-09
US20150118742A1 (en) 2015-04-30
WO2014071255A1 (en) 2014-05-08

Similar Documents

Publication Publication Date Title
US9879308B2 (en) Apparatus for ex vivo microfluidic analysis of biologic samples
US20200264205A1 (en) Methods and devices for analysis of defined multicellular combinations
US11229910B2 (en) Microfluidic devices and systems for cell culture and/or assay
Shin et al. A one-stop microfluidic-based lung cancer organoid culture platform for testing drug sensitivity
US10168317B2 (en) Microfluidic device, system, and method for tracking single cells and single cell lineages
ES2459367T3 (es) Modelos tridimensionales de enfermedades de células/tejidos perfundidos
US9029158B2 (en) Microfluidic device
Wlodkowic et al. Wormometry‐on‐a‐chip: Innovative technologies for in situ analysis of small multicellular organisms
US20150204763A1 (en) System for analyzing biological sample material
TW201109653A (en) Microfluidic device
US9663755B2 (en) Apparatus and methods for sperm separation
JP4145938B2 (ja) 細胞分離チップおよびこれを使用した細胞培養方法
WO2012163087A1 (en) Method for sperm motility evaluation and screening and its microfluidic device
Qi et al. Probing single cells using flow in microfluidic devices
US11634676B2 (en) Intracellular delivery using microfluidics-assisted cell screening (MACS)
JP5580117B2 (ja) 細胞分析装置
US20210301261A1 (en) Human liver microphysiology platform and self assembly liver acinus model and methods of their use
Han et al. Imaging technologies for microfluidic biochips
US20230415153A1 (en) Microfluidic devices and method for sampling and analysis of cells using optical forces and raman spectroscopy
Lee et al. Divide and conquer: A perspective on biochips for single-cell and rare-molecule analysis by next-generation sequencing
US20170016884A1 (en) A novel, high-throughput, nanotopographic platform for screening cell migratory behavior
US11845084B2 (en) Microchip high density hanging drop three-dimension culture platform
JP4679847B2 (ja) 細胞の解析方法
CN112955260A (zh) 生物样品保持器和处理器
Hamza An optofluidic platform for longitudinal circulating tumor cell studies in mouse models of cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE CHARLES STARK DRAPER LABORATORY, INC., MASSACH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EPSHTEYN, ALLA;HOLTON, ANGELA;LANDIS, DAVID;AND OTHERS;SIGNING DATES FROM 20140115 TO 20140318;REEL/FRAME:032751/0086

STCB Information on status: application discontinuation

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