WO2004033059A2 - Methods and apparatus for optophoretic diagnosis of cells and particles - Google Patents

Methods and apparatus for optophoretic diagnosis of cells and particles Download PDF

Info

Publication number
WO2004033059A2
WO2004033059A2 PCT/US2003/030975 US0330975W WO2004033059A2 WO 2004033059 A2 WO2004033059 A2 WO 2004033059A2 US 0330975 W US0330975 W US 0330975W WO 2004033059 A2 WO2004033059 A2 WO 2004033059A2
Authority
WO
WIPO (PCT)
Prior art keywords
cell
particle
cells
channel
detecting
Prior art date
Application number
PCT/US2003/030975
Other languages
French (fr)
Other versions
WO2004033059A3 (en
Inventor
Haichuan Zhang
Thomas D.Y. Chung
Jeff Hall
William Soohoo
Josh Kohrumel
Eugene Tu
Mark Wang
Daniel Edward Raymond
Philippe Marchand
Jonathan Diver
William F. Butler
Phan Nguyen
Mirianas Chachisvilis
Andrew Katz
Norbert Hagen
Kris Lykstad
Luis Pestana
Original Assignee
Genoptix, 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 Genoptix, Inc. filed Critical Genoptix, Inc.
Priority to AU2003272804A priority Critical patent/AU2003272804A1/en
Publication of WO2004033059A2 publication Critical patent/WO2004033059A2/en
Publication of WO2004033059A3 publication Critical patent/WO2004033059A3/en

Links

Classifications

    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N15/14Electro-optical investigation, e.g. flow cytometers
    • G01N15/1468Electro-optical investigation, e.g. flow cytometers with spatial resolution of the texture or inner structure of the particle
    • G01N15/147Electro-optical investigation, e.g. flow cytometers with spatial resolution of the texture or inner structure of the particle the analysis being performed on a sample stream
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N15/14Electro-optical investigation, e.g. flow cytometers
    • G01N15/1484Electro-optical investigation, e.g. flow cytometers microstructural devices
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N2015/1006Investigating individual particles for cytology
    • G01N2015/1027
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N15/00Investigating characteristics of particles; Investigating permeability, pore-volume, or surface-area of porous materials
    • G01N15/10Investigating individual particles
    • G01N15/14Electro-optical investigation, e.g. flow cytometers
    • G01N15/1434Electro-optical investigation, e.g. flow cytometers using an analyser being characterised by its optical arrangement
    • G01N2015/1447Spatial selection
    • G01N2015/145Spatial selection by pattern of light, e.g. fringe pattern

Definitions

  • the field of the invention relates generally to optical interrogation methods and apparatus used to determine a property of a cell, a population of cells, and/or cellular components, as well as particles.
  • the methods preferably can be used to select, identify, characterize, and sort individual cells, particles, or groups of cells or particles according to the property of interest.
  • the methods can be used in a variety of applications including, for example, drug screening applications, toxicity applications, protein expression applications, rapid clonal selection applications, biopharmaceutical monitoring applications, quality control application, biopharmaceutical enrichment applications, viral detection, bacterial drug sensitivity screening, and environmental testing applications. More particularly, the systems involved may be used to advantageously diagnose the condition or state of a cell or particle.
  • dielectrophoretic separators utilize non-uniform DC or AC electric fields for separation of particles. See, e.g., U.S. Patent No. 5,814,200, Pethig et al., entitled “Apparatus for Separating By Dielectrophoresis”. The application of dielectrophoresis to cell sorting has been attempted. In Becker (with Gascoyne) et al., PNAS USA, Vol. 92, pp. 860-864, Jan.
  • Ashkin's U.S. Patent No. 3,808,550 entitled “Apparatuses for Trapping and Accelerating Neutral Particles” disclosed systems for trapping or containing particles through radiation pressure. Lasers generating coherent optical radiation were the preferred source of optical pressure. The use of optical radiation to trap small particles grew within the Ashkin Bell Labs group to the point that ultimately the Nobel Prize was awarded to researchers from that lab, including Steven Chu. See, e.g., Chu, S., “Laser Trapping of Neutral Particles", Sci. Am., p. 71 (Feb. 1992), Chu, S., “Laser Manipulation of Atoms and Particles", Science 253, pp. 861-866 (1991).
  • the interaction of a focused beam of light with dielectric particles or matter falls into the broad categories of a gradient force and a scattering force.
  • the gradient force tends to pull materials with higher relative dielectric constants toward the areas of highest intensity in the focused beam of light.
  • the scattering force is the result of momentum transfer from the beam of light to the material, and is generally in the same direction as the beam.
  • the use of light to trap particles is also sometimes referred to as an optical tweezer arrangement.
  • the force of trapping is given by the following equation:
  • FIG. 1 shows a drawing of a particle in an optical tweezer.
  • the optical tweezer consists of a highly focused beam directed to the particle.
  • the focused beam 12 first converges on the particle 10 and then diverges.
  • the intensity pattern 14 relates to the cross-section of the intensity of the beam in the horizontal dimension
  • the intensity pattern 16 is the cross-section of intensity in the vertical dimension.
  • the trapping force is a function of the gradient of the intensity of the light.
  • the force is greater where the light intensity changes most rapidly, and contrarily, is at a minimum where the light intensity is uniform.
  • the gradient force overcomes the scattering force, which would otherwise push the particle in the direction of the light out of the focal point.
  • the laser beam is directed through a high numerical aperture microscope objective. This arrangement serves to enhance the relative contribution from the high numerical aperture illumination but decreases the effect of the scattering force.
  • the term "opticution" has become known in the art to refer to optic radiation killing biological materials.
  • Micromanipulation of particles has been demonstrated.
  • the use of optical tweezers in combination with a microbeam technique of pulsed laser cutting, sometimes also referred to as laser scissors or scalpel, for cutting moving cells and organelles was demonstrated.
  • Optical tweezers and scissors have been used in all-optical in vitro fertilization.
  • Various techniques have included the use of "handles" wherein a structure is attached to a biological material to aid in the trapping. See, e.g., Block, Nature (London), 348, pp. 348-352 (1990).
  • the polarizable matter adopts the pattern of an applied, patterned light intensity distribution. See also, “Matter Rides on Ripples of Lights", reporting on the Burns work in New Scientist, 18 Nov., 1989, No. 1691. Yet others have proposed methods for depositing atoms on a substrate utilizing a standing wave optical pattern. The system may be utilized to produce an array of structures by translating the standing wave pattern. See, Celotta et al, U.S. Patent No. 5,360,764, entitled “Method of Fabricating Laser Controlled Nanolithography”. [0016] Yet others have attempted to cause motion of particles by utilizing light.
  • the technique preferably utilizes propeller shaped molecules, such that the induced rotary motion of the molecules results in translation.
  • Sasaki et al. discloses a method and device for controlling the flow of fine particles along a pattern formed using a scanning laser. See, Sasaki et al., Pattern Formation and Flow Control of Fine Particles By Laser-Scanning Micromanipulation, Optics Letters, Vol. 16., No. 19 (October 1, 1991).
  • polystyrene latex particles were distributed on in a circular pattern of laser light. A driving force was imparted on the particles by repetitive scanning of the trapping beam at a repetition rate of 15 Hz in a clockwise manner. It was observed that all the particles moved together in an orderly fashion around the circular laser pattern.
  • the particle characteristics may include particle size, shape, magnetic susceptibility, magnetic label density, charge separation, dielectric constant, and derivatives thereof.
  • a uniform force field such as a constant, uniform magnetic force field is generated, the particle is subject to that constant force field, and the velocity determined by observing the particle at multiple locations.
  • Variations are described, such as for determining the position of the particle, though the force field is typically described as being constant.
  • a pre-determined force field magnitude and direction is applied to a particle and multiple digital images are analyzed with specified other components to characterize the particles.
  • the system utilized a detection window upstream of the "T" intersection and based upon the detected property, would sort particles within the system.
  • a forward sorting system switched fluid flow based upon a detected event.
  • the fluid flow was set to route all particles to a waste collection, but upon detection of a collectible event, reversed the fluid flow until the particle was detected a second time, after which the particle was collected.
  • a device and associated methods are described for characterizing a cell or particle.
  • the systems generally include a channel having an inlet and an outlet, the channel containing a moving fluid therein for carrying the cell or particle from the inlet to the outlet.
  • the device also includes a detector (or multiple detectors) for detecting the presence of a cell or particle along a portion of the channel.
  • the detector includes at least a first detecting position, a second detecting position, and a third detecting position.
  • the device further includes a light source providing an optical gradient disposed within the channel and between the second and third detecting positions.
  • a control system is coupled to the detector to receive and process detected signals from the detector.
  • the amount of time that a cell or particle takes to flow through a first distance i.e., its time-of-flight
  • the particle is then flowed past a second, downstream distance in the presence of an optical gradient and its time-of-flight is measured.
  • a comparison of the measured time-of-flights for the first and second distances is used to characterize the cell or particle.
  • the optical gradient serves as an Optical speed-bump', serving to slightly retard the progress of the cell or particle in an amount related to the degree of interaction between the particle and the optical gradient.
  • the method can be used to characterize cells based on one or more biological properties of the cell.
  • the characterization information may be utilized to further sort cells or particles based upon an observed parameter.
  • coherent light may be used for illumination of the particle.
  • a pattern generator disposed upstream of the particles selectively illuminates the particle.
  • a detector array determines particle position as a function of time.
  • a system utilizing coherent light scans a beam over the channel.
  • a detector determines the cell or particle positioning as a function of time.
  • Incoherent light may be used for illumination. Detection may be by any number of techniques, such as through the use of a mask and detector array, or by use of a line camera. Electrical detection of the cell or particle position may be utilized, such as where an impedance detection is utilized.
  • an additional detection beam may be utilized.
  • the beam is directed axially along the channel and an additional detector is located upstream from the other detectors.
  • additional detectors the various detectors may be optimized to determine different detectable characteristics.
  • the initial detector may be utilized to activate or otherwise tune the remaining detectors.
  • the initial detector may be used as a gating detector in the sense that it detects the presence of an incoming cell or particle.
  • the gating detector has the capability to detect whether the incoming test subject (i.e. cell or particle) is in a condition for measurement.
  • the detector can determine and reject a sample if it appears that the cells are clumped together or otherwise unrepresentative of the cells or particles of interest.
  • the device has applications in a wide variety of diagnostic applications including, but not limited to, cancer diagnostic applications and infectious disease diagnostic applications.
  • Fig. 1 is a graphical depiction of optical intensity patterns for a prior art optical tweezer system, showing both the focus beam, a particle and the cross-section of intensity of the beam.
  • Fig. 2 shows a plan view of a time-of-flight system.
  • Fig. 3 shows a generalized block diagram of a microfluidic detection system.
  • Fig. 4 is a block diagrammatic view of one apparatus and associated method for detection, namely one in which coherent light detection is utilized.
  • Fig. 5A shows a plan diagram of a coherent light detection system utilizing scanning detection.
  • Fig. 5B graphically shows the operation of scanning system.
  • Fig. 6A shows a plan view of a system utilizing an incoherent light detection system including a detector mask.
  • Fig. 6B shows a side plan view of a detector mask.
  • Fig. 7 shows a plan view of system utilizing incoherent light for detection along with a line camera.
  • Fig. 8A shows a plan view of an electronic detection system.
  • Fig. 8B shows close up view of the charmel and its associated electrodes.
  • Fig. 9 shows a side view of a gravity-based time-of-flight system.
  • Fig. 10 is a plan view of a microfluidic channel based detection system.
  • Fig. 11 is a system block diagram of various subsystems within the system.
  • Fig. 12 depicts the optical subsystem for one implementation of a time-of-flight system.
  • Fig. 13 shows a combined block diagram and processing functionality and software for the acquisition subsystem.
  • Fig. 14 shows a flow chart of one possible implementation of the software subsystem.
  • Fig. 15 depicts the forces on a particle in a time-of-flight system.
  • Fig. 16 depicts the optical force on a typical 10 micron cell by a row numerical aperture (NA) laser line.
  • Fig. 17 is a graph of time delay as a function of escape velocity normalized to flow velocity for a 10 micron bead as simulated.
  • Fig. 18 shows the data of Fig. 17 plotted in a log-log format.
  • Fig. 19 is a depiction of an optical system used to perform line scan and fast scan analysis on samples.
  • Fig. 20 depicts a preferred detection scheme utilizing multiple detectors and a detection laser.
  • Fig. 21 illustrates a perspective view of a preferred embodiment of a time-of-flight device.
  • Fig. 22 illustrates another perspective view of the preferred embodiment of the time- of-flight device shown in Fig. 21.
  • FIG. 23 schematically illustrates the preferred embodiment shown in Figs. 21 and 22.
  • Fig. 24 schematically illustrates the fluidics used in one preferred embodiment of the invention.
  • Fig. 25 schematically illustrates a preferred embodiment of the flow pump used to create a low, constant flow rate.
  • Fig. 26 illustrates a preferred embodiment of the flow pump used to create a low, constant flow rate.
  • Fig. 27 illustrates one embodiment of a massively parallel system.
  • Fig. 28 illustrates yet another embodiment of a massively parallel system.
  • Fig. 29A illustrates a device capable of both characterizing and sorting a cell or particle.
  • Fig. 29B illustrates a preferred device and method for sorting cells or particles in the device shown in Fig. 29A.
  • Fig. 29C illustrates another preferred device and method for sorting cells or particles in the device shown in Fig. 29A.
  • Fig. 30 is a histogram of the measured escape velocities of Plasmodium-infected and non-infected red blood cells.
  • Fig. 31 shows a comparison of the mean escape velocity for Plasmodium-infected and non-infected red blood cells .
  • Fig. 32 is a histogram of time-of-flight measurements for normal red blood cells
  • Fig. 33 shows a comparison of the mean time-of-flight values for the infected and control cells of Fig. 32.
  • Fig. 34 is a histogram of time-of-flight measurements for normal red blood cells
  • Fig. 35 shows a comparison of the mean time-of-flight values for the infected and control cells of Fig. 34.
  • Fig. 36 illustrates a histogram of the ratio of T 2 /T ⁇ plotted against the percentage of cancerous and non-cancerous cells from breast tissue.
  • Fig. 37 shows a comparison of the mean T 2 /T ⁇ ratio of the cancerous and non- cancerous cells of Fig. 36.
  • Fig. 38 illustrates a histogram of the ratio of T 2 /T ⁇ plotted against the percentage of cancerous skin cells and non-cancerous skin cells.
  • Fig. 39 shows a comparison of the mean T 2 /T ⁇ ratio of the cancerous and non- cancerous cells of Fig. 38.
  • Fig. 40 is a scatter plot of the T 2 /T ⁇ ratio as a function oft] for polystyrene beads and
  • Fig. 41 is a histogram of the number of particles as a function of T 2 /T ⁇ ratio for experimental data shown in Fig. 40.
  • Fig. 42 is a scatter plot of the T 2 /T ⁇ ratio as a function of event number for 24657 rho+ (wild type) yeast and MYA-1133 rho(0) (mutant) yeast.
  • Fig. 43 is a histogram of the percentage of cells as a function of T 2 /T ⁇ ratio for experimental data shown in Fig. 42.
  • Fig. 44 is a scatter plot of the T 2 /T ⁇ ratio as a function of event number for treated and non-treated HL60 cells.
  • the treated HL60 cells were treated with 1% DMSO.
  • Fig. 45 is a histogram of the percentage of cells as a function of T 2 /T ⁇ ratio for experimental data shown in Fig. 44.
  • Fig. 46 is a histogram of the percentage of unactivated and activated T cells as a function of T 2 /T] ratio.
  • Bio Property means a distinct phenotype, state, condition, or response of a cell or group of cells, for example, whether a cell is diseased, has been infected by a virus, the degree to which a cell expresses a particular protein, the stage in the cell cycle a particular cell is presently at, whether the cell is affected by the presence of a chemical compound, a particular phenotype of the cell, whether a ligand is bound to the surface of a cell, cytoskeletal changes in the cell, whether a cell is decorated with antibodies, the presence or absence of a cellular component (e.g., an organelle or inclusion body), a change in one or more cellular components, the toxicity of chemical compounds, a physical property of a cell or population of cells, a response of a cell or population of cells to an external stimulus, cellular motility, membrane fluidity, state of differentiation, viability, size, osmolarity
  • Determining is meant to indicate that a particular phenotype, state, condition, or response is ascertained.
  • Dielectric constant is defined to be that property which determines the electrostatic energy stored per unit volume for unit potential gradient. (See, e.g., the New IEEE Standard Dictionary Of Electrical And Electronics Terms, ⁇ 1993).
  • the "escape velocity” is defined as the minimum speed at which an interrogated cell or particle no longer tracks the moving optical gradient.
  • the "optical dielectric constant” is the dielectric constant of a particle or thing at optical wavelengths. Generally, the optical wavelength range is from 150 A to 30,000 A.
  • An "optical gradient field” is an optical pattern having a variation in one or more parameters including intensity, wavelength or frequency, phase, polarization or other parameters relating to the optical energy.
  • an optical gradient field or pattern may also be called an optical fringe field or fringe pattern, or variants thereof.
  • a "moving optical gradient field” is an optical gradient field that moves in space and/or time relative to other components of the system, e.g., particles or objects to be identified, characterized, selected and/or sorted, the medium, typically a fluidic medium, in contact with the particles, and/or any containment or support structure.
  • An "optical scattering force” is that force applied to a particle or thing caused by a momentum transfer from photons to material irradiated with optical energy.
  • An “optical gradient force” is one which causes a particle or object to be subject to a force based upon a difference in dielectric constant between the particle and the medium in which it is located.
  • Optophoresis or “Optophoretic” generally relates to the use of photonic or light energy to obtain information about or spatially move or otherwise usefully interact with a particle.
  • Optophoretic constant or “optophoretic signature” or “optophoretic fingerprint” refer to the parameter or parameters which distinguish or characterize particles for optical selection, identification, characterization or sorting.
  • Separatation of two objects is the relative spatial distancing over time of a particle from some other reference point or thing.
  • FIG. 2 shows a plan view of a time-of-flight system.
  • a channel 20 is defined by an inlet 22, an outlet 24 and first and second sidewalls 26.
  • the sidewalls 26 are linear, and substantially parallel to each other.
  • the outlet 24 has a width D.
  • D may be on the order of substantially 60 microns or smaller. Typically, D would exceed the size of the largest expected cell or particle to flow through the channel 20.
  • the channel 20 contains a moving fluid therein, shown by arrow A in Fig. 2, which carries the cells or particles 36 from the inlet 22 to the outlet 24.
  • Particle as used herein refers to any type of small body and includes, as an example, spores, pollen, and particulate matter such as airborne or other environmental contaminants. These include airborne as well as waterborne contaminants.
  • the moving fluid flows within the channel 20 at a constant flow rate.
  • the flow rate of the fluid within the channel 20 is preferably controllable. A flow rate is chosen such that the flow rate of the fluid exceeds the escape velocity of the cells or particles 36 transported through the channel 20, that is the flow rate is chosen such that cells or particles 36 do not get "stuck" on the optical gradient 38 (described in detail below) .
  • the system includes multiple, preferably three, detecting positions, 31, 32 and 33.
  • the detecting positions 31, 32, 33 may be associated with detection of times, e.g., ti, t 2 , t 3 .
  • the times may be absolute, relative or elapsed.
  • the two outermost detecting positions, i.e., 31 and 33 in this example, are separated by a distance L.
  • the difference between the first two detecting positions ti, t 2 define a time interval Tl.
  • Tl may also be associated with a first detection zone corresponding to the motion of the particle 36 through the zone Tl.
  • the second and third detecting positions t 2 , t 3 define a second detection zone T2.
  • the second detection zone also corresponds to the time difference between t 2 , t 3 .
  • the distance between t 2 1 3 is equal to the distance between ti t 2 .
  • the cell or particle 36 flows from the inlet 22 through the channel 20 through the outlet 24.
  • representative particle speeds are from substantially 50 to substantially 200 microns per second.
  • the time-of-flight through that zone is measured, that is the time it takes for the cell or particle 36 to move from position ti to t 2 .
  • An optical gradient 38 is disposed within zone T2.
  • the optical gradient 38 is substantially linear and has a thickness in the fluid flow direction which is substantially less than the transverse dimension.
  • the width is less than substantially 10% of the transverse length of the optical gradient 38.
  • the optical gradient 38 is disposed within a portion of the channel 20 such that the optical gradient 38 is generally orthogonal to the direction of the fluid flow.
  • the optical gradient 38 is preferably formed using a coherent light source such as a laser that is passed through a cylindrical lens.
  • the optical gradient 38 may be formed using a scanning laser system.
  • the moving cell or particle 36 moves through zone T2, it will intercept the optical gradient 38. If the optical gradient 38 has no effect on the moving particle 36, T2 equals Tl, assuming the physical difference in detecting positions is equal for zone 1 and for zone 2. If the moving cell or particle 36 does optically interact with the optical gradient 38, the cell or particle 36 will typically be slowed or retarded in its transit through zone T2. Accordingly, T2 would be greater than Tl, assuming the detection positions are uniformly spaced. [0096]
  • the optical gradient 38 may be said to be "static" relative to the underlying device, such as the device that defines the channel 20. However, the relative motion of the cell or particle 36 and the optical gradient 38 provide the discriminating force within the system.
  • the cell or particle 36 In operation, the cell or particle 36 generally moves at a speed in the range from about 50 to about 200 microns per second.
  • the detection spacing that is the distance between adjacent detecting positions, is typically on the order from approximately 20 to approximately 50 microns. While shown with three detection positions, the system may use more detection positions, or different types of detectors, as desired.
  • the throughput of the system may be in the range of approximately 500 to about 2,000 particles per hour per channel 20.
  • Fig. 3 shows a generalized block diagram of a microfluidic detection, preferably diagnostic, system 39.
  • the substrate 40 containing the channel 20 receives the moving cell or particle 36.
  • An illumination system 42 preferably including a laser as the light source, provides the optical gradient 38.
  • a detection system 44 is operatively positioned to detect the position of the cell or particle 36 at multiple, typically three or more, locations.
  • a control system 46 controls the illumination system 42 via the communication path 48.
  • the output of the detection system 44 is coupled to the control system 46 via the communication path 50.
  • the control system 46 serves to receive and process the detected signals from the detection system 44.
  • a display 54 optionally depicts the detected intensity of the particle as it passes the sections of the detection system 44. In this regard, the display 54 can show the amount of time it takes the cell or particle 36 to pass through zones Tl and T2.
  • the display 54 may further display a ratio using the values of Tl and T2 (i.e. T1/T2 or T2/T1).
  • Figs. 4, 5A, 6 and 7 show optical detection systems. More particularly, Fig. 4 is a multi-element detection system utilizing coherent light. Fig. 5A is a scanning detection system using coherent light. Fig. 6 is a detector system using a detector mask, utilizing incoherent light. Fig. 7 is a line camera system utilizing incoherent light. Finally, Fig. 8 shows an electronic detection system, specifically utilizing an impedance detector. [00100] Fig. 4 is a block diagrammatic view of one apparatus and associated method for detection, namely one in which coherent light detection is utilized.
  • a flow cell 60 such as described in connection with Fig. 2 is adapted to receive fluid flow in the direction y, where that fluid includes cells or particles 36 for analysis.
  • An illumination system 62 includes a pattern generator 64 and a light source 66.
  • the pattern generator 64 may be implemented in any number of formats. For example, a VCSEL (vertical cavity surface emitting laser) array may be directed towards the system 60. Alternately, diffractive optical elements may be used.. In yet other implementations, light modulators, such as MEMs mirror systems may be utilized. In yet another implementation, light modulator implementation, an AO (Accousto-Optical) modulator may be utilized. However implemented, the illumination system 62 serves to generate patterned illumination for the detection of the cell or particle position, timing or speed.
  • a laser bump pattern generator 68 receives the output of a laser 70 to generate the optical gradient (item 38 in Fig. 2).
  • the pattern generator 68 may be formed from any variety of technologies, e.g., a scanning system, such as an oscillating mirror scanning system, or through the use of diffractive optical elements.
  • an optical system 72 such as a beam splitter, may be utilized to combine the light from the pattern generator 64 as well as the laser bump pattern generator 68 using a lens 74.
  • the illumination source 66 may be relatively low powered.
  • the illumination laser diode may be an 8mW 635 nm laser making the light in the visible range.
  • the laser 70 for generating the optical gradient may be of relatively higher power, such as a 1W laser diode strip.
  • the laser 70 may optionally be in the infrared spectrum, such as 1064 nm.
  • a beam splitter 76 may be utilized to divert the light to imaging optics which may then be detected with camera 78, such as a CCD camera.
  • a detector array 80 receives the light path from the device 60, after passing tlirough appropriate focusing optics.
  • the detector array may be implemented in any number of technologies. For example, a PIN array may be utilized.
  • the output of the detector array 80 is optionally provided to a signal processor 82, such as a digital signal processor (DSP).
  • DSP digital signal processor
  • a time interval measurement unit 84 detects the time intervals (see, e.g., Tl and T2, or ti, t 2 , and t 3 , Fig. 2).
  • a timing diagram 86 may be displayed, such as through a graphical display or a printed display. As shown in Fig. 4, the left most curve depicts the number of counts (i.e., particles 36) in the interval Tl, whereas the right most curve depicts the number of counts in the interval T2. As can be seen, the average time to traverse region T2 was greater than the time to traverse region Tl.
  • the camera 78 may be utilized for any functions, including monitoring and alignment.
  • the signal processing system 82 optionally includes demodulation, differential peak detection and digitizing.
  • the illumination system 62 may include modulation to enhance the signal to noise ratio (SNR) for detection.
  • Fig. 5A shows a plan diagram of an coherent light detection system utilizing ' scanning detection. The description for many components in Fig. 5A is the same as that for Fig. 4, and accordingly those components have been similarly numbered.
  • a coherent light source 90 such as a laser diode, provides illumination of the system 60.
  • the output of the illumination system 90 is scanned over at least a portion of the channel device 60.
  • the scanning device 92 may be, for example, a scanning mirror system, preferably an x, y scanning system such as a system using two rotating mirrors having non-parallel axis.
  • the scanning system would typically scan in a raster scan fashion, such as shown in Fig. 5B.
  • the spacing between the scans is exaggerated in Fig. 5B for purposes of illustration.
  • the output of scanning device 92 is passed through the associated optics to scan the operative portions of the channel device 60.
  • the output of that scan is then imaged upon a detector 94.
  • a beam splitter 76 may direct the output illumination to an imaging camera 78, such as a CCD camera.
  • the detector 94 output is then provided to a signal processor 82, which in turn provides its output to an input for the time correlation and interval measurement unit 96.
  • the time correlation and interval measurement unit 96 receives a clock signal input, such as provided from a time base 98.
  • Fig. 6A shows a plan view of a system utilizing an incoherent light detection system including a detector mask.
  • Fig. 6B shows a side plan view of a detector mask 102. Similar components disclosed in Figs. 4, 5A, 6A, 7, and 8 are shown having identical element numbers and unless indicated otherwise, operate in the same manner.
  • An illumination source 100 preferably a visible illumination lamp provides a source of light for detection. The light impinges upon the particle to be analyzed, whether passing through focusing optics or not. The light output from the device 60 is then directed to the mask 102. The mask 102 is placed before the detector array 80.
  • the mask 102 includes three apertures 104.
  • the apertures are elongate, preferably rectangular, and preferably evenly spaced one from another.
  • the apertures 104 define detection windows for the cell or particle 36 passing through the device 60.
  • the width of the apertures 104 are about 1.5 ⁇ m.
  • the width of the apertures 104 should be no more than about one-half of the size of the cell or particle 36.
  • the width of the aperture 104 is chosen such that enough light passes there through to produce a signal strong enough to be picked up by the detector.
  • Fig. 7 shows a plan view of system utilizing incoherent light for detection along with a line camera.
  • Figs. 8A and 8B show a plan view of an electronic detection system. More particularly, this system utilizes impedance measurement for detection. Detection electrode 120, 122 and 124 are spaced at detection positions PI, P2 and P3, respectively. Fig.
  • FIG. 8B shows a plan view of a particle 36 passing through the channel device 60 having three detection electrodes, 120, 122, and 124.
  • An electromagnetic field is set up between the individual electrode pairs, e.g., 120.
  • the impedance of the space between the electrode pairs changes as a function of the particle 36 position.
  • the impedance detector and signal processing system 126 is electrically connected to the electrode pairs, e.g., 120.
  • the profile for the cell or particle 36 as it moves through the detector then is analyzed for the desired property, such as the interval for zones 1 and 2, or the time difference between the two zones.
  • Fig. 9 shows a side view of a gravity-based time-of-flight system.
  • a surface 130 is angled relative to horizontal, such that there is a component of gravitational force exerted on the particle or cell 131 to cause it to move down the surface 130.
  • Detectors 132, 134, 136 are disposed to detect the particle 131 as it passes the detectors.
  • a time difference ⁇ ti is detected between the first two detectors 132, 134.
  • An optical gradient 138 comprising an optophoretic "speed bump" is disposed between the second detector 134 and the third detector 136.
  • a time difference is measured between the second detector 134 and the third detector 136, and is designated ⁇ t 2 .
  • Fig. 10 is a plan view of a microfluidic channel based detection system.
  • a particle or cell 140 flows through the channel 142 in the direction of the arrow.
  • a first light source 144 such as a light emitting diode, is directed to a first detector 154, such as a photo detector to detect the time at which the particle or cell 140 reaches a first position.
  • FIG. 11 is a system block diagram of various subsystems within the system.
  • the optical subsystem 160 is depicted using the general structure shown in Fig. 6.
  • a computer control system 162 interfaces with other subsystems, including the electronics driver subsystem 164, the microftuidics subsystem 166 and the electronics acquisition system 168. As shown, various connections or buses are provided as required between the various subsystems, such as from the computer control subsystem 162 to the microfluidics subsystem 166, and from the electronics driver subsystem 164 to the microfluidics subsystem 166.
  • a power subsystem 170 connects to all subsystems.
  • Fig. 12 depicts the optical subsystem for one implementation of a time-of-flight system.
  • Fig. 13 shows a combined block diagram and processing functionality and software for the acquisition subsystem.
  • An optical subsystem 200 receives optical input from a white light source 202 and a laser 204 to generate the optical gradient. Detection may consist of detectors 208 to detect the time of crossing of the particle at a predefined position.
  • An imaging camera 206 may be utilized for general system imaging, alignment or to otherwise determine the location of the particle.
  • a power system 210 is connected to all necessary electrical components.
  • Fig. 13 shows two possible detection, analysis and display systems. Under solution A, a pattern identification step 212 may be utilized. Analog circuitry may be implemented or various digital techniques, such as digital signal processors (DSPs) may be utilized.
  • DSPs digital signal processors
  • the analysis section 214 receives the output of the pattern identification 212 and optionally the direct output of the detector 208, which is received by the data acquisition subsystem 216.
  • a data processing system 218 performs various functions, including optionally statistical analysis. The various functionalities of system 214 may be performed under control of a personal computer, such as operating in a windows based environment.
  • Solution B depicts the functionality 230 in which the output of the detectors 208 is provided to a data acquisition functionality 232 and/or a pattern identification subsystem 234. If the data acquisition functionality 232 and pattern identification functionality 234 are present, they may be performed in either order. As depicted, an analog-to-digital (A D) converter 236 is provided to convert acquired analog data to digital data.
  • a D analog-to-digital
  • an FPGA platform may be utilized.
  • the acquired data is then subject to data processing step 238, which optionally includes statistical analysis.
  • Ultimate display to the user may be under control of a graphical user interface or peripheral interface 240.
  • the system may be under microprocessor control 240.
  • Fig. 14 shows a flow chart of one possible implementation of the software subsystem.
  • Fig. 15 depicts the forces on a cell or particle in a time-of-flight system.
  • TOF Time-of-flight
  • TOF instrument measures the time delay of flowing particles or cells by a laser beam in micro fluidic environment. The time delay according to optophoretic property is used to analyze the biological differences between populations.
  • the TOF instrument is aiming for low cost and diagnostic applications.
  • the force equation is: d 2 x dx m ⁇ _- - F optical lateral (x) ⁇ F dmg (— -V f (z),z) Equation l
  • b is the drag coefficient that depends on the radius and shape of the object and the viscosity ("stiffness") of the medium.
  • is the viscosity of medium in g / cm s. Due the boundary effect, the flow velocity decreases upon the distance to the boundary of the micro-channel.
  • GLMT and geometrical simulations find that optical forces F opticalJalera! (x) and F oplic ⁇ l ⁇ (x) on a cell by low numerical aperture (NA) laser beam are typically as shown in Figure 16.
  • Fig. 16 depicts the optical force on a typical 10 micron cell by a 0.1 (NA) laser line.
  • Fig. 17 is a graph of time delay as a function of escape velocity normalized to flow velocity for a 10 micron bead as simulated. As expected, as the flow velocity approaches the escape velocity, the time of delay increases. In addition, Fig. 17 shows that lower flow velocities produce larger time delays for a given escape velocity/flow velocity. Fig. 18 shows the data of Fig. 17 plotted in a log-log format. The devices and methods described herein may take advantage of the interaction between flow velocity and delay time to optimize the sensitivity of the device. For example, as seen in Fig. 17, the time delay increases as the flow velocity approaches the particle's escape velocity.
  • Fig. 17 shows that the slower flow velocities produce the largest time delays. Consequently, slower fluid flow rates will produce more noticeable changes in the measured travel times in the detection zones (i.e., zone T2 as compared to Tl). Of course, slower flow rates will reduce the overall throughput of the device and may not be desirable in certain applications.
  • Fig. 20 shows a preferred signal detection scheme utilizing multiple detectors.
  • Fig. 20 includes components similar to those described in Fig. 2.
  • Fig. 20 includes an additional detection position 280.
  • a detection laser beam 282 is directed to the system in a direction generally parallel to the channel 20.
  • the detection laser beam 282 is a low power visible laser beam.
  • the detectors may be any of the type described generally herein.
  • the added detector may be optimized for properties other than that which the other particle detectors are optimized for.
  • the detectors may be optimized based on optical arrangement, such as optical focusing, and/or the geometry of the detector mask, and/or by electronic processing, such as by dedicated filtering and/or the use of threshold circuits.
  • the signal from the additional detector may be more representative of particle physical properties, such as particle physical size.
  • the first detectors may be optimized for detecting the particle physical position.
  • the additional detector may be physically placed upstream of the other detection positions.
  • the additional detector may provid trigger selection for the time-of-flight signal acquisition based upon a particles detected property or properties.
  • the additional detector acts as a gating detector that indicates to the other components of the device, i.e., the detectors that measure the time intervals Tl and T2 that a cell or particle is about to pass through the detection zones. Throughput and accuracy of signal acquisition generally increases by adding an additional detector.
  • Figs. 21 and 22 show a preferred embodiment of a time-of-flight device 200 that employs an additional detector as is described above.
  • the device 200 includes optical, mechanical, and fluidic components that are mounted on a base plate 202.
  • the device 200 includes a infrared (IR) laser 204 that outputs a coherent beam of infrared light.
  • IR infrared
  • the IR laser 204 has a wavelength in the range of about 780 nm to about 1064 nm. Two preferred wavelengths within this range include 808 nm and 1064 nm.
  • the IR laser 204 creates the optical gradient (e.g., optical gradient 38 as is shown in Fig. 2) that is used to differentially slow cells or particles passing there through.
  • the output of the IR laser 204 passes through a laser collimation lens 206 and is directed against two mirrors 208.
  • the light then passes through a IR cylindrical lens 210 and into an IR beam splitter 212.
  • the light passing through the IR beam splitter 212 is then directed through a focusing lens 213 into a microfluidic mounting system 214.
  • the microfluidic mounting system 214 includes at least one channel (not shown) therein through which the cells or particles pass.
  • the IR optical gradient is disposed generally perpendicular to the direction of flow through the channel contained in the microfluidic mounting system 214.
  • the time-of-flight device 200 also includes a visible (VIS) laser 216 that outputs a coherent beam of light.
  • the visible laser 216 comprises a laser diode operating at around 635 nm.
  • the visible laser beam acts as a detection laser as is described in more detail above and as shown in Fig. 20.
  • the visible laser beam is reflected off another mirror 218 and passed through two lenses 220, 222.
  • the visible laser beam is then reflected off a mirror 224 and passed through a filter 226.
  • the visible laser beam then passes through a white light beam splitter 228 and into a cylindrical lens 230.
  • the visible laser beam then passes through the focusing lens 213 and into the microfluidic mounting system 214.
  • the visible laser beam produces a line of light that is disposed within the channel (not shown) contained within the microfluidic mounting system 214 in a direction that is generally parallel to the direction of fluid flow.
  • This detection laser is shown in detail in Fig. 20 (detection laser 282).
  • both the IR laser beam and the visible laser beam pass through a collection lens 232 and into a beam splitter 234.
  • One output of the beam splitter 234 is then directed through an imaging lens and filter 236.
  • the light passing through the imaging lens and filter 236 is then directed through a mask M2 disposed in front of a first detector 238.
  • the mask M2 preferably includes a single window therein as is shown, for example, in Fig. 11.
  • the first detector 238 is preferably used as an event or gating detector as is described above and shown in Fig. 20.
  • the other output of the beam splitter 234 is directed to an imaging lens and filter 240 and into a mirror 242.
  • the reflected light is then directed through a mask Ml disposed in front of a second detector 244.
  • the mask Ml preferably includes three windows therein as is shown, for example, in Fig. 11.
  • the second detector 244 is preferably used to determine the amount of time it takes a cell or particle to travel from a first detecting position ti to a second detecting position t 2 and the amount of time it takes the same cell or particle to travel from the second detecting position t 2 to a third detecting position t 3 .
  • the three windows in the mask Ml correspond to the detecting positions ti, t 2 , and t 3 .
  • the device 200 may also include an optional camera 246 such as a CCD camera that is used to image the microfluidic mounting system 214.
  • the optional camera is used to calibrate the device 200. If the optional camera 246 is used, a white light source 248 is preferably used to provide additional light to enhance the imaging of the microfluidic mounting system 214 and the channel contained therein.
  • Fig. 33 illustrates the underside of the device 200.
  • the device 200 includes a power input 250 that is preferably connected to a conventional 110 VAC power source.
  • the device 200 also includes a computer interface 252 that allows data communication between the device 200 and an external computer (not shown).
  • the computer interface 252 is a 68 pin high-speed connection.
  • Fig. 33 also illustrates the laser driver 254 for the IR laser 204.
  • Data acquisition electronics 256 are included in a printed circuit board preferably located on the underside of the device 200.
  • the data acquisition electronics 256 includes therein a driver for the visible laser 216.
  • a power supply 258 for the device 200 is located on the underside of the base plate 202.
  • FIG. 23 schematically illustrates the preferred embodiment illustrated in Figs. 21 and 22 along with the channel 20 of the microfluidic mounting system 214.
  • the optical gradient 38 is shown disposed inside the channel 20.
  • a cell or particle 36 travels down the channel 20 in the direction of the arrow.
  • Detectors 238 and 244 are coupled to signal capturing/data processing electronics 260.
  • Fig. 24 schematically illustrates the fluidics 300 used according to one preferred embodiment to produce a substantially constant, low flow rate through a device 302 containing a channel 304 therein.
  • the channel 304 is oriented in the vertical direction.
  • a reservoir 306 is provided that contains the cells or particles in a fluid medium.
  • Tubing 308 is provided between the reservoir and the inlet to the channel 304.
  • Additional tubing 310 is provided at the outlet of the channel 310 and connects to a flow pump 312.
  • the flow pump 312 is used to provide a substantially constant yet low flow rate of fluid through the channel 304.
  • the flow pump 312 is advantageously controllable so that various flow rates can be used.
  • the bypass 314 is used to evacuate fluid from the system after passing through the device 302.
  • a controllable valve 316 is shown in Fig. 23 that is used to direct fluid from the flow pump 312 to the bypass 314.
  • Figs. 25 and 26 illustrate a preferred embodiment of the flow pump 312.
  • the flow pump 312 includes a motor 314.
  • the motor 314 is preferably a stepper motor with an integral 50x to lOOx gear reduction drive of the output rotation rate.
  • the motor 314 is coupled to rotationally drive a leadscrew 316.
  • the leadscrew 316 preferably is a high pitch leadscrew.
  • the leadscrew 316 is mechanically coupled to a stage 318. Rotation of the leadscrew 316 imparts linear motion to the stage 318 in the direction of arrow A shown in Fig. 36.
  • the stage 318 is coupled to a syringe plunger 320.
  • a stationary syringe 322 is mounted atop a housing 324.
  • the syringe 322 is a microsyringe with a volume of between about 1 ⁇ L to about 100 ⁇ L.
  • the plunger 320 has a travel distance (stroke) of between about 2 to about 5 cm. It is generally preferably to use a syringe 322 with a small cross-sectional area and a long stroke. These two conditions advantageously produce low flow rates.
  • Tubing 326 is provided at the end of the syringe 322 that is opposite to the end of the syringe 322 with the plunger 320. The tubing 326 is, in turn, connected to a device such as device 302 shown in Fig. 34.
  • the motor 314 rotates the leadscrew 316 which causes the stage 318 and its connected plunger 320 to move in the axial direction (Arrow A in Fig. 36).
  • the syringe 322 is used to withdraw fluid containing cells or particles from a reservoir such as reservoir 306 shown in Fig. 34. The cells or particles then pass with the fluid through a channel such as channel 304 shown in Fig. 34.
  • the plunger 320 can be depressed into the syringe 322 using the motor 314 operating in the reverse direction to evacuate the fluid and cells/particles contained therein using, for example, the bypass 314.
  • the syringe 322 may be preloaded with cells or particles and the fluid can then be pushed through the channel 304 by depressing the plunger 320 using the motor 314.
  • Figs. 27 and 28 show a exemplary view of various possible embodiments for massively parallel system.
  • an array 350 of multiple channels 352 are disposed parallel to one another.
  • the array 350 may be a two dimensional array, as is shown in solid in Fig. 37, or alternatively, the array 350 may be three-dimensional, as is shown in dashed lines in Fig. 37.
  • the array 350 of channels 352 are connected to a common inlet 354 as well as a common outlet 356.
  • the three detecting positions ti, t 2 , and t 3 are also shown in Fig. 37.
  • An optophoretic gradient 358 is disposed between the second and third detecting positions so as to slow down the particles or cells differentially based on their properties.
  • the optophoretic gradient 358 preferably is formed as a line as is shown in Fig. 37. If, however, the array 350 is a three-dimensional array, the optophoretic gradient 358 preferably forms a plane (not shown) that passes through the channels 352. In the three-dimensional array 350, a detector system capable of determining which "layer" of the array the cell or particle is present is needed.
  • a detector system capable of determining which "layer" of the array the cell or particle is present is needed.
  • One example is the electrode-based detector system illustrated herein in Figs. 8A and 8B.
  • Fig. 28 discloses a three-dimensional parallel system that includes a series of stacked two-dimensional arrays 360 separated by a distance D.
  • a light source 362 such as a Bessel beam, which is capable of reconstructing itself, is used to illuminate the arrays 360.
  • Fig. 28 also shows an optophoretic gradient 364 disposed across the channels contained in each array 360.
  • Figs. 29A, 29B, and 29C illustrate an embodiment of a device 400 is capable of both characterizing and sorting cells or particles 402.
  • a channel 404 is provided that contains a region having a plurality of detecting positions.
  • Fig. 29 A shows three such detecting positions ti, t 2 , and t 3 .
  • An Optophoretic gradient 406 is disposed between detecting positions t 2 and t 3 .
  • These detecting positions have one or more associated detectors (not shown) that are used to calculate the time-of-flight for region Tl and region T2. A comparison of the time-of-flight for each region can then be made to characterize the cell or particle 402.
  • the device 400 also is capable of sorting cells or particles 402.
  • Fig. 29A shows two such branches 410A and 410B, however, it should be understood that the single channel 404 may branch into any number of branches 410.
  • Fig. 29B illustrates one method used to sort the cells or particles 402. In this method a moving optical gradient 412 is used to sweep certain cells or particles 402 having certain desired characteristics into one of the braches 410. The optical gradient 412 starts at the one edge of the channel 404 (the lower edge shown in Fig. 29B) and scans generally perpendicular to the direction of fluid flow as shown by the arrows in Fig. 29B.
  • the cell or particle 402 When the cell or particle 402 encounters the moving optical gradient 412, the cell or particle 402 either passes into branch 410A or branch 410B. Those cells or particles 402 that interact more with the moving optical gradient 412 tend to be swept into the upper branch 410B (cell or particle 402B shown in Fig. 29B) while the other cells or particles 402 tend to pass into branch 410A (cell or particle 402A shown in Fig. 29B).
  • Fig. 29C illustrates an alternative method used to sort the cells or particles 402.
  • this embodiment employs a stationary optical gradient 414.
  • the stationary optical gradient 414 is oriented at an angle to the direction of fluid flow. Those cells or particles 402 that interact more with the stationary optical gradient 414 tend to travel along the angled optical gradient and pass into the upper branch 410B (cell or particle 402B shown in Fig. 29C) while the other cells or particles 402 tend to pass into branch 410A (cell or particle 402A shown in Fig. 29C).
  • EXPERIMENTAL DATA [00136] A.
  • a variety of methods have been traditionally used to diagnose malaria infection.
  • One method uses a visible stain such as Giemsa stain and subsequent microscopic evaluation.
  • infection may be detected using nucleic-acid binding stains such as, for example, the fluorescent stain acridine orange followed by fluorescence microscopy or flow cytometric analysis.
  • nucleic-acid binding stains such as, for example, the fluorescent stain acridine orange followed by fluorescence microscopy or flow cytometric analysis.
  • Still other diagnostic techniques use immunological methods that can detect the presence of Plasmodium-specific antigens. All of these methods, however, have limitations. All of the methods require the addition of exogenous reagents. Some of the methods kill the cells, thereby destroying sample integrity. Moreover, these methods often require expensive equipment that needs to be manned by skilled operators.
  • the stained cells were stained with the fluorescent nucleic-acid binding dye SybrGreen as a confirmatory test to distinguish infected RBCs (fluorescent) from non-infected RBCs (non-fluorescent).
  • Some experiments were conducted using non-synchronized infection state cultures while other tests were conducted using synchronized cultures.
  • Non-synchronized cultures comprised parasites that are at all stages of their life cycle.
  • synchronized cultures contained parasites that were at approximately the same stage of their infection cycle.
  • Still other experiments were performed with varying levels of cellular infection. Because the culture used to grow the malaria parasite was self-limiting due to the presence of breakdown products of RBCs, no more than about 15% of the cells were infected.
  • Fig. 20 shows an optical system having illumination of a sample plate 194 from the top side and imaging from the bottom side.
  • a laser 180 generates a first beam 181 which optionally passes through a spatial filter 182.
  • the spatial filter 182as shown includes lens 183 and aperture 184.
  • the output of the spatial filter 182 is directed to a mirror 185 and passes through the objective 186 and is imaged onto the sample plate 187.
  • the sample plate 187 and material supported on it may be imaged via an objective 188.
  • An optional mirror 189 directs radiation to an optional filter 190 through an imaging lens 191 onto the detector 192.
  • the detector 192 is coupled to an imaging system 193.
  • the imaging system 193 provides information to a control system 194 which controls various optical components of the system.
  • Fig. 30 shows a histogram of the measured escape velocities of both infected and non-infected RBCs. In the tested sample, about 5% of the cells were infected with the parasite. The measured escape velocities for the non-infected RBCs were significantly lower than the escape velocities of the infected RBCs.
  • Fig. 31 shows a comparison of the mean escape velocity for the infected and non-infected RBCs.
  • Time-of-flight measurements were also performed on a population of normal RBCs and a non-synchronized, mixed population of infected RBCs. With respect to the infected population, the population contained in excess of 70% infected cells. Time-of-flight measurements were made using a 2.6 W laser focused with a cylindrical lens.
  • Fig. 32 shows a histogram of the time-of-flight measurements for normal RBCs and infected RBCs. As seen in Fig. 32, the infected RBCs show a slight shift upward in time-of-flight (TOF) values as compared to the non-infected (control) RBCs.
  • Fig. 33 shows the comparison of the mean TOF values for the infected and control cells.
  • Figs. 34 and 35 illustrate the results of another time-of-flight experiment performed on infected and non-infected RBCs.
  • the same experimental setup was used as in the prior experiment (i.e., 2.6W laser with cylindrical lens).
  • synchronized cells were tested, h addition, in this experiment, over 95% of the cells were infected.
  • Fig. 34 shows a histogram of TOF data for the infected and non-infected cells.
  • the infected cells show a noticeable increase in TOF values as compared to their non-infected counterparts.
  • Fig. 35 shows the mean TOF values for both the infected and non-infected populations.
  • Optophoretic interrogation using time-of-flight analysis has been used to distinguish cancer cells from normal cells for breast carcinoma and skin melanoma.
  • Optophoretic interrogation can be used as a diagnostic tool to determine whether cells show the Optophoretic characteristics of cancer cells or normal cells.
  • the Optophoretic methods can also be used to detect whether the sample cells are primary or metastatic cells. These provide a relatively quick way of diagnosing whether a sample contains cancerous cells. The techniques advantageously may be used with relatively small sample sizes.
  • Experiments have been conducted on human breast carcinoma cells as well as human melanoma cells. Tumor cell lines were purchased from ATCC and, when available, their normal counterparts were matched from the same patient. Cells were grown in culture until the time of testing. Adherent cells were detached from culture flasks using trypsin and resuspended in buffer. Cells were then subject to Optophoretic interrogation.
  • Fig. 36 illustrates a histogram of the ratio of T 2 /T ⁇ plotted against the percentage of cells.
  • the cancerous cells (HS578T) exhibited a larger T 2 /T] ratio as compared to the normal cells (HS578BST).
  • Fig. 37 illustrates the mean T 2 /T ⁇ ratio for the cancerous and non-cancerous cells.
  • normal skin cells (CCD 1037) and malignant melanoma cells were subject to time-of-flight analysis.
  • Fig. 38 illustrates the ratio of T 2 /T ⁇ plotted against the percentage of cells.
  • the cancerous cells (WM 115) exhibited a larger T 2 /T ⁇ ratio as compared to the normal cells (CCD 1037). These results are consistent with the results seen in the time-of-flight data for breast carcinoma cells, namely, that the cancerous cells exhibit generally higher T 2 /T ⁇ ratios.
  • Fig. 39 illustrates the mean T 2 /T ⁇ ratio for the cancerous and non-cancerous cells. [00150] C.
  • Fig. 40 illustrates a scatter plot of the time-of-flight (TOF) ratio as a function of ti.
  • Fig. 41 illustrates a histogram of the number of particles as a function of the TOF ratio ⁇ _l ⁇ ⁇ . As seen in Figs.
  • the polystyrene beads have a larger TOF ratio as compared to the PMMA beads.
  • the beads may be used as vehicles to carry different cells or populations of cells. Interrogation may then be performed on the underlying vehicle which, in turn, allows for the characterization and possible sorting of cells adhered to the surface of the beads.
  • beads made from polystyrene may have one or more compounds bound thereto that are specific to a particular cell or cell type.
  • Other beads made from another material, i.e., PMMA may include one or more different compounds thereon that bind to a different type of cell. Characterization and sorting may be performed using the Optophoretic differences in the carrier beads.
  • the beads may be used in an agglomeration assay in which beads have specific ligands attached to their surfaces.
  • the ligand laden beads are then able to bind to cells having corresponding binding sites, hi this type of assay, a single cell might be bound to multiple beads, each bead having a different ligand.
  • These bead-ligand-cell complexes may then be analyzed optophoretically to analyze and differentiate the cells of interest.
  • Fig. 42 illustrates a scatter plot of the time-of-flight (TOF) ratio as a function of the event number for the mutant and wild type cells.
  • Fig. 43 illustrates a histogram of the percentage of yeast cells as a function of the TOF ratio t 2 /t ⁇ . As seen in Figs. 42 and 43, the mutant strain rho(0) generally has a lower TOF ratio as compared to the wild type strain.
  • HL60 cells were treated with 1% dimethyl sulfoxide (DMSO) and subject to time-of-flight testing on a diagnostic device of the type shown in Figs. 32-34.
  • DMSO dimethyl sulfoxide
  • a set of HL60 cells that were not treated with DMSO were also subject to time-of-flight testing. Measurements were made after 40 hours of treatment with DMSO.
  • DMSO is a known cell differentiation inducer.
  • Fig. 44 illustrates a scatter plot of the time-of-flight (TOF) ratio as a function of the event number for the treated and non-treated HL60 cells.
  • TOF time-of-flight
  • FIG. 45 illustrates a histogram of the percentage of cells as a function of the TOF ratio t 2 /t ⁇ . As seen in Figs. 44 and 45, the HL60 cells treated with DMSO generally had a lower TOF ratio as compared to the non- treated HL60 cells.
  • Figs. 32-34 Phorbol mystirate acetate (PMA) and ionomycin were used to activate the T cells.
  • T cells were cultured at a cell density on the order of 10 "6 cells per ml in the presence of 0.5 ⁇ g/ml PMA and 50 ng/ml ionomycin overnight in a CO 2 incubator.
  • Fig. 46 illustrates a histogram of the percentage of cells as a function of the TOF ratio t 2 /t ⁇ . As seen in Fig. 46, the stimulated T cells generally had a lower TOF ratio as compared to the unstimulated T cells.

Abstract

A device for characterizing a cell or particle includes a channel having an inlet and an outlet, the channel containing a moving fluid therein for carrying the cell or particle from the inlet to the outlet. The device includes detectors for monitorng the position of the cell or particle along the channel. The device further comprises a light source to provide an optical gradient and a control system to receive and process signals from the detector.The device can be used to characterize and sort cells based on a biological property.

Description

METHODS AND APPARATUS FOR OPTOPHORETIC DIAGNOSIS OF CELLS AND PARTICLES
Field of the Invention
[0001] The field of the invention relates generally to optical interrogation methods and apparatus used to determine a property of a cell, a population of cells, and/or cellular components, as well as particles. The methods preferably can be used to select, identify, characterize, and sort individual cells, particles, or groups of cells or particles according to the property of interest. The methods can be used in a variety of applications including, for example, drug screening applications, toxicity applications, protein expression applications, rapid clonal selection applications, biopharmaceutical monitoring applications, quality control application, biopharmaceutical enrichment applications, viral detection, bacterial drug sensitivity screening, and environmental testing applications. More particularly, the systems involved may be used to advantageously diagnose the condition or state of a cell or particle.
Related Applications
[0002] This application is related to Application Serial No. 10/240,611, filed September 12, 2002, entitled "Methods of Using Optical Interrogation to Determine a Biological Property of a Cell or Population of Cells", which is a continuation-in-part of U.S. Application Serial No. 10/053,507, filed January 17, 2002, entitled "Methods and Apparatus For Generating and Utilizing Linear Moving Optical Gradients," which itself is a continuation-in-part of U.S. Application Serial No. 09/993,377, filed November 14, 2001, entitled "Methods and Apparatus for Generating and Utilizing a Moving Optical Gradient," which itself is a continuation-in-part of U.S. Application Serial No. 09/845,245, filed April 27, 2001, entitled "Methods and Apparatus for Use of Optical Forces for Identification, Characterization and/or Sorting of Particles." This Application is also related to U.S. provisional Application Serial No. 60/377,145, filed on, May 1, 2002, entitled, "Cellular Analysis Using Infrared Moving Optical Gradient Fields". The above-identified U.S. Applications are incorporated by reference as if set forth fully herein.
Background of the Invention
[0003] In the field of biology, there often is a need to discriminate and sort cells or groups of cells based on a particular biological property of interest. For example, the discrimination and separation of cells has numerous applications in pharmaceutical drug discovery, medicine, and biotechnology. As just one example, when cells are used to produce a new protein or biopharmaceutical compound, it is desirable to select those cells or groups of cells that have the highest yield levels. Historically, sorting technologies have utilized some affinity interaction, such as receptor-ligand interactions or reactions with immunologic targets. Sorting technologies using affinity interaction, however, often are labor intensive, costly, require tags or labels, and change the nature or state of the cells.
[0004] While biological applications are of particular interest to discriminate and sort cells, similar methods and techniques can be employed in other applications ranging from industrial applications to environmental applications.
[0005] Attempts have been made to sort and characterize particles, including cells, based on the electromagnetic response properties of materials. For example, dielectrophoretic separators utilize non-uniform DC or AC electric fields for separation of particles. See, e.g., U.S. Patent No. 5,814,200, Pethig et al., entitled "Apparatus for Separating By Dielectrophoresis". The application of dielectrophoresis to cell sorting has been attempted. In Becker (with Gascoyne) et al., PNAS USA, Vol. 92, pp. 860-864, Jan. 1995, Cell Biology, in the article entitled "Separation of Human Breast Cancer Cells from Blood by Differential Dielectric Affinity", the authors reported that the dielectric properties of diseased cells differed sufficiently to enable separation of the cancer cells from normal blood cells. The system balanced hydrodynamic and dielectrophoretic forces acting on cells within a dielectric affinity column containing a microelectrode array. More sophisticated separation systems have been implemented. Yet others have attempted to use electrostatic forces for separation of particles. See, e.g., Judy et al, U.S. Patent No. 4,440,638, entitled "Surface Field-Effect Device for Manipulation of Charged Species", and Washizu "Electrostatic Manipulation of Biological Objects", Journal of Electrostatics, Vol. 25, No. 1, June 1990, pp. 109-103. Yet others have utilized various microfluidic systems to move and sort particles. See, e.g., Ramsey, U.S. Patent No. 6,033,546, entitled "Apparatus and Method For Performing Microfluidic Manipulations For Chemical Analysis and Synthesis." [0006] Still others in the field have used light to sort and trap particles. One of the earliest workers in the field was Arthur Ashkin at Bell Laboratories, who used a laser for manipulating transparent, μm-size latex beads. Ashkin's U.S. Patent No. 3,808,550 entitled "Apparatuses for Trapping and Accelerating Neutral Particles" disclosed systems for trapping or containing particles through radiation pressure. Lasers generating coherent optical radiation were the preferred source of optical pressure. The use of optical radiation to trap small particles grew within the Ashkin Bell Labs group to the point that ultimately the Nobel Prize was awarded to researchers from that lab, including Steven Chu. See, e.g., Chu, S., "Laser Trapping of Neutral Particles", Sci. Am., p. 71 (Feb. 1992), Chu, S., "Laser Manipulation of Atoms and Particles", Science 253, pp. 861-866 (1991).
[0007] Generally, the interaction of a focused beam of light with dielectric particles or matter falls into the broad categories of a gradient force and a scattering force. The gradient force tends to pull materials with higher relative dielectric constants toward the areas of highest intensity in the focused beam of light. The scattering force is the result of momentum transfer from the beam of light to the material, and is generally in the same direction as the beam. The use of light to trap particles is also sometimes referred to as an optical tweezer arrangement. Generally, utilizing the Rayleigh approximation, the force of trapping is given by the following equation:
Figure imgf000005_0001
Where Fg is the optical gradient force on the particle in the direction toward the higher intensity, r is the radius of the particle, SB is the dielectric constant of the background medium, ε is the dielectric constant of the particle, I is the light intensity in watts per square centimeter and V is the spatial derivative. Fig. 1 shows a drawing of a particle in an optical tweezer. The optical tweezer consists of a highly focused beam directed to the particle. [0008] As shown in Fig. 1, the focused beam 12 first converges on the particle 10 and then diverges. The intensity pattern 14 relates to the cross-section of the intensity of the beam in the horizontal dimension, and the intensity pattern 16 is the cross-section of intensity in the vertical dimension. As can be seen from the equation, the trapping force is a function of the gradient of the intensity of the light. Thus, the force is greater where the light intensity changes most rapidly, and contrarily, is at a minimum where the light intensity is uniform.
[0009] Early stable optical traps levitated particles with a vertical laser beam, balancing the upward scattering force against the downward gravitational force. The gradient force of the light served to keep the particle on the optical axis. See, e.g., Ashkin, "Optical Levitation by Radiation Pressure", Appl. Phys. Lett., 19(6), pp. 283-285 (1971). In 1986, Ashkin disclosed a trap based upon a highly focused laser beam, as opposed to light propagating along an axis. The highly focused beam results in a small point in space having an extremely high intensity. The extreme focusing causes a large gradient force to pull the dielectric particle toward that point. Under certain conditions, the gradient force overcomes the scattering force, which would otherwise push the particle in the direction of the light out of the focal point. Typically, to realize such a high level of focusing, the laser beam is directed through a high numerical aperture microscope objective. This arrangement serves to enhance the relative contribution from the high numerical aperture illumination but decreases the effect of the scattering force. [0010] Optical trapping methods have been employed to manipulate biological materials. In 1987, Ashkin reported an experimental demonstration of optical trapping and manipulation of biological materials with a single beam gradient force optical trap system. Ashkin, et al., "Optical Trapping and Manipulation of Viruses and Bacteria", Science, 20 March, 1987, Vol. 235, No. 4795, pp. 1517-1520. In U.S. Patent No. 4,893,886, Ashkin et al., entitled "Non- Destructive Optical Trap for Biological Particles and Method of Doing Same", reported successful trapping of biological particles in a single beam gradient force optical trap utilizing an infrared light source. The use of an infrared laser emitting coherent light in substantially infrared range of wavelengths, there stated to be 0.8 μm to 1.8 μm, was said to permit the biological materials to exhibit normal motility in continued reproductivity even after trapping for several life cycles in a laser power of 160 mW. The term "opticution" has become known in the art to refer to optic radiation killing biological materials.
[0011] The use of light to investigate biological materials has been utilized by a number of researchers. Internal cell manipulation in plant cells has been demonstrated. Ashkin, et al., PNAS USA, Vol. 86, 7914-7918 (1989). See also, the summary article by Ashkin, A., "Optical Trapping and Manipulation of Neutral Particles Using Lasers", PNAS USA, Vol. 94, pp. 4853- 4860, May 1997, Physics. Various mechanical and force measurements have been made including the measurement of torsional compliance of bacterial flagella by twisting a bacterium about a tethered flagellum. Block, S., et al, Nature (London), 338, pp. 514-518 (1989). Micromanipulation of particles has been demonstrated. For example, the use of optical tweezers in combination with a microbeam technique of pulsed laser cutting, sometimes also referred to as laser scissors or scalpel, for cutting moving cells and organelles was demonstrated. Seeger, et al., Cytometry, 12, pp. 497-504 (1991). Optical tweezers and scissors have been used in all-optical in vitro fertilization. Tadir, Y., Human Reproduction, 6, pp. 1011- 1016 (1991). Various techniques have included the use of "handles" wherein a structure is attached to a biological material to aid in the trapping. See, e.g., Block, Nature (London), 348, pp. 348-352 (1990). [0012] Various measurements have been made of biological systems utilizing optical trapping and interferometric position monitoring with subnanometer resolution. Svoboda, Nature (London), 365, pp. 721-727 (1993). Yet others have proposed feedback based systems in which a tweezer trap is utilized. Molloy, et al., Biophys. J., 68, pp. 2985-3055 (1995). [0013] A number of workers have sought to distort or stretch biological materials. Ashkin in Nature (London), 330 pp. 769-771 (1987), utilized optical tweezers to distort the shape of red blood cells. Multiple optical tweezers have been utilized to form an assay to measure the shape recovery time of red blood cells. Bronkhorst, Biophys. J., 69, pp. 1666-1673 (1995). Kas, et al., has proposed an "optical stretcher" in U.S. Patent No. 6,067,859 which suggests the use of a tunable laser to trap and deform cells between two counter-propagating beams generated by a laser. The system is utilized to detect single malignant cancer cells. Yet another assay proposed colliding two cells or particles under controlled conditions, termed the OPTCOL for optical collision. See, e.g., Mammer, Chem & Biol., 3, pp. 757,763 (1996). [0014] Yet others have proposed utilizing optical forces to measure a property of an object. See, e.g., Guanming, Lai et al., "Determination of Spring Constant of Laser-Trapped Particle by Self-Mining Interferometry", Proc. of SPIE, 3921, pp. 197-204 (2000). Yet others have utilized the optical trapping force balanced against a fluidic drag force as a method to calibrate the force of an optical trap. These systems utilize the high degree of dependence on the drag force, particularly Stokes drag force. [0015] Yet others have utilized light intensity patterns for positioning materials. In U.S. Patent No. 5,245,466, Burns et al., entitled "Optical Matter", arrays of extended crystalline and non-crystalline structures are created using light beams coupled to microscopic polarizable matter. The polarizable matter adopts the pattern of an applied, patterned light intensity distribution. See also, "Matter Rides on Ripples of Lights", reporting on the Burns work in New Scientist, 18 Nov., 1989, No. 1691. Yet others have proposed methods for depositing atoms on a substrate utilizing a standing wave optical pattern. The system may be utilized to produce an array of structures by translating the standing wave pattern. See, Celotta et al, U.S. Patent No. 5,360,764, entitled "Method of Fabricating Laser Controlled Nanolithography". [0016] Yet others have attempted to cause motion of particles by utilizing light. With a technique termed by its authors as "photophoresis", Brian Space, et al., utilized a polarized beam to induce rotary motion in molecules to induce translation of the molecules, the desired goal being to form a concentration gradient of the molecules. The technique preferably utilizes propeller shaped molecules, such that the induced rotary motion of the molecules results in translation.
[0017] Sasaki et al. discloses a method and device for controlling the flow of fine particles along a pattern formed using a scanning laser. See, Sasaki et al., Pattern Formation and Flow Control of Fine Particles By Laser-Scanning Micromanipulation, Optics Letters, Vol. 16., No. 19 (October 1, 1991). In one demonstration in Sasaki et al., polystyrene latex particles were distributed on in a circular pattern of laser light. A driving force was imparted on the particles by repetitive scanning of the trapping beam at a repetition rate of 15 Hz in a clockwise manner. It was observed that all the particles moved together in an orderly fashion around the circular laser pattern. Experiments were also conducted that varied the repetition rate of the trapping beam. The investigators found that particle flow rates became slower as the scan rate increased. [0018] Various efforts have been described relating to cellular response. By way of example, Ransom et al. U.S. Patent No 6,280,967 entitled "Cell Flow Apparatus and Method for Real- Time (Sic.) of Cellular Responses" describes an apparatus and method for the real-time measurement of a cellular response of a test compound or series of test compounds on a flowing suspension of cells. The cells and test compound or compounds are combined and then flowed through a detection zone. Typically, a label is detected indicating the response. Libraries of compounds are described. As stated, generally the detectable event requires a label. [0019] In Zborowski et al. U.S. Patent No. 5,974,901, entitled "Method for Determining Particle Characteristics", and U.S. Patent No. 6,082,205, entitled "System and Device For Determining Particle Characteristics", methods and apparatus are described for determining at least one of a plurality of particle physical characteristics. Particularly, the particle characteristics may include particle size, shape, magnetic susceptibility, magnetic label density, charge separation, dielectric constant, and derivatives thereof. In one aspect, a uniform force field, such as a constant, uniform magnetic force field is generated, the particle is subject to that constant force field, and the velocity determined by observing the particle at multiple locations. Variations are described, such as for determining the position of the particle, though the force field is typically described as being constant. In another aspect, a pre-determined force field magnitude and direction is applied to a particle and multiple digital images are analyzed with specified other components to characterize the particles.
[0020] Various researchers have attempted to combine microfabricated devices with optical systems. In "A Microfabricated Device for Sizing and Sorting DNA Molecules", Chou, et al., PNAS USA, Vol. 96, pp. 11-13, Jan. 1999, Applied Physical Sciences, Biophysics, a microfabricated device is described for sizing and sorting microscopic objects based upon a measurement of fluorescent properties. The paper describes a system for determining the length of DNA by measuring the fluorescent properties, including the amount of intercalated fluorescent dye within the DNA. In "A Microfabricated Fluorescence-Activated Cells Sorter", Nature Biotechnology, Vol. 17, Nov. 1999, pp. 1109-1111, a "T" microfabricated structure was used for cell sorting. The system utilized a detection window upstream of the "T" intersection and based upon the detected property, would sort particles within the system. A forward sorting system switched fluid flow based upon a detected event. In a reverse sorting mode, the fluid flow was set to route all particles to a waste collection, but upon detection of a collectible event, reversed the fluid flow until the particle was detected a second time, after which the particle was collected. Certain of these systems are described in Quake et al., PCT Publication WO 99/61888, entitled "Microfabricated Cell Sorter".
[0021] Yet others have attempted to characterize biological systems based upon measuring various properties, including electromagnetic radiation related properties. Various efforts to explore dielectric properties of materials, especially biological materials, in the microwave range have been made. See, e.g., Larson et al., U.S. Patent No. 4,247,815, entitled "Method and Apparatus for Physiologic Facsimile Imaging of Biologic Targets Based on Complex Permittivity Measurements Using Remote Microwave Interrogation", and PCT Publication WO 99/39190, named inventor Hefti, entitled "Method and Apparatus for Detecting Molecular Binding Events".
Summary of the Invention
[0022] A device and associated methods are described for characterizing a cell or particle. The systems generally include a channel having an inlet and an outlet, the channel containing a moving fluid therein for carrying the cell or particle from the inlet to the outlet. The device also includes a detector (or multiple detectors) for detecting the presence of a cell or particle along a portion of the channel. The detector includes at least a first detecting position, a second detecting position, and a third detecting position. The device further includes a light source providing an optical gradient disposed within the channel and between the second and third detecting positions. A control system is coupled to the detector to receive and process detected signals from the detector.
[0023] In operation, the amount of time that a cell or particle takes to flow through a first distance (i.e., its time-of-flight) is measured. The particle is then flowed past a second, downstream distance in the presence of an optical gradient and its time-of-flight is measured. A comparison of the measured time-of-flights for the first and second distances is used to characterize the cell or particle. The optical gradient serves as an Optical speed-bump', serving to slightly retard the progress of the cell or particle in an amount related to the degree of interaction between the particle and the optical gradient. In the case of a biological particle such as a cell, the method can be used to characterize cells based on one or more biological properties of the cell. Optionally, the characterization information may be utilized to further sort cells or particles based upon an observed parameter. [0024] A variety of detection systems are described. Within the realm of optical detection systems, coherent light may be used for illumination of the particle. In one embodiment, a pattern generator disposed upstream of the particles selectively illuminates the particle. A detector array determines particle position as a function of time. In an alternative embodiment, a system utilizing coherent light scans a beam over the channel. A detector determines the cell or particle positioning as a function of time. Incoherent light may be used for illumination. Detection may be by any number of techniques, such as through the use of a mask and detector array, or by use of a line camera. Electrical detection of the cell or particle position may be utilized, such as where an impedance detection is utilized.
[0025] In a preferred embodiment of the system, an additional detection beam may be utilized. Preferably, the beam is directed axially along the channel and an additional detector is located upstream from the other detectors. By utilizing additional detectors, the various detectors may be optimized to determine different detectable characteristics. The initial detector may be utilized to activate or otherwise tune the remaining detectors. In addition, the initial detector may be used as a gating detector in the sense that it detects the presence of an incoming cell or particle. Preferably the gating detector has the capability to detect whether the incoming test subject (i.e. cell or particle) is in a condition for measurement. For example, if cells are being analyzed on the system, the detector can determine and reject a sample if it appears that the cells are clumped together or otherwise unrepresentative of the cells or particles of interest. [0026] It is an object of this invention to provide a simple, inexpensive, scalable system for Optophoretic diagnostics of a cell or particle. It is a further object of the invention to provide a system that uses low volume, substantially constant velocity flow regulation coupled with optical measurement and interrogation components to serve as a diagnostic device. The device has applications in a wide variety of diagnostic applications including, but not limited to, cancer diagnostic applications and infectious disease diagnostic applications.
Brief Description of the Drawings [0027] Fig. 1 is a graphical depiction of optical intensity patterns for a prior art optical tweezer system, showing both the focus beam, a particle and the cross-section of intensity of the beam.
[0028] Fig. 2 shows a plan view of a time-of-flight system.
[0029] Fig. 3 shows a generalized block diagram of a microfluidic detection system. [0030] Fig. 4 is a block diagrammatic view of one apparatus and associated method for detection, namely one in which coherent light detection is utilized.
[0031] Fig. 5A shows a plan diagram of a coherent light detection system utilizing scanning detection.
[0032] Fig. 5B graphically shows the operation of scanning system. [0033] Fig. 6A shows a plan view of a system utilizing an incoherent light detection system including a detector mask.
[0034] Fig. 6B shows a side plan view of a detector mask.
[0035] Fig. 7 shows a plan view of system utilizing incoherent light for detection along with a line camera. [0036] Fig. 8A shows a plan view of an electronic detection system.
[0037] Fig. 8B shows close up view of the charmel and its associated electrodes.
[0038] Fig. 9 shows a side view of a gravity-based time-of-flight system.
[0039] Fig. 10 is a plan view of a microfluidic channel based detection system.
[0040] Fig. 11 is a system block diagram of various subsystems within the system. [0041] Fig. 12 depicts the optical subsystem for one implementation of a time-of-flight system.
[0042] Fig. 13 shows a combined block diagram and processing functionality and software for the acquisition subsystem.
[0043] Fig. 14 shows a flow chart of one possible implementation of the software subsystem.
[0044] Fig. 15 depicts the forces on a particle in a time-of-flight system.
[0045] Fig. 16 depicts the optical force on a typical 10 micron cell by a row numerical aperture (NA) laser line. [0046] Fig. 17 is a graph of time delay as a function of escape velocity normalized to flow velocity for a 10 micron bead as simulated.
[0047] Fig. 18 shows the data of Fig. 17 plotted in a log-log format.
[0048] Fig. 19 is a depiction of an optical system used to perform line scan and fast scan analysis on samples.
[0049] Fig. 20 depicts a preferred detection scheme utilizing multiple detectors and a detection laser.
[0050] Fig. 21 illustrates a perspective view of a preferred embodiment of a time-of-flight device. [0051] Fig. 22 illustrates another perspective view of the preferred embodiment of the time- of-flight device shown in Fig. 21.
[0052] Fig. 23 schematically illustrates the preferred embodiment shown in Figs. 21 and 22.
[0053] Fig. 24 schematically illustrates the fluidics used in one preferred embodiment of the invention. [0054] Fig. 25 schematically illustrates a preferred embodiment of the flow pump used to create a low, constant flow rate.
[0055] Fig. 26 illustrates a preferred embodiment of the flow pump used to create a low, constant flow rate.
[0056] Fig. 27 illustrates one embodiment of a massively parallel system. [0057] Fig. 28 illustrates yet another embodiment of a massively parallel system.
[0058] Fig. 29A illustrates a device capable of both characterizing and sorting a cell or particle.
[0059] Fig. 29B illustrates a preferred device and method for sorting cells or particles in the device shown in Fig. 29A. [0060] Fig. 29C illustrates another preferred device and method for sorting cells or particles in the device shown in Fig. 29A.
[0061] Fig. 30 is a histogram of the measured escape velocities of Plasmodium-infected and non-infected red blood cells.
[0062] Fig. 31 shows a comparison of the mean escape velocity for Plasmodium-infected and non-infected red blood cells .
[0063] Fig. 32 is a histogram of time-of-flight measurements for normal red blood cells and
Plasmodium-infected red blood cells. [0064] Fig. 33 shows a comparison of the mean time-of-flight values for the infected and control cells of Fig. 32.
[0065] Fig. 34 is a histogram of time-of-flight measurements for normal red blood cells and
Plasmodium-infected red blood cells. The cells tested in this experiment were synchronized. [0066] Fig. 35 shows a comparison of the mean time-of-flight values for the infected and control cells of Fig. 34.
[0067] Fig. 36 illustrates a histogram of the ratio of T2/Tι plotted against the percentage of cancerous and non-cancerous cells from breast tissue.
[0068] Fig. 37 shows a comparison of the mean T2/Tι ratio of the cancerous and non- cancerous cells of Fig. 36.
[0069] Fig. 38 illustrates a histogram of the ratio of T2/Tι plotted against the percentage of cancerous skin cells and non-cancerous skin cells.
[0070] Fig. 39 shows a comparison of the mean T2/Tι ratio of the cancerous and non- cancerous cells of Fig. 38. [0071] Fig. 40 is a scatter plot of the T2/Tι ratio as a function oft] for polystyrene beads and
PMMA beads.
[0072] Fig. 41 is a histogram of the number of particles as a function of T2/Tι ratio for experimental data shown in Fig. 40.
[0073] Fig. 42 is a scatter plot of the T2/Tι ratio as a function of event number for 24657 rho+ (wild type) yeast and MYA-1133 rho(0) (mutant) yeast.
[0074] Fig. 43 is a histogram of the percentage of cells as a function of T2/Tι ratio for experimental data shown in Fig. 42.
[0075] Fig. 44 is a scatter plot of the T2/Tι ratio as a function of event number for treated and non-treated HL60 cells. The treated HL60 cells were treated with 1% DMSO. [0076] Fig. 45 is a histogram of the percentage of cells as a function of T2/Tι ratio for experimental data shown in Fig. 44.
[0077] Fig. 46 is a histogram of the percentage of unactivated and activated T cells as a function of T2/T] ratio.
Detailed Description of the Invention Definitions
[0078] The following definitions are provided for an understanding of the invention disclosed herein. [0079] "Biological Property" means a distinct phenotype, state, condition, or response of a cell or group of cells, for example, whether a cell is diseased, has been infected by a virus, the degree to which a cell expresses a particular protein, the stage in the cell cycle a particular cell is presently at, whether the cell is affected by the presence of a chemical compound, a particular phenotype of the cell, whether a ligand is bound to the surface of a cell, cytoskeletal changes in the cell, whether a cell is decorated with antibodies, the presence or absence of a cellular component (e.g., an organelle or inclusion body), a change in one or more cellular components, the toxicity of chemical compounds, a physical property of a cell or population of cells, a response of a cell or population of cells to an external stimulus, cellular motility, membrane fluidity, state of differentiation, viability, size, osmolarity, adhesion, secretion, cell/cell interactions, activation, and cell growth.
[0080] "Determining" is meant to indicate that a particular phenotype, state, condition, or response is ascertained. [0081] "Dielectric constant" is defined to be that property which determines the electrostatic energy stored per unit volume for unit potential gradient. (See, e.g., the New IEEE Standard Dictionary Of Electrical And Electronics Terms, ©1993).
[0082] The "escape velocity" is defined as the minimum speed at which an interrogated cell or particle no longer tracks the moving optical gradient. [0083] The "optical dielectric constant" is the dielectric constant of a particle or thing at optical wavelengths. Generally, the optical wavelength range is from 150 A to 30,000 A.
[0084] An "optical gradient field" is an optical pattern having a variation in one or more parameters including intensity, wavelength or frequency, phase, polarization or other parameters relating to the optical energy. When generated by an interferometer, an optical gradient field or pattern may also be called an optical fringe field or fringe pattern, or variants thereof.
[0085] A "moving optical gradient field" is an optical gradient field that moves in space and/or time relative to other components of the system, e.g., particles or objects to be identified, characterized, selected and/or sorted, the medium, typically a fluidic medium, in contact with the particles, and/or any containment or support structure. [0086] An "optical scattering force" is that force applied to a particle or thing caused by a momentum transfer from photons to material irradiated with optical energy. [0087] An "optical gradient force" is one which causes a particle or object to be subject to a force based upon a difference in dielectric constant between the particle and the medium in which it is located.
[0088] "Optophoresis" or "Optophoretic" generally relates to the use of photonic or light energy to obtain information about or spatially move or otherwise usefully interact with a particle.
[0089] "Optophoretic constant" or "optophoretic signature" or "optophoretic fingerprint" refer to the parameter or parameters which distinguish or characterize particles for optical selection, identification, characterization or sorting. [0090] "Separation" of two objects is the relative spatial distancing over time of a particle from some other reference point or thing.
[0091] "Sorting" involves the separation of two or more particles in a meaningful way. [0092] Fig. 2 shows a plan view of a time-of-flight system. A channel 20 is defined by an inlet 22, an outlet 24 and first and second sidewalls 26. Preferably, the sidewalls 26 are linear, and substantially parallel to each other. The outlet 24 has a width D. For biologic diagnostics, such as cell diagnostics, D may be on the order of substantially 60 microns or smaller. Typically, D would exceed the size of the largest expected cell or particle to flow through the channel 20. The channel 20 contains a moving fluid therein, shown by arrow A in Fig. 2, which carries the cells or particles 36 from the inlet 22 to the outlet 24. Particle as used herein refers to any type of small body and includes, as an example, spores, pollen, and particulate matter such as airborne or other environmental contaminants. These include airborne as well as waterborne contaminants. Preferably, the moving fluid flows within the channel 20 at a constant flow rate. The flow rate of the fluid within the channel 20 is preferably controllable. A flow rate is chosen such that the flow rate of the fluid exceeds the escape velocity of the cells or particles 36 transported through the channel 20, that is the flow rate is chosen such that cells or particles 36 do not get "stuck" on the optical gradient 38 (described in detail below) . [0093] The system includes multiple, preferably three, detecting positions, 31, 32 and 33. The detecting positions 31, 32, 33, may be associated with detection of times, e.g., ti, t2, t3. The times may be absolute, relative or elapsed. The two outermost detecting positions, i.e., 31 and 33 in this example, are separated by a distance L. A representative length L when detecting biological particles would be substantially 200 microns or less. The difference between the first two detecting positions ti, t2, define a time interval Tl. Tl may also be associated with a first detection zone corresponding to the motion of the particle 36 through the zone Tl. The second and third detecting positions t2, t3, define a second detection zone T2. The second detection zone also corresponds to the time difference between t2, t3. In one preferred embodiment, the distance between t213 is equal to the distance between ti t2.
[0094] In operation, the cell or particle 36 flows from the inlet 22 through the channel 20 through the outlet 24. For biological detection purposes, representative particle speeds are from substantially 50 to substantially 200 microns per second. As the cell or particle 36 flows through the first detection zone Tl, the time-of-flight through that zone is measured, that is the time it takes for the cell or particle 36 to move from position ti to t2. An optical gradient 38 is disposed within zone T2. Preferably, the optical gradient 38 is substantially linear and has a thickness in the fluid flow direction which is substantially less than the transverse dimension. Preferably, the width is less than substantially 10% of the transverse length of the optical gradient 38. In addition, the optical gradient 38 is disposed within a portion of the channel 20 such that the optical gradient 38 is generally orthogonal to the direction of the fluid flow. The optical gradient 38 is preferably formed using a coherent light source such as a laser that is passed through a cylindrical lens. Alternatively, the optical gradient 38 may be formed using a scanning laser system.
[0095] As the moving cell or particle 36 moves through zone T2, it will intercept the optical gradient 38. If the optical gradient 38 has no effect on the moving particle 36, T2 equals Tl, assuming the physical difference in detecting positions is equal for zone 1 and for zone 2. If the moving cell or particle 36 does optically interact with the optical gradient 38, the cell or particle 36 will typically be slowed or retarded in its transit through zone T2. Accordingly, T2 would be greater than Tl, assuming the detection positions are uniformly spaced. [0096] The optical gradient 38 may be said to be "static" relative to the underlying device, such as the device that defines the channel 20. However, the relative motion of the cell or particle 36 and the optical gradient 38 provide the discriminating force within the system.
[0097] In operation, the cell or particle 36 generally moves at a speed in the range from about 50 to about 200 microns per second. For biological applications, the detection spacing, that is the distance between adjacent detecting positions, is typically on the order from approximately 20 to approximately 50 microns. While shown with three detection positions, the system may use more detection positions, or different types of detectors, as desired. In operation, the throughput of the system may be in the range of approximately 500 to about 2,000 particles per hour per channel 20. [0098] Fig. 3 shows a generalized block diagram of a microfluidic detection, preferably diagnostic, system 39. The substrate 40 containing the channel 20 receives the moving cell or particle 36. An illumination system 42, preferably including a laser as the light source, provides the optical gradient 38. A detection system 44 is operatively positioned to detect the position of the cell or particle 36 at multiple, typically three or more, locations. A control system 46 controls the illumination system 42 via the communication path 48. The output of the detection system 44 is coupled to the control system 46 via the communication path 50. The control system 46 serves to receive and process the detected signals from the detection system 44. A display 54 optionally depicts the detected intensity of the particle as it passes the sections of the detection system 44. In this regard, the display 54 can show the amount of time it takes the cell or particle 36 to pass through zones Tl and T2. The display 54 may further display a ratio using the values of Tl and T2 (i.e. T1/T2 or T2/T1).
[0099] Various detection system may be utilized in connection with such systems. While not meant to be limiting, various exemplary detection systems will be described. Figs. 4, 5A, 6 and 7 show optical detection systems. More particularly, Fig. 4 is a multi-element detection system utilizing coherent light. Fig. 5A is a scanning detection system using coherent light. Fig. 6 is a detector system using a detector mask, utilizing incoherent light. Fig. 7 is a line camera system utilizing incoherent light. Finally, Fig. 8 shows an electronic detection system, specifically utilizing an impedance detector. [00100] Fig. 4 is a block diagrammatic view of one apparatus and associated method for detection, namely one in which coherent light detection is utilized. A flow cell 60, such as described in connection with Fig. 2 is adapted to receive fluid flow in the direction y, where that fluid includes cells or particles 36 for analysis. An illumination system 62 includes a pattern generator 64 and a light source 66. The pattern generator 64 may be implemented in any number of formats. For example, a VCSEL (vertical cavity surface emitting laser) array may be directed towards the system 60. Alternately, diffractive optical elements may be used.. In yet other implementations, light modulators, such as MEMs mirror systems may be utilized. In yet another implementation, light modulator implementation, an AO (Accousto-Optical) modulator may be utilized. However implemented, the illumination system 62 serves to generate patterned illumination for the detection of the cell or particle position, timing or speed. A laser bump pattern generator 68 receives the output of a laser 70 to generate the optical gradient (item 38 in Fig. 2). The pattern generator 68 may be formed from any variety of technologies, e.g., a scanning system, such as an oscillating mirror scanning system, or through the use of diffractive optical elements. As shown, an optical system 72, such as a beam splitter, may be utilized to combine the light from the pattern generator 64 as well as the laser bump pattern generator 68 using a lens 74. The illumination source 66 may be relatively low powered. For example, the illumination laser diode may be an 8mW 635 nm laser making the light in the visible range. The laser 70 for generating the optical gradient may be of relatively higher power, such as a 1W laser diode strip. The laser 70 may optionally be in the infrared spectrum, such as 1064 nm. As the particle 36 passes in front of the patterned illumination, modified light passes out from the device 60. Optionally, a beam splitter 76 may be utilized to divert the light to imaging optics which may then be detected with camera 78, such as a CCD camera. A detector array 80 receives the light path from the device 60, after passing tlirough appropriate focusing optics. The detector array may be implemented in any number of technologies. For example, a PIN array may be utilized. The output of the detector array 80 is optionally provided to a signal processor 82, such as a digital signal processor (DSP). A time interval measurement unit 84 detects the time intervals (see, e.g., Tl and T2, or ti, t2, and t3, Fig. 2). A timing diagram 86 may be displayed, such as through a graphical display or a printed display. As shown in Fig. 4, the left most curve depicts the number of counts (i.e., particles 36) in the interval Tl, whereas the right most curve depicts the number of counts in the interval T2. As can be seen, the average time to traverse region T2 was greater than the time to traverse region Tl. [00101] The camera 78 may be utilized for any functions, including monitoring and alignment. The signal processing system 82 optionally includes demodulation, differential peak detection and digitizing. Optionally, the illumination system 62 may include modulation to enhance the signal to noise ratio (SNR) for detection. [00102] Fig. 5A shows a plan diagram of an coherent light detection system utilizing ' scanning detection. The description for many components in Fig. 5A is the same as that for Fig. 4, and accordingly those components have been similarly numbered. In Fig. 5A, a coherent light source 90, such as a laser diode, provides illumination of the system 60. The output of the illumination system 90 is scanned over at least a portion of the channel device 60. The scanning device 92 may be, for example, a scanning mirror system, preferably an x, y scanning system such as a system using two rotating mirrors having non-parallel axis. The scanning system would typically scan in a raster scan fashion, such as shown in Fig. 5B. The spacing between the scans is exaggerated in Fig. 5B for purposes of illustration. The output of scanning device 92 is passed through the associated optics to scan the operative portions of the channel device 60. The output of that scan is then imaged upon a detector 94. Optionally, a beam splitter 76 may direct the output illumination to an imaging camera 78, such as a CCD camera. The detector 94 output is then provided to a signal processor 82, which in turn provides its output to an input for the time correlation and interval measurement unit 96. The time correlation and interval measurement unit 96 receives a clock signal input, such as provided from a time base 98. The time base 98 may also control the operation of the scanning device 92. Overall system control may be achieved through such an integrated arrangement. [00103] Fig. 6A shows a plan view of a system utilizing an incoherent light detection system including a detector mask. Fig. 6B shows a side plan view of a detector mask 102. Similar components disclosed in Figs. 4, 5A, 6A, 7, and 8 are shown having identical element numbers and unless indicated otherwise, operate in the same manner. An illumination source 100, preferably a visible illumination lamp provides a source of light for detection. The light impinges upon the particle to be analyzed, whether passing through focusing optics or not. The light output from the device 60 is then directed to the mask 102. The mask 102 is placed before the detector array 80. As shown, the mask 102 includes three apertures 104. Generally, the apertures are elongate, preferably rectangular, and preferably evenly spaced one from another. The apertures 104 define detection windows for the cell or particle 36 passing through the device 60. In one preferred embodiment, the width of the apertures 104 are about 1.5μm. Generally, the width of the apertures 104 should be no more than about one-half of the size of the cell or particle 36. In addition, the width of the aperture 104 is chosen such that enough light passes there through to produce a signal strong enough to be picked up by the detector. [00104] Fig. 7 shows a plan view of system utilizing incoherent light for detection along with a line camera. Incoherent light from a source 100 illuminates the cell or particle 36 in the device 60 and the output light is then imaged upon the line camera 110. The output of the line camera 110 is provided to an analysis engine 112. The analysis engine 112 may calculate the desired parameters or properties, such as velocity, acceleration, deceleration, position and/or time intervals. The output of the analysis engine may optionally be displayed on display 86. [00105] Figs. 8A and 8B show a plan view of an electronic detection system. More particularly, this system utilizes impedance measurement for detection. Detection electrode 120, 122 and 124 are spaced at detection positions PI, P2 and P3, respectively. Fig. 8B shows a plan view of a particle 36 passing through the channel device 60 having three detection electrodes, 120, 122, and 124. An electromagnetic field is set up between the individual electrode pairs, e.g., 120. The impedance of the space between the electrode pairs changes as a function of the particle 36 position. The impedance detector and signal processing system 126 is electrically connected to the electrode pairs, e.g., 120. The profile for the cell or particle 36 as it moves through the detector then is analyzed for the desired property, such as the interval for zones 1 and 2, or the time difference between the two zones. [00106] Fig. 9 shows a side view of a gravity-based time-of-flight system. In this regard, an external source of fluid flow such as a pump or the like is not needed. A surface 130 is angled relative to horizontal, such that there is a component of gravitational force exerted on the particle or cell 131 to cause it to move down the surface 130. Detectors 132, 134, 136 are disposed to detect the particle 131 as it passes the detectors. A time difference Δti is detected between the first two detectors 132, 134. An optical gradient 138 comprising an optophoretic "speed bump" is disposed between the second detector 134 and the third detector 136. A time difference is measured between the second detector 134 and the third detector 136, and is designated Δt2. By analyzing the relative time differences, as described previously, the particle may be characterized. [00107] It should be understood that with respect to the systems and methods described herein that rely an external source of fluid flow such as a pump, the channel 20 (or 142 as described below) may be oriented in any number of orientations including, for example, horizontal and vertical orientations. [00108] Fig. 10 is a plan view of a microfluidic channel based detection system. A particle or cell 140 flows through the channel 142 in the direction of the arrow. A first light source 144, such as a light emitting diode, is directed to a first detector 154, such as a photo detector to detect the time at which the particle or cell 140 reaches a first position. A second light source 146 illuminates the channel 142 and the second detector 156 determines when the particle or cell 140 has reached the second position. Finally, a third source 148 illuminates the charmel 142 and third detector 158 detects the crossing of the particle or cell 140 at a third position. An optical gradient 150 provides a potential force against the particle or cell 140. By comparison of the time differences Δti and Δt2, the particle may be characterized. For cellular applications, the channel would typically be in the range from substantially 10 microns to substantially 100 microns in both width and depth. [00109] Fig. 11 is a system block diagram of various subsystems within the system. The optical subsystem 160 is depicted using the general structure shown in Fig. 6. However, the description of the system and the various subsystems applies to all of the various methods, especially optical subsystems, described herein. A computer control system 162 interfaces with other subsystems, including the electronics driver subsystem 164, the microftuidics subsystem 166 and the electronics acquisition system 168. As shown, various connections or buses are provided as required between the various subsystems, such as from the computer control subsystem 162 to the microfluidics subsystem 166, and from the electronics driver subsystem 164 to the microfluidics subsystem 166. A power subsystem 170 connects to all subsystems. [00110] Fig. 12 depicts the optical subsystem for one implementation of a time-of-flight system.
[00111] Fig. 13 shows a combined block diagram and processing functionality and software for the acquisition subsystem. An optical subsystem 200 receives optical input from a white light source 202 and a laser 204 to generate the optical gradient. Detection may consist of detectors 208 to detect the time of crossing of the particle at a predefined position. An imaging camera 206 may be utilized for general system imaging, alignment or to otherwise determine the location of the particle. A power system 210 is connected to all necessary electrical components. Fig. 13 shows two possible detection, analysis and display systems. Under solution A, a pattern identification step 212 may be utilized. Analog circuitry may be implemented or various digital techniques, such as digital signal processors (DSPs) may be utilized. The analysis section 214 receives the output of the pattern identification 212 and optionally the direct output of the detector 208, which is received by the data acquisition subsystem 216. A data processing system 218 performs various functions, including optionally statistical analysis. The various functionalities of system 214 may be performed under control of a personal computer, such as operating in a windows based environment. [00112] Solution B depicts the functionality 230 in which the output of the detectors 208 is provided to a data acquisition functionality 232 and/or a pattern identification subsystem 234. If the data acquisition functionality 232 and pattern identification functionality 234 are present, they may be performed in either order. As depicted, an analog-to-digital (A D) converter 236 is provided to convert acquired analog data to digital data. Optionally, an FPGA platform may be utilized. The acquired data is then subject to data processing step 238, which optionally includes statistical analysis. Ultimate display to the user may be under control of a graphical user interface or peripheral interface 240. In this embodiment, the system may be under microprocessor control 240.
[00113] Fig. 14 shows a flow chart of one possible implementation of the software subsystem. [00114] Fig. 15 depicts the forces on a cell or particle in a time-of-flight system. TOF (Time-of-flight) system measures the time delay of flowing particles or cells by a laser beam in micro fluidic environment. The time delay according to optophoretic property is used to analyze the biological differences between populations. The TOF instrument is aiming for low cost and diagnostic applications. For a particle or cell flowing thought an optical field inside a micro- channel, the forces applied are shown in Figure 15. The force equation is: d2x dx m~^_- - Foptical lateral(x) ~Fdmg(— -Vf(z),z) Equation l
Assume: z = z0 constant before particle interacts with laser beam (z0is the middle of the channel), and gravity force is balanced with buoyant force. Then: dx dx Fdmg(~ -Vf(z0),Zϋ) = b(— -Vf) Equation 2
where b is the drag coefficient that depends on the radius and shape of the object and the viscosity ("stiffness") of the medium. For a sphere of radius r, the drag coefficient is b = 6π- rη Equation 3
[00115] where η is the viscosity of medium in g / cm s. Due the boundary effect, the flow velocity decreases upon the distance to the boundary of the micro-channel. Both GLMT and geometrical simulations find that optical forces FopticalJalera!(x) and Foplicαl ^(x) on a cell by low numerical aperture (NA) laser beam are typically as shown in Figure 16. Fig. 16 depicts the optical force on a typical 10 micron cell by a 0.1 (NA) laser line.
[00116] Simulations have been performed since Equation 1 is difficult to solve because F op ed lateral (x) ^s m nonlinear and in complicated form. Several numerical simulations have been conducted as shown in Figures 17 and 18. These simulations have not considered the impact from the axial optical force.
[00117] Fig. 17 is a graph of time delay as a function of escape velocity normalized to flow velocity for a 10 micron bead as simulated. As expected, as the flow velocity approaches the escape velocity, the time of delay increases. In addition, Fig. 17 shows that lower flow velocities produce larger time delays for a given escape velocity/flow velocity. Fig. 18 shows the data of Fig. 17 plotted in a log-log format. The devices and methods described herein may take advantage of the interaction between flow velocity and delay time to optimize the sensitivity of the device. For example, as seen in Fig. 17, the time delay increases as the flow velocity approaches the particle's escape velocity. This fact may be exploited by modifying the flow velocity to a level just above the escape velocity of the particle to thereby create a large degree of time delay. In addition, Fig. 17 shows that the slower flow velocities produce the largest time delays. Consequently, slower fluid flow rates will produce more noticeable changes in the measured travel times in the detection zones (i.e., zone T2 as compared to Tl). Of course, slower flow rates will reduce the overall throughput of the device and may not be desirable in certain applications.
[00118] Regardless the accuracy and parameters of the simulations, the non-linearity of TOF system is clear. These simulations are supported by experimental results from fast scan instruments, which have strong similarities to the TOF system. The TOF system is sensitive to optophoretic differences in biological cells. The biological sensitivities of TOF system have been demonstrated.
[00119] Fig. 20 shows a preferred signal detection scheme utilizing multiple detectors. Fig. 20 includes components similar to those described in Fig. 2. Fig. 20 includes an additional detection position 280. A detection laser beam 282 is directed to the system in a direction generally parallel to the channel 20. Preferably, the detection laser beam 282 is a low power visible laser beam. The detectors may be any of the type described generally herein. The added detector may be optimized for properties other than that which the other particle detectors are optimized for. For example, the detectors may be optimized based on optical arrangement, such as optical focusing, and/or the geometry of the detector mask, and/or by electronic processing, such as by dedicated filtering and/or the use of threshold circuits. In this way, the signal from the additional detector may be more representative of particle physical properties, such as particle physical size. In contrast, the first detectors may be optimized for detecting the particle physical position. In a preferred embodiment, the additional detector may be physically placed upstream of the other detection positions. In this arrangement, the additional detector may provid trigger selection for the time-of-flight signal acquisition based upon a particles detected property or properties. In this regard, the additional detector acts as a gating detector that indicates to the other components of the device, i.e., the detectors that measure the time intervals Tl and T2 that a cell or particle is about to pass through the detection zones. Throughput and accuracy of signal acquisition generally increases by adding an additional detector. Optionally, yet additional detectors having different optical and/or electrical arrangements may provide more information regarding the particle measurement. Such additional properties or parameters might include, e.g., light scatter, absorption by the particle, size, autofluorescence, fluorescence, luminescence, and other reporter-based properties. [00120] Figs. 21 and 22 show a preferred embodiment of a time-of-flight device 200 that employs an additional detector as is described above. The device 200 includes optical, mechanical, and fluidic components that are mounted on a base plate 202. With reference to Fig. 32, the device 200 includes a infrared (IR) laser 204 that outputs a coherent beam of infrared light. Preferably, the IR laser 204 has a wavelength in the range of about 780 nm to about 1064 nm. Two preferred wavelengths within this range include 808 nm and 1064 nm. The IR laser 204 creates the optical gradient (e.g., optical gradient 38 as is shown in Fig. 2) that is used to differentially slow cells or particles passing there through. The output of the IR laser 204 passes through a laser collimation lens 206 and is directed against two mirrors 208. The light then passes through a IR cylindrical lens 210 and into an IR beam splitter 212. The light passing through the IR beam splitter 212 is then directed through a focusing lens 213 into a microfluidic mounting system 214. The microfluidic mounting system 214 includes at least one channel (not shown) therein through which the cells or particles pass. The IR optical gradient is disposed generally perpendicular to the direction of flow through the channel contained in the microfluidic mounting system 214.
[00121] The time-of-flight device 200 also includes a visible (VIS) laser 216 that outputs a coherent beam of light. Preferably, the visible laser 216 comprises a laser diode operating at around 635 nm. The visible laser beam acts as a detection laser as is described in more detail above and as shown in Fig. 20. The visible laser beam is reflected off another mirror 218 and passed through two lenses 220, 222. The visible laser beam is then reflected off a mirror 224 and passed through a filter 226. The visible laser beam then passes through a white light beam splitter 228 and into a cylindrical lens 230. The visible laser beam then passes through the focusing lens 213 and into the microfluidic mounting system 214. The visible laser beam produces a line of light that is disposed within the channel (not shown) contained within the microfluidic mounting system 214 in a direction that is generally parallel to the direction of fluid flow. This detection laser is shown in detail in Fig. 20 (detection laser 282). [00122] After passing through the microfluidic mounting system 214, both the IR laser beam and the visible laser beam pass through a collection lens 232 and into a beam splitter 234. One output of the beam splitter 234 is then directed through an imaging lens and filter 236. The light passing through the imaging lens and filter 236 is then directed through a mask M2 disposed in front of a first detector 238. The mask M2 preferably includes a single window therein as is shown, for example, in Fig. 11. The first detector 238 is preferably used as an event or gating detector as is described above and shown in Fig. 20.
[00123] The other output of the beam splitter 234 is directed to an imaging lens and filter 240 and into a mirror 242. The reflected light is then directed through a mask Ml disposed in front of a second detector 244. The mask Ml preferably includes three windows therein as is shown, for example, in Fig. 11. The second detector 244 is preferably used to determine the amount of time it takes a cell or particle to travel from a first detecting position ti to a second detecting position t2 and the amount of time it takes the same cell or particle to travel from the second detecting position t2 to a third detecting position t3. The three windows in the mask Ml correspond to the detecting positions ti, t2, and t3.
[00124] The device 200 may also include an optional camera 246 such as a CCD camera that is used to image the microfluidic mounting system 214. The optional camera is used to calibrate the device 200. If the optional camera 246 is used, a white light source 248 is preferably used to provide additional light to enhance the imaging of the microfluidic mounting system 214 and the channel contained therein.
[00125] Fig. 33 illustrates the underside of the device 200. The device 200 includes a power input 250 that is preferably connected to a conventional 110 VAC power source. The device 200 also includes a computer interface 252 that allows data communication between the device 200 and an external computer (not shown). Preferably, the computer interface 252 is a 68 pin high-speed connection. Fig. 33 also illustrates the laser driver 254 for the IR laser 204. Data acquisition electronics 256 are included in a printed circuit board preferably located on the underside of the device 200. The data acquisition electronics 256 includes therein a driver for the visible laser 216. A power supply 258 for the device 200 is located on the underside of the base plate 202. [00126] Fig. 23 schematically illustrates the preferred embodiment illustrated in Figs. 21 and 22 along with the channel 20 of the microfluidic mounting system 214. The optical gradient 38 is shown disposed inside the channel 20. A cell or particle 36 travels down the channel 20 in the direction of the arrow. Detectors 238 and 244 are coupled to signal capturing/data processing electronics 260. [00127] Fig. 24 schematically illustrates the fluidics 300 used according to one preferred embodiment to produce a substantially constant, low flow rate through a device 302 containing a channel 304 therein. hi Fig. 23 the channel 304 is oriented in the vertical direction. A reservoir 306 is provided that contains the cells or particles in a fluid medium. Tubing 308 is provided between the reservoir and the inlet to the channel 304. Additional tubing 310 is provided at the outlet of the channel 310 and connects to a flow pump 312. The flow pump 312 is used to provide a substantially constant yet low flow rate of fluid through the channel 304. The flow pump 312 is advantageously controllable so that various flow rates can be used. Also preferably included in the fluidics is a bypass 314. The bypass 314 is used to evacuate fluid from the system after passing through the device 302. A controllable valve 316 is shown in Fig. 23 that is used to direct fluid from the flow pump 312 to the bypass 314. [00128] Figs. 25 and 26 illustrate a preferred embodiment of the flow pump 312. In this embodiment, the flow pump 312 includes a motor 314. The motor 314 is preferably a stepper motor with an integral 50x to lOOx gear reduction drive of the output rotation rate. The motor 314 is coupled to rotationally drive a leadscrew 316. The leadscrew 316 preferably is a high pitch leadscrew. The leadscrew 316 is mechanically coupled to a stage 318. Rotation of the leadscrew 316 imparts linear motion to the stage 318 in the direction of arrow A shown in Fig. 36. As best seen in Fig. 36, the stage 318 is coupled to a syringe plunger 320. A stationary syringe 322 is mounted atop a housing 324. Preferably the syringe 322 is a microsyringe with a volume of between about 1 μL to about 100 μL. Preferably the plunger 320 has a travel distance (stroke) of between about 2 to about 5 cm. It is generally preferably to use a syringe 322 with a small cross-sectional area and a long stroke. These two conditions advantageously produce low flow rates. Tubing 326 is provided at the end of the syringe 322 that is opposite to the end of the syringe 322 with the plunger 320. The tubing 326 is, in turn, connected to a device such as device 302 shown in Fig. 34.
[00129] During operation, the motor 314 rotates the leadscrew 316 which causes the stage 318 and its connected plunger 320 to move in the axial direction (Arrow A in Fig. 36). In one preferred embodiment, the syringe 322 is used to withdraw fluid containing cells or particles from a reservoir such as reservoir 306 shown in Fig. 34. The cells or particles then pass with the fluid through a channel such as channel 304 shown in Fig. 34. After the plunger 320 has traveled its maximum distance, the plunger 320 can be depressed into the syringe 322 using the motor 314 operating in the reverse direction to evacuate the fluid and cells/particles contained therein using, for example, the bypass 314. In an alternative embodiment, the syringe 322 may be preloaded with cells or particles and the fluid can then be pushed through the channel 304 by depressing the plunger 320 using the motor 314.
[00130] Figs. 27 and 28 show a exemplary view of various possible embodiments for massively parallel system. In Fig. 37, an array 350 of multiple channels 352 are disposed parallel to one another. The array 350 may be a two dimensional array, as is shown in solid in Fig. 37, or alternatively, the array 350 may be three-dimensional, as is shown in dashed lines in Fig. 37. The array 350 of channels 352 are connected to a common inlet 354 as well as a common outlet 356. The three detecting positions ti, t2, and t3 are also shown in Fig. 37. An optophoretic gradient 358 is disposed between the second and third detecting positions so as to slow down the particles or cells differentially based on their properties. If the array 350 is two- dimensional, the optophoretic gradient 358 preferably is formed as a line as is shown in Fig. 37. If, however, the array 350 is a three-dimensional array, the optophoretic gradient 358 preferably forms a plane (not shown) that passes through the channels 352. In the three-dimensional array 350, a detector system capable of determining which "layer" of the array the cell or particle is present is needed. One example is the electrode-based detector system illustrated herein in Figs. 8A and 8B.
[00131] Fig. 28 discloses a three-dimensional parallel system that includes a series of stacked two-dimensional arrays 360 separated by a distance D. hi this embodiment a light source 362 such as a Bessel beam, which is capable of reconstructing itself, is used to illuminate the arrays 360. Fig. 28 also shows an optophoretic gradient 364 disposed across the channels contained in each array 360.
[00132] Figs. 29A, 29B, and 29C illustrate an embodiment of a device 400 is capable of both characterizing and sorting cells or particles 402. In this embodiment a channel 404 is provided that contains a region having a plurality of detecting positions. Fig. 29 A shows three such detecting positions ti, t2, and t3. An Optophoretic gradient 406 is disposed between detecting positions t2 and t3. These detecting positions have one or more associated detectors (not shown) that are used to calculate the time-of-flight for region Tl and region T2. A comparison of the time-of-flight for each region can then be made to characterize the cell or particle 402. The device 400 also is capable of sorting cells or particles 402. The sorting step is carried out at a fork region 408 in which the channel 404 branches into two or more branches 410. Fig. 29A shows two such branches 410A and 410B, however, it should be understood that the single channel 404 may branch into any number of branches 410. [00133] Fig. 29B illustrates one method used to sort the cells or particles 402. In this method a moving optical gradient 412 is used to sweep certain cells or particles 402 having certain desired characteristics into one of the braches 410. The optical gradient 412 starts at the one edge of the channel 404 (the lower edge shown in Fig. 29B) and scans generally perpendicular to the direction of fluid flow as shown by the arrows in Fig. 29B. When the cell or particle 402 encounters the moving optical gradient 412, the cell or particle 402 either passes into branch 410A or branch 410B. Those cells or particles 402 that interact more with the moving optical gradient 412 tend to be swept into the upper branch 410B (cell or particle 402B shown in Fig. 29B) while the other cells or particles 402 tend to pass into branch 410A (cell or particle 402A shown in Fig. 29B).
[00134] Fig. 29C illustrates an alternative method used to sort the cells or particles 402. In contrast to the prior embodiment, this embodiment employs a stationary optical gradient 414. The stationary optical gradient 414 is oriented at an angle to the direction of fluid flow. Those cells or particles 402 that interact more with the stationary optical gradient 414 tend to travel along the angled optical gradient and pass into the upper branch 410B (cell or particle 402B shown in Fig. 29C) while the other cells or particles 402 tend to pass into branch 410A (cell or particle 402A shown in Fig. 29C). [00135] EXPERIMENTAL DATA [00136] A. Infection of Red Blood Cells with Plasmodium falciparum (malaria) [00137] In one experiment, line scan as well as time-of-flight analysis was performed on cells to experimentally diagnose infection of red blood cells (RBCs) caused by Plasmodium falciparum, the parasite that causes malaria.
[00138] A variety of methods have been traditionally used to diagnose malaria infection. One method uses a visible stain such as Giemsa stain and subsequent microscopic evaluation. Alternatively, infection may be detected using nucleic-acid binding stains such as, for example, the fluorescent stain acridine orange followed by fluorescence microscopy or flow cytometric analysis. Still other diagnostic techniques use immunological methods that can detect the presence of Plasmodium-specific antigens. All of these methods, however, have limitations. All of the methods require the addition of exogenous reagents. Some of the methods kill the cells, thereby destroying sample integrity. Moreover, these methods often require expensive equipment that needs to be manned by skilled operators.
[00139] As an alternative to these diagnostic methods, Optophoretic interrogation of RBCs has been experimentally used to diagnose infection by Plasmodium falciparum. Plasmodium falciparum-mfected RBC cell cultures and non-infected cell cultures were maintained in RPMI medium supplemented with HEPES buffer, NaHCO3 and gentamicin. Prior to Optophoretic interrogation, cells were washed and diluted in phosphate-buffered saline containing 1% w/v bovine serum albumin (PBS/BSA). [00140] Cells were subject to Optophoretic interrogation either in an unstained or stained condition. The stained cells were stained with the fluorescent nucleic-acid binding dye SybrGreen as a confirmatory test to distinguish infected RBCs (fluorescent) from non-infected RBCs (non-fluorescent). Some experiments were conducted using non-synchronized infection state cultures while other tests were conducted using synchronized cultures. Non-synchronized cultures comprised parasites that are at all stages of their life cycle. In contrast, synchronized cultures contained parasites that were at approximately the same stage of their infection cycle. Still other experiments were performed with varying levels of cellular infection. Because the culture used to grow the malaria parasite was self-limiting due to the presence of breakdown products of RBCs, no more than about 15% of the cells were infected. Higher infection rates with enriched populations of cells were achieved through the Percoll centrifugation-based method. Optophoretic interrogation was performed using line scan analysis as well as time-of- flight (TOF) analysis. [00141] The EV data was generated using non-enriched, non-synchronized samples. The TOF data was generated using enriched, synchronized samples. Escape velocity measurements were taken using optical system similar to that shown in Fig. 20. Fig. 20 shows an optical system having illumination of a sample plate 194 from the top side and imaging from the bottom side. A laser 180 generates a first beam 181 which optionally passes through a spatial filter 182. The spatial filter 182as shown includes lens 183 and aperture 184. The output of the spatial filter 182 is directed to a mirror 185 and passes through the objective 186 and is imaged onto the sample plate 187. The sample plate 187 and material supported on it may be imaged via an objective 188. An optional mirror 189 directs radiation to an optional filter 190 through an imaging lens 191 onto the detector 192. The detector 192 is coupled to an imaging system 193. Preferably, the imaging system 193 provides information to a control system 194 which controls various optical components of the system.
[00142] In this particular experimental setup, the laser was run at 100 mW power. Fig. 30 shows a histogram of the measured escape velocities of both infected and non-infected RBCs. In the tested sample, about 5% of the cells were infected with the parasite. The measured escape velocities for the non-infected RBCs were significantly lower than the escape velocities of the infected RBCs. Fig. 31 shows a comparison of the mean escape velocity for the infected and non-infected RBCs.
[00143] Time-of-flight measurements were also performed on a population of normal RBCs and a non-synchronized, mixed population of infected RBCs. With respect to the infected population, the population contained in excess of 70% infected cells. Time-of-flight measurements were made using a 2.6 W laser focused with a cylindrical lens. Fig. 32 shows a histogram of the time-of-flight measurements for normal RBCs and infected RBCs. As seen in Fig. 32, the infected RBCs show a slight shift upward in time-of-flight (TOF) values as compared to the non-infected (control) RBCs. Fig. 33 shows the comparison of the mean TOF values for the infected and control cells.
[00144] Figs. 34 and 35 illustrate the results of another time-of-flight experiment performed on infected and non-infected RBCs. The same experimental setup was used as in the prior experiment (i.e., 2.6W laser with cylindrical lens). In this experiment, however, synchronized cells were tested, h addition, in this experiment, over 95% of the cells were infected. Fig. 34 shows a histogram of TOF data for the infected and non-infected cells. As seen in Fig. 34, the infected cells show a noticeable increase in TOF values as compared to their non-infected counterparts. Fig. 35 shows the mean TOF values for both the infected and non-infected populations. [00145] B. Characterization of Normal and Cancerous Cells
[00146] Optophoretic interrogation using time-of-flight analysis has been used to distinguish cancer cells from normal cells for breast carcinoma and skin melanoma. In this regard, Optophoretic interrogation can be used as a diagnostic tool to determine whether cells show the Optophoretic characteristics of cancer cells or normal cells. In addition, the Optophoretic methods can also be used to detect whether the sample cells are primary or metastatic cells. These provide a relatively quick way of diagnosing whether a sample contains cancerous cells. The techniques advantageously may be used with relatively small sample sizes. [00147] Experiments have been conducted on human breast carcinoma cells as well as human melanoma cells. Tumor cell lines were purchased from ATCC and, when available, their normal counterparts were matched from the same patient. Cells were grown in culture until the time of testing. Adherent cells were detached from culture flasks using trypsin and resuspended in buffer. Cells were then subject to Optophoretic interrogation.
[00148] Samples of matched cancerous and non-cancerous cells from breast tissue (HS578T and HS578BST) were tested using a time-of-flight system. Fig. 36 illustrates a histogram of the ratio of T2/Tι plotted against the percentage of cells. The cancerous cells (HS578T) exhibited a larger T2/T] ratio as compared to the normal cells (HS578BST). Fig. 37 illustrates the mean T2/Tι ratio for the cancerous and non-cancerous cells. [00149] In yet another experiment, normal skin cells (CCD 1037) and malignant melanoma cells (WM 115) were subject to time-of-flight analysis. The time-of-flight analysis was performed using a laser powered at 2.6W using a cylindrical lens . Fig. 38 illustrates the ratio of T2/Tι plotted against the percentage of cells. The cancerous cells (WM 115) exhibited a larger T2/Tι ratio as compared to the normal cells (CCD 1037). These results are consistent with the results seen in the time-of-flight data for breast carcinoma cells, namely, that the cancerous cells exhibit generally higher T2/Tι ratios. Fig. 39 illustrates the mean T2/Tι ratio for the cancerous and non-cancerous cells. [00150] C. Characterization of Cells Using Beads [00151] In this experiment, 5.1 μm polystyrene and 5.0 μm polymethylmethacrylate (PMMA) beads were subject to time-of-flight analysis on a diagnostic device of the type shown in Figs. 32-34. These two samples of particles have different refractive indexes. The small difference in diameters was small enough that characterization was based on refractive index difference and not particle diameter. Fig. 40 illustrates a scatter plot of the time-of-flight (TOF) ratio as a function of ti. Fig. 41 illustrates a histogram of the number of particles as a function of the TOF ratio \_l\\. As seen in Figs. 40 and 41, the polystyrene beads have a larger TOF ratio as compared to the PMMA beads. The beads may be used as vehicles to carry different cells or populations of cells. Interrogation may then be performed on the underlying vehicle which, in turn, allows for the characterization and possible sorting of cells adhered to the surface of the beads. For example, beads made from polystyrene may have one or more compounds bound thereto that are specific to a particular cell or cell type. Other beads made from another material, i.e., PMMA, may include one or more different compounds thereon that bind to a different type of cell. Characterization and sorting may be performed using the Optophoretic differences in the carrier beads. In another related application, the beads may be used in an agglomeration assay in which beads have specific ligands attached to their surfaces. The ligand laden beads are then able to bind to cells having corresponding binding sites, hi this type of assay, a single cell might be bound to multiple beads, each bead having a different ligand. These bead-ligand-cell complexes may then be analyzed optophoretically to analyze and differentiate the cells of interest. [00152] D. Characterization of Wild Type and Mutant Yeast Strains
[00153] In this experiment, two strains of yeast, 24657 rho+ (wild type) and MYA-1133 rho(0) (mutant) were subject to time-of-flight testing on a diagnostic device of the type shown in Figs. 32-34. The difference between the wild type and the mutant yeast strain is that the rho(0) strain lacks mitochondrial DNA. Fig. 42 illustrates a scatter plot of the time-of-flight (TOF) ratio as a function of the event number for the mutant and wild type cells. Fig. 43 illustrates a histogram of the percentage of yeast cells as a function of the TOF ratio t2/tι. As seen in Figs. 42 and 43, the mutant strain rho(0) generally has a lower TOF ratio as compared to the wild type strain.
[00154] E. Characterization of HL60 Cells hi Response to Treatment with DMSO [00155] In this experiment, HL60 cells were treated with 1% dimethyl sulfoxide (DMSO) and subject to time-of-flight testing on a diagnostic device of the type shown in Figs. 32-34. As a control, a set of HL60 cells that were not treated with DMSO were also subject to time-of-flight testing. Measurements were made after 40 hours of treatment with DMSO. DMSO is a known cell differentiation inducer. Fig. 44 illustrates a scatter plot of the time-of-flight (TOF) ratio as a function of the event number for the treated and non-treated HL60 cells. Fig. 45 illustrates a histogram of the percentage of cells as a function of the TOF ratio t2/tι. As seen in Figs. 44 and 45, the HL60 cells treated with DMSO generally had a lower TOF ratio as compared to the non- treated HL60 cells.
[00156] F. Characterization of Activated and Unactivated T Cells
[00157] In this experiment, activated and unactivated T cells were subject to time-of-flight testing on a diagnostic device of the type shown in Figs. 32-34. Phorbol mystirate acetate (PMA) and ionomycin were used to activate the T cells. T cells were cultured at a cell density on the order of 10"6 cells per ml in the presence of 0.5 μg/ml PMA and 50 ng/ml ionomycin overnight in a CO2 incubator. Fig. 46 illustrates a histogram of the percentage of cells as a function of the TOF ratio t2/tι. As seen in Fig. 46, the stimulated T cells generally had a lower TOF ratio as compared to the unstimulated T cells.
[00158] While the invention is susceptible to various modifications, and alternative forms, specific examples thereof have been shown in the drawings and are herein described in detail. It should be understood, however, that the mvention is not to be limited to the particular forms or methods disclosed, but to the contrary, the invention is to cover all modifications, equivalents and alternatives falling within the spirit and scope of the appended claims.

Claims

We Claim:
1. A device for characterizing cells or particles comprising: a channel having an inlet and an outlet, a source of fluid for flowing through the channel from the inlet to the outlet, the source of fluid carrying at least one cell or particle, detectors for detecting the position of the at least one cell or particle within the channel at at least three points in time, a light source for defining an optical gradient across at least a portion of the channel in a direction generally orthogonal to the fluid flow, and an analysis system coupled to the detectors to characterize the at least one cell or particle.
2. The device according to claim 1, wherein the channel is defined in a substrate.
3. The device according to claim 1, wherein the device further includes an inlet reservoir.
4. The device according to claim 1, wherein the device further includes an outlet reservoir.
5. The device according to claim 1, wherein the detectors are discrete detectors.
6. The device according to claim 1, wherein the detectors are integrated detectors.
7. The device according to claim 1, further comprising an illumination system having a light source for illuminating a portion of the channel.
8. The device according to claim 7, wherein the illumination system comprises a pattern generator.
9. The device according to claim 7, wherem the illumination system comprises a scanning device.
10. The device according to claim 7, further comprising a detector mask.
11. The device according to claim 1, wherein the analysis system includes a display.
12. The device according to claim 1, further comprising a sorting system.
13. The device according to claim 1, wherein the analysis system controls the sorting system.
14. A device for characterizing a cell or particle comprising: a channel having an inlet and an outlet, the channel containing a moving fluid therein for carrying the cell or particle from the inlet to the outlet; a detector for detecting the presence of a cell or particle along a portion of the channel, the detector including a first detecting position, a second detecting position located downstream of the first detecting position, and a third detecting position located downstream of the second detecting position; a light source providing an optical gradient disposed within the channel and between the second and third detection positions of the detector; a control system coupled to the detector to receive and process detected signals from the detector.
15. The device according to claim 14, wherein the detector comprises a plurality of discrete detectors.
16. The device according to claim 14, wherein the detector comprises an integrated detector.
17. The device according to claim 14, wherein the light source comprises a laser for generating the optical gradient.
18. The device according to claim 14, further comprising an illumination system having a light source for illuminating a portion of the channel.
19. The device according to claim 18, wherein the illumination system comprises a pattern generator.
20. The device according to claim 18, wherein the illumination system comprises a scanning device.
21. The device according to claim 18, further comprising a detector mask.
22. The device according to claim 14, wherein the control system includes a display.
23. The device according to claim 14, wherein the distance between the first and third detecting positions is less than 200 microns.
24. The device according to claim 14, wherein the distance between the first and second detecting positions is equal to the distance between the second and third detecting positions.
25. The device according to claim 14, wherein the moving fluid has a substantially constant flow rate.
26. The device according to claim 22, wherein a timing diagram is displayable on the display.
27. The device according to claim 14, wherein the flow rate of the moving fluid is adjustable.
28. The device according to claim 14, wherein the flow rate of the moving fluid exceeds the escape velocity of the cell or particle.
29. A method for characterizing a cell or particle comprising the steps of: flowing a cell or particle past first and second points defining a first zone; measuring the time it takes the cell or particle to pass between the first and second points in the first zone; flowing a cell or particle past first and second points defining a second zone; subjecting the cell or particle to an optical gradient positioned in the second zone; measuring the time it takes the cell or particle to pass between the first and second points in the second zone; and comparing the measured times for the first and second zones for characterizing the cell or particle.
30. The method according to claim 29, further comprising the step of sorting the cell or particle based on the measured times for the first and second zones.
31. The method according to claim 29, wherein the second point in the first zone is also the first point in the second zone.
32. A method of determining a biological property of a cell or population of cells comprising the steps of: flowing a cell past first and second points defining a first zone; measuring the time it takes the cell to pass between the first and second points in the first zone; flowing the cell past first and second points defining a second zone; subjecting the cell to an optical gradient positioned in the second zone; measuring the time it takes the cell to pass between the first and second points in the second zone; and comparing the measured times for the first and second zones for the cell so as to determine a biological property of the cell based at least in part on the comparison.
33. The method according to claim 32, wherein the biological property comprises whether the cell is infected with an infectious agent.
34. The method according to claim 32, wherein the biological property comprises whether the cell is cancerous.
35. The method according to claim 34, wherein the biological property comprises the metastatic potential of the cell.
36. The method according to claim 32, wherein the biological property comprises detecting a phenotype change in the cell.
37. The method according to claim 32, wherein the biological property comprises detecting whether the cell is wild type or mutant.
38. The method according to claim 32, wherein the cell is a T cell.
39. The method according to claim 38, wherein the biological property comprises the activation level of the T cell.
40. A method of diagnosing a diseased state of one or more cells in a sample containing a plurality of cells comprising the steps of: flowing the sample of cells through a first detecting region; measuring the time it takes the cells to pass through the first detecting region; flowing the cells through a second detecting region located downstream of the first detecting region; subjecting the cells to an optical gradient positioned in the second detecting region; measuring the time it takes the cells pass through the second detecting region; and comparing the measured times for the first and second detecting regions for characterizing at least a portion of the cells in the sample as being in a diseased state or in a normal state.
41. The method according to claim 40, wherein the diseased state is cancer.
42. The method according to claim 40, wherein the diseased state is infection.
43. A method of analysis of an environmental sample containing a plurality of particles comprising the steps of: flowing the sample of particles through a first detecting region; measuring the time it takes the particles to pass through the first detecting region; flowing the particles through a second detecting region located downstream of the first detecting region; subjecting the particles to an optical gradient positioned in the second detecting region; measuring the time it takes the particles pass through the second detecting region; and comparing the measured times for the first and second detecting regions for characterizing at least a portion of the particles in the sample.
44. The method according to claim 43, wherein the sample is an airborne sample.
45. The method according to claim 43, wherein the sample is a waterborne sample.
46. A device for characterizing cells or particles comprising: at least one channel having an inlet and an outlet; a source of fluid for flowing through the at least one channel from the inlet to the outlet, the source of fluid carrying cells or particles; a first light source for defining a detection beam within the at least one channel, the
detection beam being disposed in the channel and generally parallel to the direction of fluid flow; a first detector for detecting the presence of a cell or particle along a portion of the at least one channel; a second detector for detecting the presence of a cell or particle along another portion of the at least one channel, the second detector including a first detecting position, a second detecting position located downstream of the first detection position, and a third detecting position located downstream of the second detecting position; a second light source for providing an optical gradient disposed within the at least one channel and between the second and third detection positions of the second detector; and a control system coupled to the first and second detectors to receive and process detected signals from the first and second detectors.
47. The device according to claim 46, the first light source comprising a coherent light source.
48. The device according to claim 46, the second light source comprising a coherent light source.
49. The device according to claim 46, further comprising a flow pump coupled to one of the inlet and outlet of the channel.
50. The device according to claim 46, wherein the channel is disposed inside a microfluidic mounting system.
51. The device according to claim 46, further comprising an external computer coupled to the device via a computer interface.
52. The device according to claim 46, further comprising a plurality of channels.
PCT/US2003/030975 2002-10-08 2003-09-30 Methods and apparatus for optophoretic diagnosis of cells and particles WO2004033059A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003272804A AU2003272804A1 (en) 2002-10-08 2003-09-30 Methods and apparatus for optophoretic diagnosis of cells and particles

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/267,914 US20040067167A1 (en) 2002-10-08 2002-10-08 Methods and apparatus for optophoretic diagnosis of cells and particles
US10/267,914 2002-10-08

Publications (2)

Publication Number Publication Date
WO2004033059A2 true WO2004033059A2 (en) 2004-04-22
WO2004033059A3 WO2004033059A3 (en) 2006-01-26

Family

ID=32042845

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/030975 WO2004033059A2 (en) 2002-10-08 2003-09-30 Methods and apparatus for optophoretic diagnosis of cells and particles

Country Status (3)

Country Link
US (1) US20040067167A1 (en)
AU (1) AU2003272804A1 (en)
WO (1) WO2004033059A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1950552A2 (en) * 2007-01-26 2008-07-30 Palo Alto Research Center Incorporated Method andsystem implementing spatially modulated excitation or emission for particle characterization with enhanced sensitivity
CN101910821B (en) * 2007-12-04 2012-09-05 粒子监测系统有限公司 Non-orthogonal particle detection systems and methods
US9029800B2 (en) 2011-08-09 2015-05-12 Palo Alto Research Center Incorporated Compact analyzer with spatial modulation and multiple intensity modulated excitation sources
US9164037B2 (en) 2007-01-26 2015-10-20 Palo Alto Research Center Incorporated Method and system for evaluation of signals received from spatially modulated excitation and emission to accurately determine particle positions and distances

Families Citing this family (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040121474A1 (en) * 2002-12-19 2004-06-24 Genoptix, Inc Detection and evaluation of chemically-mediated and ligand-mediated t-cell activation using optophoretic analysis
US20040121307A1 (en) * 2002-12-19 2004-06-24 Genoptix, Inc Early detection of cellular differentiation using optophoresis
ES2544944T3 (en) * 2003-05-08 2015-09-07 The University Court Of The University Of St. Andrews Particle fractionation
FR2860886B1 (en) 2003-10-14 2005-12-23 Commissariat Energie Atomique DEVICE FOR DISPLACING PARTICLES
EP1765228A4 (en) * 2004-06-28 2009-06-10 Haemonetics Corp Blood component separation system with stationary separation chamber
US7522786B2 (en) * 2005-12-22 2009-04-21 Palo Alto Research Center Incorporated Transmitting light with photon energy information
US7630076B2 (en) * 2005-07-26 2009-12-08 University Of Connecticut Dual-detector systems and methods having utility in biomolecular measurements
US8437582B2 (en) * 2005-12-22 2013-05-07 Palo Alto Research Center Incorporated Transmitting light with lateral variation
US7420677B2 (en) * 2005-12-22 2008-09-02 Palo Alto Research Center Incorporated Sensing photon energies of optical signals
US7386199B2 (en) 2005-12-22 2008-06-10 Palo Alto Research Center Incorporated Providing light to channels or portions
US7433552B2 (en) 2005-12-22 2008-10-07 Palo Alto Research Center Incorporated Obtaining analyte information
US7547904B2 (en) 2005-12-22 2009-06-16 Palo Alto Research Center Incorporated Sensing photon energies emanating from channels or moving objects
US7358476B2 (en) * 2005-12-22 2008-04-15 Palo Alto Research Center Incorporated Sensing photons from objects in channels
US9878326B2 (en) 2007-09-26 2018-01-30 Colorado School Of Mines Fiber-focused diode-bar optical trapping for microfluidic manipulation
US9885644B2 (en) * 2006-01-10 2018-02-06 Colorado School Of Mines Dynamic viscoelasticity as a rapid single-cell biomarker
JP2007301534A (en) * 2006-05-15 2007-11-22 Ebara Corp Atomizer
GB0614297D0 (en) * 2006-07-19 2006-08-30 Shaw Water Engineering Ltd Apparatus, system and method for detecting particles
GB0618605D0 (en) * 2006-09-21 2006-11-01 Univ St Andrews Optical sorting
GB0618606D0 (en) * 2006-09-21 2006-11-01 Univ St Andrews Optical sorting
EP1905755A1 (en) * 2006-09-26 2008-04-02 Lonza Ag Process for the synthesis of 2,2 ,6-tribromobiphenyl
US20080094608A1 (en) * 2006-10-23 2008-04-24 The Regents Of The University Of California Laser velocimetry system
US7936463B2 (en) 2007-02-05 2011-05-03 Palo Alto Research Center Incorporated Containing analyte in optical cavity structures
US7817276B2 (en) 2007-02-05 2010-10-19 Palo Alto Research Center Incorporated Distinguishing objects
US7554673B2 (en) * 2007-02-05 2009-06-30 Palo Alto Research Center Incorporated Obtaining information about analytes using optical cavity output light
US7633629B2 (en) * 2007-02-05 2009-12-15 Palo Alto Research Center Incorporated Tuning optical cavities
US7545513B2 (en) * 2007-02-05 2009-06-09 Palo Alto Research Center Incorporated Encoding optical cavity output light
US7502123B2 (en) * 2007-02-05 2009-03-10 Palo Alto Research Center Incorporated Obtaining information from optical cavity output light
US8308926B2 (en) 2007-08-20 2012-11-13 Purdue Research Foundation Microfluidic pumping based on dielectrophoresis
US10722250B2 (en) 2007-09-04 2020-07-28 Colorado School Of Mines Magnetic-field driven colloidal microbots, methods for forming and using the same
JP5260919B2 (en) * 2007-09-05 2013-08-14 浜松ホトニクス株式会社 Blood test equipment
US8320983B2 (en) * 2007-12-17 2012-11-27 Palo Alto Research Center Incorporated Controlling transfer of objects affecting optical characteristics
EP2085760B1 (en) * 2008-01-30 2018-07-04 Palo Alto Research Center Incorporated Producing time variation in emanating light
US7763856B2 (en) * 2008-01-31 2010-07-27 Palo Alto Research Center Incorporated Producing time variation in emanating light
US8263955B2 (en) * 2008-12-18 2012-09-11 Palo Alto Research Center Incorporated Causing relative motion
US7701580B2 (en) * 2008-02-01 2010-04-20 Palo Alto Research Center Incorporated Transmitting/reflecting emanating light with time variation
US8153949B2 (en) * 2008-12-18 2012-04-10 Palo Alto Research Center Incorporated Obtaining sensing results indicating time variation
US8153950B2 (en) * 2008-12-18 2012-04-10 Palo Alto Research Center Incorporated Obtaining sensing results and/or data in response to object detection
US7817254B2 (en) * 2008-01-30 2010-10-19 Palo Alto Research Center Incorporated Obtaining information from time variation of sensing results
US7894068B2 (en) * 2008-02-04 2011-02-22 Palo Alto Research Center Incorporated Producing filters with combined transmission and/or reflection functions
US8629981B2 (en) 2008-02-01 2014-01-14 Palo Alto Research Center Incorporated Analyzers with time variation based on color-coded spatial modulation
US8373860B2 (en) * 2008-02-01 2013-02-12 Palo Alto Research Center Incorporated Transmitting/reflecting emanating light with time variation
EP2383551B1 (en) * 2009-01-06 2020-04-08 The Furukawa Electric Co., Ltd. Optical measurement apparatus and sample identifying and dispensing apparatus
WO2010088761A1 (en) * 2009-02-06 2010-08-12 Maziyar Khorasani Method and apparatus for manipulating and detecting analytes
US8343440B2 (en) * 2009-03-27 2013-01-01 Seiko Epson Corporation Cell separating apparatus and cell separating method
WO2011071912A1 (en) * 2009-12-07 2011-06-16 Yale University Label-free cellular manipulation and sorting via biocompatible ferrofluids
JP5617530B2 (en) * 2010-10-29 2014-11-05 ソニー株式会社 Cell sorting device and cell sorting method
US20120181450A1 (en) * 2011-01-18 2012-07-19 Electronics And Telecommunications Research Institute Method and apparatus for detecting bio material using photoelectric conversion device, and method for manufacturing photoelectric conversion device
US8723140B2 (en) 2011-08-09 2014-05-13 Palo Alto Research Center Incorporated Particle analyzer with spatial modulation and long lifetime bioprobes
US20160296945A1 (en) 2013-03-15 2016-10-13 Ancera, Inc. Systems and methods for active particle separation
WO2014145765A1 (en) 2013-03-15 2014-09-18 Ancera, Inc. Systems and methods for bead-based assays in ferrofluids
US9372143B2 (en) * 2013-05-15 2016-06-21 Captl Llc Scanning image flow cytometer
US9315846B2 (en) * 2014-02-13 2016-04-19 The United States Of America As Represented By Secretary Of The Navy Fluidic channel based on a filtered, free-space electromagnetic wave source
US9528925B2 (en) * 2014-02-14 2016-12-27 Palo Alto Research Center Incorporated Spatial modulation of light to determine object position
WO2016100954A1 (en) 2014-12-19 2016-06-23 Captl Llc Flow cytometry using hydrodynamically planar flow
US10036698B2 (en) 2015-06-19 2018-07-31 Captl Llc Time-sequential cytometry
WO2016210348A2 (en) 2015-06-26 2016-12-29 Ancera, Inc. Background defocusing and clearing in ferrofluid-based capture assays
TWI569016B (en) * 2015-10-19 2017-02-01 國立高雄應用科技大學 Light-trapping cancer cell stage testing method
US10228317B1 (en) * 2016-08-25 2019-03-12 Verily Life Sciences Llc Multiplexed microfluidic cell sorting using laser induced cavitation bubbles
US10605733B1 (en) 2016-12-15 2020-03-31 Verily Life Sciences Llc Light sheet imaging flow cytometer
CN108704677A (en) * 2018-04-27 2018-10-26 广州万孚生物技术股份有限公司 A kind of micro-fluidic chip and the analytical instrument containing it
CN108716938A (en) * 2018-04-27 2018-10-30 广州万孚生物技术股份有限公司 A kind of liquid quantifying device and its application

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020108859A1 (en) * 2000-11-13 2002-08-15 Genoptix Methods for modifying interaction between dielectric particles and surfaces

Family Cites Families (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3638139A (en) * 1964-09-29 1972-01-25 Bell Telephone Labor Inc Frequency-selective laser devices
US3558877A (en) * 1966-12-19 1971-01-26 Gca Corp Method and apparatus for mass separation by selective light absorption
US3710279A (en) * 1969-12-15 1973-01-09 Bell Telephone Labor Inc Apparatuses for trapping and accelerating neutral particles
CA944466A (en) * 1970-01-26 1974-03-26 Western Electric Company, Incorporated Guided raman devices
US3808432A (en) * 1970-06-04 1974-04-30 Bell Telephone Labor Inc Neutral particle accelerator utilizing radiation pressure
US3662183A (en) * 1970-12-28 1972-05-09 Bell Telephone Labor Inc Continuously tunable optical parametric oscillator
US3725810A (en) * 1971-04-23 1973-04-03 Bell Telephone Labor Inc Optical stimulated emission devices employing split optical guides
US3761721A (en) * 1972-07-06 1973-09-25 Trw Inc Matter wave interferometric apparatus
US4092535A (en) * 1977-04-22 1978-05-30 Bell Telephone Laboratories, Incorporated Damping of optically levitated particles by feedback and beam shaping
US4247815A (en) * 1979-05-22 1981-01-27 The United States Of America As Represented By The Secretary Of The Army Method and apparatus for physiologic facsimile imaging of biologic targets based on complex permittivity measurements using remote microwave interrogation
US4253846A (en) * 1979-11-21 1981-03-03 Technicon Instruments Corporation Method and apparatus for automated analysis of fluid samples
US4327288A (en) * 1980-09-29 1982-04-27 Bell Telephone Laboratories, Incorporated Method for focusing neutral atoms, molecules and ions
US4386274A (en) * 1980-11-10 1983-05-31 Saul Altshuler Isotope separation by standing waves
US4453805A (en) * 1981-02-19 1984-06-12 Bell Telephone Laboratories, Incorporated Optical grating using a liquid suspension of dielectric particles
FR2506530A1 (en) * 1981-05-22 1982-11-26 Thomson Csf COHERENT RADIATION SOURCE GENERATING AN ADJUSTABLE SPREAD DIRECTION BEAM
US4390403A (en) * 1981-07-24 1983-06-28 Batchelder J Samuel Method and apparatus for dielectrophoretic manipulation of chemical species
FR2519777A1 (en) * 1982-01-12 1983-07-18 Thomson Csf METHOD FOR MANUFACTURING DIFFRACTANT PHASE STRUCTURES
US4440638A (en) * 1982-02-16 1984-04-03 U.T. Board Of Regents Surface field-effect device for manipulation of charged species
FR2537768A1 (en) * 1982-12-08 1984-06-15 Commissariat Energie Atomique METHOD AND DEVICE FOR OBTAINING SPATIALLY MODULATED DENSITY PARTICLE BEAMS, APPLICATION TO ION ETCHING AND IMPLANTATION
DE3574617D1 (en) * 1984-09-11 1990-01-11 Partec Ag METHOD AND DEVICE FOR SORTING MICROSCOPIC PARTICLES.
US4827125A (en) * 1987-04-29 1989-05-02 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Confocal scanning laser microscope having no moving parts
US4893886A (en) * 1987-09-17 1990-01-16 American Telephone And Telegraph Company Non-destructive optical trap for biological particles and method of doing same
US4908112A (en) * 1988-06-16 1990-03-13 E. I. Du Pont De Nemours & Co. Silicon semiconductor wafer for analyzing micronic biological samples
US5100627A (en) * 1989-11-30 1992-03-31 The Regents Of The University Of California Chamber for the optical manipulation of microscopic particles
FR2655435B1 (en) * 1989-12-01 1992-02-21 Thomson Csf COHERENT ADDITION DEVICE OF LASER BEAMS.
CA2031716C (en) * 1989-12-07 1996-06-18 Hiroaki Misawa Laser microprocessing and the device therefor
US5795457A (en) * 1990-01-30 1998-08-18 British Technology Group Ltd. Manipulation of solid, semi-solid or liquid materials
US5029791A (en) * 1990-03-08 1991-07-09 Candela Laser Corporation Optics X-Y positioner
US5198369A (en) * 1990-04-25 1993-03-30 Canon Kabushiki Kaisha Sample measuring method using agglomeration reaction of microcarriers
US5079169A (en) * 1990-05-22 1992-01-07 The Regents Of The Stanford Leland Junior University Method for optically manipulating polymer filaments
US5338930A (en) * 1990-06-01 1994-08-16 Research Corporation Technologies Frequency standard using an atomic fountain of optically trapped atoms
US5245466A (en) * 1990-08-15 1993-09-14 President And Fellows Of Harvard University And Rowland Institute Optical matter
US5113286A (en) * 1990-09-27 1992-05-12 At&T Bell Laboratories Diffraction grating apparatus and method of forming a surface relief pattern in diffraction grating apparatus
CA2057506C (en) * 1990-12-13 2003-05-13 Keiji Sasaki Laser trapping and method for applications thereof
US5919645A (en) * 1991-04-09 1999-07-06 Azur Environmental Method for the direct determination of the toxicity of particulate solids
JP3129471B2 (en) * 1991-06-01 2001-01-29 科学技術振興事業団 Multi-beam particle operation method
US5206504A (en) * 1991-11-01 1993-04-27 The United States Of America As Represented By The Administrator, National Aeronautics And Space Administration Sample positioning in microgravity
JP3018687B2 (en) * 1991-12-12 2000-03-13 松下電器産業株式会社 Scanning laser microscope
US5495105A (en) * 1992-02-20 1996-02-27 Canon Kabushiki Kaisha Method and apparatus for particle manipulation, and measuring apparatus utilizing the same
JPH0693038B2 (en) * 1992-06-11 1994-11-16 東京工業大学長 Method and apparatus for controlling the motion of a small number of neutral atoms
US5189294A (en) * 1992-07-08 1993-02-23 The United States Of America As Represented By The Secretary Of The Air Force Transform lens with a plurality of sliced lens segments
US6399397B1 (en) * 1992-09-14 2002-06-04 Sri International Up-converting reporters for biological and other assays using laser excitation techniques
LU88184A1 (en) * 1992-10-28 1994-09-09 Europ Communities Optical modulator
US5452123A (en) * 1992-12-30 1995-09-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Method of making an optically nonlinear switched optical device and related devices
US5327515A (en) * 1993-01-14 1994-07-05 At&T Laboratories Method for forming a Bragg grating in an optical medium
GB9301122D0 (en) * 1993-01-21 1993-03-10 Scient Generics Ltd Method of analysis/separation
GB9306729D0 (en) * 1993-03-31 1993-05-26 British Tech Group Improvements in separators
DE69413470T2 (en) * 1993-07-08 1999-03-04 Canon Kk Particle separation method and apparatus
JP3390173B2 (en) * 1993-07-22 2003-03-24 ビーティージー・インターナショナル・リミテッド Intelligent sensors for near-field optical devices
EP0714348A4 (en) * 1993-07-27 1998-05-06 Physical Optics Corp Light source destructuring and shaping device
US5445011A (en) * 1993-09-21 1995-08-29 Ghislain; Lucien P. Scanning force microscope using an optical trap
US5900160A (en) * 1993-10-04 1999-05-04 President And Fellows Of Harvard College Methods of etching articles via microcontact printing
US5512745A (en) * 1994-03-09 1996-04-30 Board Of Trustees Of The Leland Stanford Jr. University Optical trap system and method
JP3355021B2 (en) * 1994-03-26 2002-12-09 科学技術振興事業団 Micro memory and micro sensor
DE4411268C2 (en) * 1994-03-31 2001-02-01 Danfoss As Analysis method and device
US6071394A (en) * 1996-09-06 2000-06-06 Nanogen, Inc. Channel-less separation of bioparticles on a bioelectronic chip by dielectrophoresis
US5637458A (en) * 1994-07-20 1997-06-10 Sios, Inc. Apparatus and method for the detection and assay of organic molecules
US6001229A (en) * 1994-08-01 1999-12-14 Lockheed Martin Energy Systems, Inc. Apparatus and method for performing microfluidic manipulations for chemical analysis
JP3474652B2 (en) * 1994-11-11 2003-12-08 株式会社モリテックス Multi-point laser trapping apparatus and method
US5629802A (en) * 1995-01-05 1997-05-13 The United States Of America As Represented By The Secretary Of The Air Force Spatially multiplexed optical signal processor
US5795782A (en) * 1995-03-17 1998-08-18 President & Fellows Of Harvard College Characterization of individual polymer molecules based on monomer-interface interactions
US5793485A (en) * 1995-03-20 1998-08-11 Sandia Corporation Resonant-cavity apparatus for cytometry or particle analysis
US5608519A (en) * 1995-03-20 1997-03-04 Gourley; Paul L. Laser apparatus and method for microscopic and spectroscopic analysis and processing of biological cells
US5953166A (en) * 1995-03-22 1999-09-14 Moritex Corporation Laser trapping apparatus
US6797942B2 (en) * 2001-09-13 2004-09-28 University Of Chicago Apparatus and process for the lateral deflection and separation of flowing particles by a static array of optical tweezers
US5631141A (en) * 1995-05-05 1997-05-20 The Regents Of The University Of California High resolution biosensor for in-situ microthermometry
US5776674A (en) * 1995-06-05 1998-07-07 Seq, Ltd Chemical biochemical and biological processing in thin films
US5659561A (en) * 1995-06-06 1997-08-19 University Of Central Florida Spatial solitary waves in bulk quadratic nonlinear materials and their applications
US5620857A (en) * 1995-06-07 1997-04-15 United States Of America, As Represented By The Secretary Of Commerce Optical trap for detection and quantitation of subzeptomolar quantities of analytes
US5950071A (en) * 1995-11-17 1999-09-07 Lightforce Technology, Inc. Detachment and removal of microscopic surface contaminants using a pulsed detach light
US5888370A (en) * 1996-02-23 1999-03-30 Board Of Regents, The University Of Texas System Method and apparatus for fractionation using generalized dielectrophoresis and field flow fractionation
US6078681A (en) * 1996-03-18 2000-06-20 Marine Biological Laboratory Analytical imaging system and process
US5942443A (en) * 1996-06-28 1999-08-24 Caliper Technologies Corporation High throughput screening assay systems in microscale fluidic devices
US5760395A (en) * 1996-04-18 1998-06-02 Universities Research Assoc., Inc. Method and apparatus for laser-controlled proton beam radiology
US5952651A (en) * 1996-06-10 1999-09-14 Moritex Corporation Laser manipulation apparatus and cell plate used therefor
US6280967B1 (en) * 1996-08-02 2001-08-28 Axiom Biotechnologies, Inc. Cell flow apparatus and method for real-time of cellular responses
US5804436A (en) * 1996-08-02 1998-09-08 Axiom Biotechnologies, Inc. Apparatus and method for real-time measurement of cellular response
US6221654B1 (en) * 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
US5858192A (en) * 1996-10-18 1999-01-12 Board Of Regents, The University Of Texas System Method and apparatus for manipulation using spiral electrodes
DE19649048C1 (en) * 1996-11-27 1998-04-09 Evotec Biosystems Gmbh Particle identification method for enzyme-linked immunoassay using fast Fourier transform
US5939716A (en) * 1997-04-02 1999-08-17 Sandia Corporation Three-dimensional light trap for reflective particles
US6215134B1 (en) * 1997-05-09 2001-04-10 California Institute Of Technology Semiconductor surface lenses and shaped structures
GB2326229A (en) * 1997-06-13 1998-12-16 Robert Jeffrey Geddes Carr Detecting and analysing submicron particles
US6111398A (en) * 1997-07-03 2000-08-29 Coulter International Corp. Method and apparatus for sensing and characterizing particles
DE19801139B4 (en) * 1998-01-14 2016-05-12 Till Photonics Gmbh Point Scanning Luminescence Microscope
US6055106A (en) * 1998-02-03 2000-04-25 Arch Development Corporation Apparatus for applying optical gradient forces
US6082205A (en) * 1998-02-06 2000-07-04 Ohio State University System and device for determining particle characteristics
US6740497B2 (en) * 1998-03-06 2004-05-25 The Regents Of The University Of California Method and apparatus for detecting cancerous cells using molecules that change electrophoretic mobility
US6088376A (en) * 1998-03-16 2000-07-11 California Institute Of Technology Vertical-cavity-surface-emitting semiconductor devices with fiber-coupled optical cavity
US6067859A (en) * 1999-03-04 2000-05-30 The Board Of Regents, The University Of Texas System Optical stretcher
JP5087192B2 (en) * 1999-11-30 2012-11-28 インテレクソン コーポレイション Method and apparatus for selectively aiming a specific cell in a cell group
AU2001290879A1 (en) * 2000-09-15 2002-03-26 California Institute Of Technology Microfabricated crossflow devices and methods
US6744038B2 (en) * 2000-11-13 2004-06-01 Genoptix, Inc. Methods of separating particles using an optical gradient
US6778724B2 (en) * 2000-11-28 2004-08-17 The Regents Of The University Of California Optical switching and sorting of biological samples and microparticles transported in a micro-fluidic device, including integrated bio-chip devices
WO2003008943A1 (en) * 2001-07-19 2003-01-30 Tufts University Optical array device and methods of use thereof for screening, analysis and manipulation of particles

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020108859A1 (en) * 2000-11-13 2002-08-15 Genoptix Methods for modifying interaction between dielectric particles and surfaces

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
IMASAKA T ET AL: 'Optical chromatography. A new tool for separation of particles.' ANALYSIS MAGAZINE. vol. 26, no. 5, 1998, pages M53 - M55, XP002992369 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1950552A2 (en) * 2007-01-26 2008-07-30 Palo Alto Research Center Incorporated Method andsystem implementing spatially modulated excitation or emission for particle characterization with enhanced sensitivity
JP2008185585A (en) * 2007-01-26 2008-08-14 Palo Alto Research Center Inc Method and system for evaluating particle characteristic
EP1950552A3 (en) * 2007-01-26 2010-11-17 Palo Alto Research Center Incorporated Method andsystem implementing spatially modulated excitation or emission for particle characterization with enhanced sensitivity
US9164037B2 (en) 2007-01-26 2015-10-20 Palo Alto Research Center Incorporated Method and system for evaluation of signals received from spatially modulated excitation and emission to accurately determine particle positions and distances
US9638637B2 (en) 2007-01-26 2017-05-02 Palo Alto Research Center Incorporated Method and system implementing spatially modulated excitation or emission for particle characterization with enhanced sensitivity
CN101910821B (en) * 2007-12-04 2012-09-05 粒子监测系统有限公司 Non-orthogonal particle detection systems and methods
US9029800B2 (en) 2011-08-09 2015-05-12 Palo Alto Research Center Incorporated Compact analyzer with spatial modulation and multiple intensity modulated excitation sources

Also Published As

Publication number Publication date
AU2003272804A8 (en) 2004-05-04
US20040067167A1 (en) 2004-04-08
WO2004033059A3 (en) 2006-01-26
AU2003272804A1 (en) 2004-05-04

Similar Documents

Publication Publication Date Title
US20040067167A1 (en) Methods and apparatus for optophoretic diagnosis of cells and particles
AU2002241760B2 (en) Methods and apparatus for use of optical forces for identification, characterization and/or sorting of particles
US6833542B2 (en) Method for sorting particles
US6744038B2 (en) Methods of separating particles using an optical gradient
US6784420B2 (en) Method of separating particles using an optical gradient
US20020108859A1 (en) Methods for modifying interaction between dielectric particles and surfaces
US20020160470A1 (en) Methods and apparatus for generating and utilizing linear moving optical gradients
AU2002241760A1 (en) Methods and apparatus for use of optical forces for identification, characterization and/or sorting of particles
Yang et al. Review and perspectives on microfluidic flow cytometers
US20020115163A1 (en) Methods for sorting particles by size and elasticity
JP6420297B2 (en) Analyzing and sorting objects in fluids
US20030124516A1 (en) Method of using optical interrogation to determine a biological property of a cell or population of cells
US20020123112A1 (en) Methods for increasing detection sensitivity in optical dielectric sorting systems
US10710074B2 (en) Method for laser separation and characterization of particles and molecular species
US20020121443A1 (en) Methods for the combined electrical and optical identification, characterization and/or sorting of particles
US20040033539A1 (en) Method of using optical interrogation to determine a biological property of a cell or population of cells
US20020113204A1 (en) Apparatus for collection of sorted particles
US20040009540A1 (en) Detection and evaluation of cancer cells using optophoretic analysis
US20030211461A1 (en) Optophoretic detection of durgs exhibiting inhibitory effect on Bcr-Abl positive tumor cells
US20030194755A1 (en) Early detection of apoptotic events and apoptosis using optophoretic analysis
US20040053209A1 (en) Detection and evaluation of topoisomerase inhibitors using optophoretic analysis
US20040121474A1 (en) Detection and evaluation of chemically-mediated and ligand-mediated t-cell activation using optophoretic analysis
AU2003202909A1 (en) Methods and apparatus for generating and utilizing linear moving optical gradients
US20040121307A1 (en) Early detection of cellular differentiation using optophoresis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP